1
|
Singh M, Chatzinakos C, Barr PB, Gentry AE, Bigdeli TB, Webb BT, Peterson RE. Trans-ancestry Genome-Wide Analyses in UK Biobank Yield Novel Risk Loci for Major Depression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.22.25322721. [PMID: 40061314 PMCID: PMC11888526 DOI: 10.1101/2025.02.22.25322721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Most genome-wide association studies (GWASs) of depression focus on broad, heterogeneous outcomes, limiting the discovery of genomic risk loci specific to major depressive disorder (MDD). Previous UK Biobank (UKB) studies had limited ability to pinpoint MDD-associated loci due to a smaller sample with strictly defined MDD outcomes and further exclusion of many participants based on ancestry or relatedness, significantly underutilizing this resource's potential for elucidating the genetic architecture of MDD. Here, we present novel genomic insights into MDD by fully utilizing existing UKB data through (1) a trans-ancestry GWAS pipeline using two complementary approaches controlling for population structure and relatedness and (2) an increased sample with MDD symptom-level data across two mental health assessments. We identified strict MDD outcomes among 211,535 participants, representing a 38% increase in eligible participants from prior studies with only one assessment. Ancestrally inclusive analyses yielded 61 genomic risk loci across depression phenotypes, compared to 47 in the analyses restricted to participants genetically similar to European ancestry. Fourteen of these loci, including five novel, were associated with strict MDD phenotypes, whereas only one locus has been previously reported in UKB. MDD-associated genomic loci and predicted gene expression levels showed little overlap with broad depression, indicating higher specificity. Notably, polygenic scores based on these results were significantly associated with depression diagnoses across ancestry groups in the All of Us Research Program, highlighting the shared genetic architecture across populations. While the trans-ancestry analyses, which included non-European participants, increased the number of associated loci, the discovery of non-European ancestry-specific loci was limited, underscoring the need for larger, globally representative studies of MDD. Importantly, beyond these results, our GWAS pipeline will facilitate inclusive analyses of other traits and disorders, helping improve statistical power, representation, and generalizability in genomic studies.
Collapse
Affiliation(s)
- Madhurbain Singh
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Chris Chatzinakos
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Peter B Barr
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Amanda Elswick Gentry
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Tim B Bigdeli
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Bradley T Webb
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC, USA
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Roseann E Peterson
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
2
|
Choi KW, Tubbs JD, Lee YH, He Y, Tsuo K, Yohannes MT, Nkambule LL, Madsen E, Ghimire DJ, Hermosilla S, Ge T, Martin AR, Axinn WG, Smoller JW. Genetic architecture and socio-environmental risk factors for major depressive disorder in Nepal. Psychol Med 2024; 54:3126-3134. [PMID: 39282852 PMCID: PMC12050005 DOI: 10.1017/s0033291724001284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is the leading cause of disability globally, with moderate heritability and well-established socio-environmental risk factors. Genetic studies have been mostly restricted to European settings, with polygenic scores (PGS) demonstrating low portability across diverse global populations. METHODS This study examines genetic architecture, polygenic prediction, and socio-environmental correlates of MDD in a family-based sample of 10 032 individuals from Nepal with array genotyping data. We used genome-based restricted maximum likelihood to estimate heritability, applied S-LDXR to estimate the cross-ancestry genetic correlation between Nepalese and European samples, and modeled PGS trained on a GWAS meta-analysis of European and East Asian ancestry samples. RESULTS We estimated the narrow-sense heritability of lifetime MDD in Nepal to be 0.26 (95% CI 0.18-0.34, p = 8.5 × 10-6). Our analysis was underpowered to estimate the cross-ancestry genetic correlation (rg = 0.26, 95% CI -0.29 to 0.81). MDD risk was associated with higher age (beta = 0.071, 95% CI 0.06-0.08), female sex (beta = 0.160, 95% CI 0.15-0.17), and childhood exposure to potentially traumatic events (beta = 0.050, 95% CI 0.03-0.07), while neither the depression PGS (beta = 0.004, 95% CI -0.004 to 0.01) or its interaction with childhood trauma (beta = 0.007, 95% CI -0.01 to 0.03) were strongly associated with MDD. CONCLUSIONS Estimates of lifetime MDD heritability in this Nepalese sample were similar to previous European ancestry samples, but PGS trained on European data did not predict MDD in this sample. This may be due to differences in ancestry-linked causal variants, differences in depression phenotyping between the training and target data, or setting-specific environmental factors that modulate genetic effects. Additional research among under-represented global populations will ensure equitable translation of genomic findings.
Collapse
Affiliation(s)
- Karmel W. Choi
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston Massachusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
| | - Justin D. Tubbs
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston Massachusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
| | - Younga H. Lee
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston Massachusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
| | - Yixuan He
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kristin Tsuo
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston Massachusetts, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mary T. Yohannes
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston Massachusetts, USA
| | - Lethukuthula L. Nkambule
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston Massachusetts, USA
| | - Emily Madsen
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston Massachusetts, USA
| | - Dirgha J. Ghimire
- Population Studies Center, Institute for Social Research, University of Michigan, Ann Arbor Michigan, USA
| | - Sabrina Hermosilla
- Population Studies Center, Institute for Social Research, University of Michigan, Ann Arbor Michigan, USA
- Department of Population and Family Health, Mailman School of Public Health, Columbia University Irving Medical Center, New York New York, USA
| | - Tian Ge
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston Massachusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
| | - Alicia R. Martin
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston Massachusetts, USA
| | - William G. Axinn
- Population Studies Center, Institute for Social Research, University of Michigan, Ann Arbor Michigan, USA
| | - Jordan W. Smoller
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston Massachusetts, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston Massachusetts, USA
- Stanley Center for Psychiatric Research, Broad Institute, Boston Massachusetts, USA
| |
Collapse
|
3
|
Zonca V, Marizzoni M, Saleri S, Zajkowska Z, Manfro PH, Souza L, Viduani A, Sforzini L, Swartz JR, Fisher HL, Kohrt BA, Kieling C, Riva MA, Cattaneo A, Mondelli V. Inflammation and immune system pathways as biological signatures of adolescent depression-the IDEA-RiSCo study. Transl Psychiatry 2024; 14:230. [PMID: 38824135 PMCID: PMC11144232 DOI: 10.1038/s41398-024-02959-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024] Open
Abstract
The biological mechanisms underlying the onset of major depressive disorder (MDD) have predominantly been studied in adult populations from high-income countries, despite the onset of depression typically occurring in adolescence and the majority of the world's adolescents living in low- and middle-income countries (LMIC). Taking advantage of a unique adolescent sample in an LMIC (Brazil), this study aimed to identify biological pathways characterizing the presence and increased risk of depression in adolescence, and sex-specific differences in such biological signatures. We collected blood samples from a risk-stratified cohort of 150 Brazilian adolescents (aged 14-16 years old) comprising 50 adolescents with MDD, 50 adolescents at high risk of developing MDD but without current MDD, and 50 adolescents at low risk of developing MDD and without MDD (25 females and 25 males in each group). We conducted RNA-Seq and pathway analysis on whole blood. Inflammatory-related biological pathways, such as role of hypercytokinemia/hyperchemokinemia in the pathogenesis of influenza (z-score = 3.464, p < 0.001), interferon signaling (z-score = 2.464, p < 0.001), interferon alpha/beta signaling (z-score = 3.873, p < 0.001), and complement signaling (z-score = 2, p = 0.002) were upregulated in adolescents with MDD compared with adolescents without MDD independently from their level of risk. The up-regulation of such inflammation-related pathways was observed in females but not in males. Inflammatory-related pathways involved in the production of cytokines and in interferon and complement signaling were identified as key indicators of adolescent depression, and this effect was present only in females.
Collapse
Affiliation(s)
- Valentina Zonca
- King's College London, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, London, UK.
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| | - Moira Marizzoni
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni, 4, 25125, Brescia, Italy
| | - Samantha Saleri
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni, 4, 25125, Brescia, Italy
| | - Zuzanna Zajkowska
- King's College London, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, London, UK
| | - Pedro H Manfro
- Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Child & Adolescent Psychiatry Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350-400N, Porto Alegre, RS, 90035-903, Brazil
| | - Laila Souza
- Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Child & Adolescent Psychiatry Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350-400N, Porto Alegre, RS, 90035-903, Brazil
| | - Anna Viduani
- Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Child & Adolescent Psychiatry Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350-400N, Porto Alegre, RS, 90035-903, Brazil
| | - Luca Sforzini
- King's College London, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, London, UK
- National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, King's College London, London, UK
| | - Johnna R Swartz
- Department of Human Ecology, University of California, Davis, Davis, CA, 95616, USA
| | - Helen L Fisher
- King's College London, Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, London, UK
- ESRC Centre for Society and Mental Health, King's College London, London, UK
| | - Brandon A Kohrt
- Center for Global Mental Health Equity, Department of Psychiatry and Behavioral Health, School of Medicine and Health Sciences, The George Washington University, 2120 L St NW, Ste 600, Washington, DC, 20037D, USA
| | - Christian Kieling
- Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Child & Adolescent Psychiatry Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350-400N, Porto Alegre, RS, 90035-903, Brazil
| | - Marco Andrea Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni, 4, 25125, Brescia, Italy
| | - Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Via Pilastroni, 4, 25125, Brescia, Italy
| | - Valeria Mondelli
- King's College London, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, London, UK
- National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, King's College London, London, UK
| |
Collapse
|
4
|
Zhang S, Jiang Z, Zeng P. Incorporating genetic similarity of auxiliary samples into eGene identification under the transfer learning framework. J Transl Med 2024; 22:258. [PMID: 38461317 PMCID: PMC10924384 DOI: 10.1186/s12967-024-05053-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/01/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND The term eGene has been applied to define a gene whose expression level is affected by at least one independent expression quantitative trait locus (eQTL). It is both theoretically and empirically important to identify eQTLs and eGenes in genomic studies. However, standard eGene detection methods generally focus on individual cis-variants and cannot efficiently leverage useful knowledge acquired from auxiliary samples into target studies. METHODS We propose a multilocus-based eGene identification method called TLegene by integrating shared genetic similarity information available from auxiliary studies under the statistical framework of transfer learning. We apply TLegene to eGene identification in ten TCGA cancers which have an explicit relevant tissue in the GTEx project, and learn genetic effect of variant in TCGA from GTEx. We also adopt TLegene to the Geuvadis project to evaluate its usefulness in non-cancer studies. RESULTS We observed substantial genetic effect correlation of cis-variants between TCGA and GTEx for a larger number of genes. Furthermore, consistent with the results of our simulations, we found that TLegene was more powerful than existing methods and thus identified 169 distinct candidate eGenes, which was much larger than the approach that did not consider knowledge transfer across target and auxiliary studies. Previous studies and functional enrichment analyses provided empirical evidence supporting the associations of discovered eGenes, and it also showed evidence of allelic heterogeneity of gene expression. Furthermore, TLegene identified more eGenes in Geuvadis and revealed that these eGenes were mainly enriched in cells EBV transformed lymphocytes tissue. CONCLUSION Overall, TLegene represents a flexible and powerful statistical method for eGene identification through transfer learning of genetic similarity shared across auxiliary and target studies.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Zhou Jiang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Center for Medical Statistics and Data Analysis, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Key Laboratory of Environment and Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Xuzhou Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
5
|
Schat E, Tuerlinckx F, De Ketelaere B, Ceulemans E. Real-time detection of mean and variance changes in experience sampling data: A comparison of existing and novel statistical process control approaches. Behav Res Methods 2024; 56:1459-1475. [PMID: 37118646 DOI: 10.3758/s13428-023-02103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2023] [Indexed: 04/30/2023]
Abstract
Retrospective analyses of experience sampling (ESM) data have shown that changes in mean and variance levels may serve as early warning signs of an imminent depression. Detecting such early warning signs prospectively would pave the way for timely intervention and prevention. The exponentially weighted moving average (EWMA) procedure seems a promising method to scan ESM data for the presence of mean changes in real-time. Based on simulation and empirical studies, computing and monitoring day averages using EWMA works particularly well. We therefore expand this idea to the detection of variance changes and propose to use EWMA to prospectively scan for mean changes in day variability statistics (i.e.,s 2 , s , ln( s )). When both mean and variance changes are of interest, the multivariate extension of EWMA (MEWMA) can be applied to both the day averages and a day statistic of variability. We evaluate these novel approaches to detecting variance changes by comparing them to EWMA-type procedures that have been specifically developed to detect a combination of mean and variance changes in the raw data: EWMA-S 2 , EWMA-ln(S 2 ), and EWMA- X ¯ -S 2 . We ran a simulation study to examine the performance of the two approaches in detecting mean, variance, or both types of changes. The results indicate that monitoring day statistics using (M)EWMA works well and outperforms EWMA-S 2 and EWMA-ln(S 2 ); the performance difference with EWMA- X ¯ -S 2 is smaller but notable. Based on the results, we provide recommendations on which statistic of variability to monitor based on the type of change (i.e., variance increase or decrease) one expects.
Collapse
Affiliation(s)
- Evelien Schat
- Quantitative Psychology and Individual Differences, Faculty of Psychology and Educational Sciences, KU Leuven, Tiensestraat 102 Box 3713, 3000, Leuven, Belgium.
| | - Francis Tuerlinckx
- Quantitative Psychology and Individual Differences, Faculty of Psychology and Educational Sciences, KU Leuven, Tiensestraat 102 Box 3713, 3000, Leuven, Belgium
| | - Bart De Ketelaere
- Mechatronics, Biostatistics and Sensors, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Eva Ceulemans
- Quantitative Psychology and Individual Differences, Faculty of Psychology and Educational Sciences, KU Leuven, Tiensestraat 102 Box 3713, 3000, Leuven, Belgium
| |
Collapse
|
6
|
Meng X, Navoly G, Giannakopoulou O, Levey DF, Koller D, Pathak GA, Koen N, Lin K, Adams MJ, Rentería ME, Feng Y, Gaziano JM, Stein DJ, Zar HJ, Campbell ML, van Heel DA, Trivedi B, Finer S, McQuillin A, Bass N, Chundru VK, Martin HC, Huang QQ, Valkovskaya M, Chu CY, Kanjira S, Kuo PH, Chen HC, Tsai SJ, Liu YL, Kendler KS, Peterson RE, Cai N, Fang Y, Sen S, Scott LJ, Burmeister M, Loos RJF, Preuss MH, Actkins KV, Davis LK, Uddin M, Wani AH, Wildman DE, Aiello AE, Ursano RJ, Kessler RC, Kanai M, Okada Y, Sakaue S, Rabinowitz JA, Maher BS, Uhl G, Eaton W, Cruz-Fuentes CS, Martinez-Levy GA, Campos AI, Millwood IY, Chen Z, Li L, Wassertheil-Smoller S, Jiang Y, Tian C, Martin NG, Mitchell BL, Byrne EM, Awasthi S, Coleman JRI, Ripke S, Sofer T, Walters RG, McIntosh AM, Polimanti R, Dunn EC, Stein MB, Gelernter J, Lewis CM, Kuchenbaecker K. Multi-ancestry genome-wide association study of major depression aids locus discovery, fine mapping, gene prioritization and causal inference. Nat Genet 2024; 56:222-233. [PMID: 38177345 PMCID: PMC10864182 DOI: 10.1038/s41588-023-01596-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/26/2023] [Indexed: 01/06/2024]
Abstract
Most genome-wide association studies (GWAS) of major depression (MD) have been conducted in samples of European ancestry. Here we report a multi-ancestry GWAS of MD, adding data from 21 cohorts with 88,316 MD cases and 902,757 controls to previously reported data. This analysis used a range of measures to define MD and included samples of African (36% of effective sample size), East Asian (26%) and South Asian (6%) ancestry and Hispanic/Latin American participants (32%). The multi-ancestry GWAS identified 53 significantly associated novel loci. For loci from GWAS in European ancestry samples, fewer than expected were transferable to other ancestry groups. Fine mapping benefited from additional sample diversity. A transcriptome-wide association study identified 205 significantly associated novel genes. These findings suggest that, for MD, increasing ancestral and global diversity in genetic studies may be particularly important to ensure discovery of core genes and inform about transferability of findings.
Collapse
Affiliation(s)
| | | | | | - Daniel F Levey
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Dora Koller
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona, Spain
| | - Gita A Pathak
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Nastassja Koen
- SAMRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Mark J Adams
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Miguel E Rentería
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - J Michael Gaziano
- Department of Medicine, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Dan J Stein
- SAMRC Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Heather J Zar
- SAMRC Unit on Child and Adolescent Health, Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Megan L Campbell
- Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | | | - Bhavi Trivedi
- Blizard Institute, Queen Mary University of London, London, UK
| | - Sarah Finer
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | | | - Nick Bass
- Division of Psychiatry, UCL, London, UK
| | | | | | | | | | | | - Susan Kanjira
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Po-Hsiu Kuo
- Department of Public Health and Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
- Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsi-Chung Chen
- Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan
- Center of Sleep Disorders, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Jen Tsai
- Institute of Brain Science and Division of Psychiatry, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | | | - Roseann E Peterson
- Department of Psychiatry, VCU, Richmond, VA, USA
- Department of Psychiatry, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Na Cai
- Helmholtz Pioneer Campus, Helmholtz Munich, Neuherberg, Germany
- Computational Health Centre, Helmholtz Munich, Neuherberg, Germany
- Department of Medicine, Technical University of Munich, Munich, Germany
| | - Yu Fang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Srijan Sen
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Laura J Scott
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Margit Burmeister
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael H Preuss
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ky'Era V Actkins
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lea K Davis
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Monica Uddin
- College of Public Health, University of South Florida, Tampa, FL, USA
| | - Agaz H Wani
- College of Public Health, University of South Florida, Tampa, FL, USA
| | - Derek E Wildman
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Allison E Aiello
- Robert N. Butler Columbia Aging Center, Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Robert J Ursano
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ronald C Kessler
- Department of Health Care Policy, Harvard Medical School, Boston, MA, USA
| | - Masahiro Kanai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Saori Sakaue
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jill A Rabinowitz
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Brion S Maher
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - George Uhl
- Neurology and Pharmacology, University of Maryland, Maryland VA Healthcare System, Baltimore, MD, USA
| | - William Eaton
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Carlos S Cruz-Fuentes
- Departamento de Genética, Instituto Nacional de Psiquiatría 'Ramón de la Fuente Muñíz', Mexico City, Mexico
| | - Gabriela A Martinez-Levy
- Departamento de Genética, Instituto Nacional de Psiquiatría 'Ramón de la Fuente Muñíz', Mexico City, Mexico
| | - Adrian I Campos
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Iona Y Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Zhengming Chen
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Peking University, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | | | - Yunxuan Jiang
- Department of Biostatistics, Emory University, Atlanta, GA, USA
- 23andMe, Inc., Mountain View, CA, USA
| | - Chao Tian
- 23andMe, Inc., Mountain View, CA, USA
| | - Nicholas G Martin
- Mental Health and Neuroscience Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Brittany L Mitchell
- Mental Health and Neuroscience Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Enda M Byrne
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
| | - Swapnil Awasthi
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
| | - Jonathan R I Coleman
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Stephan Ripke
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin, Berlin, Germany
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Cambridge, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Robin G Walters
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Andrew M McIntosh
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
- Institute for Genomics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Renato Polimanti
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- VA Connecticut Healthcare Center, West Haven, CT, USA
| | - Erin C Dunn
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit (PNGU), Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Murray B Stein
- Department of Psychiatry, UC San Diego School of Medicine, La Jolla, CA, USA
- Herbert Wertheim School of Public Health and Human Longevity, University of California San Diego, La Jolla, CA, USA
- Psychiatry Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Joel Gelernter
- Department of Psychiatry, VA CT Healthcare Center, West Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Cathryn M Lewis
- Social Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | | |
Collapse
|
7
|
Qiao J, Wu Y, Zhang S, Xu Y, Zhang J, Zeng P, Wang T. Evaluating significance of European-associated index SNPs in the East Asian population for 31 complex phenotypes. BMC Genomics 2023; 24:324. [PMID: 37312035 DOI: 10.1186/s12864-023-09425-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/01/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) have identified many single-nucleotide polymorphisms (SNPs) associated with complex phenotypes in the European (EUR) population; however, the extent to which EUR-associated SNPs can be generalized to other populations such as East Asian (EAS) is not clear. RESULTS By leveraging summary statistics of 31 phenotypes in the EUR and EAS populations, we first evaluated the difference in heritability between the two populations and calculated the trans-ethnic genetic correlation. We observed the heritability estimates of some phenotypes varied substantially across populations and 53.3% of trans-ethnic genetic correlations were significantly smaller than one. Next, we examined whether EUR-associated SNPs of these phenotypes could be identified in EAS using the trans-ethnic false discovery rate method while accounting for winner's curse for SNP effect in EUR and difference of sample sizes in EAS. We found on average 54.5% of EUR-associated SNPs were also significant in EAS. Furthermore, we discovered non-significant SNPs had higher effect heterogeneity, and significant SNPs showed more consistent linkage disequilibrium and allele frequency patterns between the two populations. We also demonstrated non-significant SNPs were more likely to undergo natural selection. CONCLUSIONS Our study revealed the extent to which EUR-associated SNPs could be significant in the EAS population and offered deep insights into the similarity and diversity of genetic architectures underlying phenotypes in distinct ancestral groups.
Collapse
Affiliation(s)
- Jiahao Qiao
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuxuan Wu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Shuo Zhang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yue Xu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jinhui Zhang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Center for Medical Statistics and Data Analysis, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Key Laboratory of Environment and Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Ting Wang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
8
|
Zhang J, Zhang S, Qiao J, Wang T, Zeng P. Similarity and diversity of genetic architecture for complex traits between East Asian and European populations. BMC Genomics 2023; 24:314. [PMID: 37308816 DOI: 10.1186/s12864-023-09434-x] [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: 01/18/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Genome-wide association studies have detected a large number of single-nucleotide polymorphisms (SNPs) associated with complex traits in diverse ancestral groups. However, the trans-ethnic similarity and diversity of genetic architecture is not well understood currently. RESULTS By leveraging summary statistics of 37 traits from East Asian (Nmax=254,373) or European (Nmax=693,529) populations, we first evaluated the trans-ethnic genetic correlation (ρg) and found substantial evidence of shared genetic overlap underlying these traits between the two populations, with [Formula: see text] ranging from 0.53 (se = 0.11) for adult-onset asthma to 0.98 (se = 0.17) for hemoglobin A1c. However, 88.9% of the genetic correlation estimates were significantly less than one, indicating potential heterogeneity in genetic effect across populations. We next identified common associated SNPs using the conjunction conditional false discovery rate method and observed 21.7% of trait-associated SNPs can be identified simultaneously in both populations. Among these shared associated SNPs, 20.8% showed heterogeneous influence on traits between the two ancestral populations. Moreover, we demonstrated that population-common associated SNPs often exhibited more consistent linkage disequilibrium and allele frequency pattern across ancestral groups compared to population-specific or null ones. We also revealed population-specific associated SNPs were much likely to undergo natural selection compared to population-common associated SNPs. CONCLUSIONS Our study provides an in-depth understanding of similarity and diversity regarding genetic architecture for complex traits across diverse populations, and can assist in trans-ethnic association analysis, genetic risk prediction, and causal variant fine mapping.
Collapse
Affiliation(s)
- Jinhui Zhang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Shuo Zhang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jiahao Qiao
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Ting Wang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Center for Medical Statistics and Data Analysis, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Key Laboratory of Environment and Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
9
|
Yang G, Bozek J, Noble S, Han M, Wu X, Xue M, Kang J, Jia T, Fu J, Ge J, Cui Z, Li X, Feng J, Gao JH. Global diversity in individualized cortical network topography. Cereb Cortex 2023:6992941. [PMID: 36657772 DOI: 10.1093/cercor/bhad002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/24/2022] [Accepted: 12/27/2022] [Indexed: 01/21/2023] Open
Abstract
Individualized cortical network topography (ICNT) varies between people and exhibits great variability in the association networks in the human brain. However, these findings were mainly discovered in Western populations. It remains unclear whether and how ICNT is shaped by the non-Western populations. Here, we leveraged a multisession hierarchical Bayesian model to define individualized functional networks in White American and Han Chinese populations with data from both US and Chinese Human Connectome Projects. We found that both the size and spatial topography of individualized functional networks differed between White American and Han Chinese groups, especially in the heteromodal association cortex (including the ventral attention, control, language, dorsal attention, and default mode networks). Employing a support vector machine, we then demonstrated that ethnicity-related ICNT diversity can be used to identify an individual's ethnicity with high accuracy (74%, pperm < 0.0001), with heteromodal networks contributing most to the classification. This finding was further validated through mass-univariate analyses with generalized additive models. Moreover, we reveal that the spatial heterogeneity of ethnic diversity in ICNT correlated with fundamental properties of cortical organization, including evolutionary cortical expansion, brain myelination, and cerebral blood flow. Altogether, this case study highlights a need for more globally diverse and publicly available neuroimaging datasets.
Collapse
Affiliation(s)
- Guoyuan Yang
- Advanced Research Institute of Multidisciplinary Sciences, School of Medical Technology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jelena Bozek
- Faculty of Electrical Engineering and Computing, University of Zagreb, Zagreb 10000, Croatia
| | - Stephanie Noble
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, United States
| | - Meizhen Han
- McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Xinyu Wu
- Advanced Research Institute of Multidisciplinary Sciences, School of Medical Technology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Mufan Xue
- Advanced Research Institute of Multidisciplinary Sciences, School of Medical Technology, School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Jujiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, Shanghai 200433, China
| | - Tianye Jia
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, Shanghai 200433, China.,Institute of Psychiatry, Psychology & Neuroscience, SGDP Centre, King's College London, London SE5 8AF, United Kingdom
| | - Jilian Fu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300203, China
| | - Jianqiao Ge
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Zaixu Cui
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Xuesong Li
- School of Computer Science and Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Fudan University, Shanghai 200433, China.,Department of Computer Science, University of Warwick, Coventry CV4 7AL, United Kingdom.,MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200433, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.,McGovern Institute for Brain Research, Peking University, Beijing 100871, China.,Beijing City Key Laboratory for Medical Physics and Engineering, Institution of Heavy Ion Physics, School of Physics, Peking University, Beijing 100871, China
| |
Collapse
|
10
|
Qiao J, Shao Z, Wu Y, Zeng P, Wang T. Detecting associated genes for complex traits shared across East Asian and European populations under the framework of composite null hypothesis testing. Lab Invest 2022; 20:424. [PMID: 36138484 PMCID: PMC9503281 DOI: 10.1186/s12967-022-03637-8] [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: 05/08/2022] [Accepted: 09/12/2022] [Indexed: 11/21/2022]
Abstract
Background Detecting trans-ethnic common associated genetic loci can offer important insights into shared genetic components underlying complex diseases/traits across diverse continental populations. However, effective statistical methods for such a goal are currently lacking. Methods By leveraging summary statistics available from global-scale genome-wide association studies, we herein proposed a novel genetic overlap detection method called CONTO (COmposite Null hypothesis test for Trans-ethnic genetic Overlap) from the perspective of high-dimensional composite null hypothesis testing. Unlike previous studies which generally analyzed individual genetic variants, CONTO is a gene-centric method which focuses on a set of genetic variants located within a gene simultaneously and assesses their joint significance with the trait of interest. By borrowing the similar principle of joint significance test (JST), CONTO takes the maximum P value of multiple associations as the significance measurement. Results Compared to JST which is often overly conservative, CONTO is improved in two aspects, including the construction of three-component mixture null distribution and the adjustment of trans-ethnic genetic correlation. Consequently, CONTO corrects the conservativeness of JST with well-calibrated P values and is much more powerful validated by extensive simulation studies. We applied CONTO to discover common associated genes for 31 complex diseases/traits between the East Asian and European populations, and identified many shared trait-associated genes that had otherwise been missed by JST. We further revealed that population-common genes were generally more evolutionarily conserved than population-specific or null ones. Conclusion Overall, CONTO represents a powerful method for detecting common associated genes across diverse ancestral groups; our results provide important implications on the transferability of GWAS discoveries in one population to others. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03637-8.
Collapse
Affiliation(s)
- Jiahao Qiao
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Zhonghe Shao
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuxuan Wu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China. .,Center for Medical Statistics and Data Analysis, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China. .,Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China. .,Key Laboratory of Environment and Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China. .,Engineering Research Innovation Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Ting Wang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
11
|
Sun J, Wang W, Zhang R, Duan H, Tian X, Xu C, Li X, Zhang D. Multivariate genome-wide association study of depression, cognition, and memory phenotypes and validation analysis identify 12 cross-ethnic variants. Transl Psychiatry 2022; 12:304. [PMID: 35907915 PMCID: PMC9338946 DOI: 10.1038/s41398-022-02074-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 11/10/2022] Open
Abstract
To date, little is known about the pleiotropic genetic variants among depression, cognition, and memory. The current research aimed to identify the potential pleiotropic single nucleotide polymorphisms (SNPs), genes, and pathways of the three phenotypes by conducting a multivariate genome-wide association study and an additional pleiotropy analysis among Chinese individuals and further validate the top variants in the UK Biobank (UKB). In the discovery phase, the participants were 139 pairs of dizygotic twins from the Qingdao Twins Registry. The genome-wide efficient mixed-model analysis identified 164 SNPs reaching suggestive significance (P < 1 × 10-5). Among them, rs3967317 (P = 1.21 × 10-8) exceeded the genome-wide significance level (P < 5 × 10-8) and was also demonstrated to be associated with depression and memory in pleiotropy analysis, followed by rs9863698, rs3967316, and rs9261381 (P = 7.80 × 10-8-5.68 × 10-7), which were associated with all three phenotypes. After imputation, a total of 457 SNPs reached suggestive significance. The top SNP chr6:24597173 was located in the KIAA0319 gene, which had biased expression in brain tissues. Genes and pathways related to metabolism, immunity, and neuronal systems demonstrated nominal significance (P < 0.05) in gene-based and pathway enrichment analyses. In the validation phase, 12 of the abovementioned SNPs reached the nominal significance level (P < 0.05) in the UKB. Among them, three SNPs were located in the KIAA0319 gene, and four SNPs were identified as significant expression quantitative trait loci in brain tissues. These findings may provide evidence for pleiotropic variants among depression, cognition, and memory and clues for further exploring the shared genetic pathogenesis of depression with Alzheimer's disease.
Collapse
Affiliation(s)
- Jing Sun
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, Qingdao, Shandong Province, China
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, Qingdao, Shandong Province, China
| | - Ronghui Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, Qingdao, Shandong Province, China
| | - Haiping Duan
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Shibei District, Qingdao, Shandong Province, China
| | - Xiaocao Tian
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Shibei District, Qingdao, Shandong Province, China
| | - Chunsheng Xu
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Shibei District, Qingdao, Shandong Province, China
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, Qingdao, Shandong Province, China.
| |
Collapse
|
12
|
Shen X, Caramaschi D, Adams MJ, Walker RM, Min JL, Kwong A, Hemani G, Barbu MC, Whalley HC, Harris SE, Deary IJ, Morris SW, Cox SR, Relton CL, Marioni RE, Evans KL, McIntosh AM. DNA methylome-wide association study of genetic risk for depression implicates antigen processing and immune responses. Genome Med 2022; 14:36. [PMID: 35354486 PMCID: PMC8969265 DOI: 10.1186/s13073-022-01039-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 03/14/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Depression is a disabling and highly prevalent condition where genetic and epigenetic, such as DNA methylation (DNAm), differences contribute to disease risk. DNA methylation is influenced by genetic variation but the association between polygenic risk of depression and DNA methylation is unknown. METHODS We investigated the association between polygenic risk scores (PRS) for depression and DNAm by conducting a methylome-wide association study (MWAS) in Generation Scotland (N = 8898, mean age = 49.8 years) with replication in the Lothian Birth Cohorts of 1921 and 1936 and adults in the Avon Longitudinal Study of Parents and Children (ALSPAC) (Ncombined = 2049, mean age = 79.1, 69.6 and 47.2 years, respectively). We also conducted a replication MWAS in the ALSPAC children (N = 423, mean age = 17.1 years). Gene ontology analysis was conducted for the cytosine-guanine dinucleotide (CpG) probes significantly associated with depression PRS, followed by Mendelian randomisation (MR) analysis to infer the causal relationship between depression and DNAm. RESULTS Widespread associations (NCpG = 71, pBonferroni < 0.05, p < 6.3 × 10-8) were found between PRS constructed using genetic risk variants for depression and DNAm in CpG probes that localised to genes involved in immune responses and neural development. The effect sizes for the significant associations were highly correlated between the discovery and replication samples in adults (r = 0.79) and in adolescents (r = 0.82). Gene Ontology analysis showed that significant CpG probes are enriched in immunological processes in the human leukocyte antigen system. Additional MWAS was conducted for each lead genetic risk variant. Over 47.9% of the independent genetic risk variants included in the PRS showed associations with DNAm in CpG probes located in both the same (cis) and distal (trans) locations to the genetic loci (pBonferroni < 0.045). Subsequent MR analysis showed that there are a greater number of causal effects found from DNAm to depression than vice versa (DNAm to depression: pFDR ranged from 0.024 to 7.45 × 10-30; depression to DNAm: pFDR ranged from 0.028 to 0.003). CONCLUSIONS PRS for depression, especially those constructed from genome-wide significant genetic risk variants, showed methylome-wide differences associated with immune responses. Findings from MR analysis provided evidence for causal effect of DNAm to depression.
Collapse
Affiliation(s)
- Xueyi Shen
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park, Edinburgh, EH10 5HF, UK.
| | - Doretta Caramaschi
- College of Life and Environmental Sciences, Psychology, University of Exeter, Exeter, UK
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Mark J Adams
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park, Edinburgh, EH10 5HF, UK
| | - Rosie M Walker
- Centre for Clinical Brain Sciences, Chancellor's Building, 49 Little France Crescent, Edinburgh BioQuarter, Edinburgh, UK
| | - Josine L Min
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Alex Kwong
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park, Edinburgh, EH10 5HF, UK
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Gibran Hemani
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Miruna C Barbu
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park, Edinburgh, EH10 5HF, UK
| | - Heather C Whalley
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park, Edinburgh, EH10 5HF, UK
| | - Sarah E Harris
- Lothian Birth Cohorts group, Department of Psychology, The University of Edinburgh, Edinburgh, UK
| | - Ian J Deary
- Lothian Birth Cohorts group, Department of Psychology, The University of Edinburgh, Edinburgh, UK
| | - Stewart W Morris
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Simon R Cox
- Lothian Birth Cohorts group, Department of Psychology, The University of Edinburgh, Edinburgh, UK
| | - Caroline L Relton
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Andrew M McIntosh
- Division of Psychiatry, University of Edinburgh, Royal Edinburgh Hospital, Morningside Park, Edinburgh, EH10 5HF, UK.
| |
Collapse
|
13
|
Chen X, Lu B, Li HX, Li XY, Wang YW, Castellanos FX, Cao LP, Chen NX, Chen W, Cheng YQ, Cui SX, Deng ZY, Fang YR, Gong QY, Guo WB, Hu ZJY, Kuang L, Li BJ, Li L, Li T, Lian T, Liao YF, Liu YS, Liu ZN, Lu JP, Luo QH, Meng HQ, Peng DH, Qiu J, Shen YD, Si TM, Tang YQ, Wang CY, Wang F, Wang HN, Wang K, Wang X, Wang Y, Wang ZH, Wu XP, Xie CM, Xie GR, Xie P, Xu XF, Yang H, Yang J, Yao SQ, Yu YQ, Yuan YG, Zhang KR, Zhang W, Zhang ZJ, Zhu JJ, Zuo XN, Zhao JP, Zang YF, Yan CG, Chen X, Cao LP, Chen W, Cheng YQ, Fang YR, Gong QY, Guo WB, Kuang L, Li BJ, Li T, Liu YS, Liu ZN, Lu JP, Luo QH, Meng HQ, Peng DH, Qiu J, Shen YD, Si TM, Tang YQ, Wang CY, Wang F, Wang HN, Wang K, Wang X, Wang Y, Wu XP, Xie CM, Xie GR, Xie P, Xu XF, Yang H, Yang J, Yao SQ, Yu YQ, Yuan YG, Zhang KR, Zhang W, Zhang ZJ, Zhu JJ, Zuo XN, Zhao JP, Zang YF, et alChen X, Lu B, Li HX, Li XY, Wang YW, Castellanos FX, Cao LP, Chen NX, Chen W, Cheng YQ, Cui SX, Deng ZY, Fang YR, Gong QY, Guo WB, Hu ZJY, Kuang L, Li BJ, Li L, Li T, Lian T, Liao YF, Liu YS, Liu ZN, Lu JP, Luo QH, Meng HQ, Peng DH, Qiu J, Shen YD, Si TM, Tang YQ, Wang CY, Wang F, Wang HN, Wang K, Wang X, Wang Y, Wang ZH, Wu XP, Xie CM, Xie GR, Xie P, Xu XF, Yang H, Yang J, Yao SQ, Yu YQ, Yuan YG, Zhang KR, Zhang W, Zhang ZJ, Zhu JJ, Zuo XN, Zhao JP, Zang YF, Yan CG, Chen X, Cao LP, Chen W, Cheng YQ, Fang YR, Gong QY, Guo WB, Kuang L, Li BJ, Li T, Liu YS, Liu ZN, Lu JP, Luo QH, Meng HQ, Peng DH, Qiu J, Shen YD, Si TM, Tang YQ, Wang CY, Wang F, Wang HN, Wang K, Wang X, Wang Y, Wu XP, Xie CM, Xie GR, Xie P, Xu XF, Yang H, Yang J, Yao SQ, Yu YQ, Yuan YG, Zhang KR, Zhang W, Zhang ZJ, Zhu JJ, Zuo XN, Zhao JP, Zang YF, Yan CG. The DIRECT consortium and the REST-meta-MDD project: towards neuroimaging biomarkers of major depressive disorder. PSYCHORADIOLOGY 2022; 2:32-42. [PMID: 38665141 PMCID: PMC10917197 DOI: 10.1093/psyrad/kkac005] [Show More Authors] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/06/2022] [Accepted: 05/06/2022] [Indexed: 02/05/2023]
Abstract
Despite a growing neuroimaging literature on the pathophysiology of major depressive disorder (MDD), reproducible findings are lacking, probably reflecting mostly small sample sizes and heterogeneity in analytic approaches. To address these issues, the Depression Imaging REsearch ConsorTium (DIRECT) was launched. The REST-meta-MDD project, pooling 2428 functional brain images processed with a standardized pipeline across all participating sites, has been the first effort from DIRECT. In this review, we present an overview of the motivations, rationale, and principal findings of the studies so far from the REST-meta-MDD project. Findings from the first round of analyses of the pooled repository have included alterations in functional connectivity within the default mode network, in whole-brain topological properties, in dynamic features, and in functional lateralization. These well-powered exploratory observations have also provided the basis for future longitudinal hypothesis-driven research. Following these fruitful explorations, DIRECT has proceeded to its second stage of data sharing that seeks to examine ethnicity in brain alterations in MDD by extending the exclusive Chinese original sample to other ethnic groups through international collaborations. A state-of-the-art, surface-based preprocessing pipeline has also been introduced to improve sensitivity. Functional images from patients with bipolar disorder and schizophrenia will be included to identify shared and unique abnormalities across diagnosis boundaries. In addition, large-scale longitudinal studies targeting brain network alterations following antidepressant treatment, aggregation of diffusion tensor images, and the development of functional magnetic resonance imaging-guided neuromodulation approaches are underway. Through these endeavours, we hope to accelerate the translation of functional neuroimaging findings to clinical use, such as evaluating longitudinal effects of antidepressant medications and developing individualized neuromodulation targets, while building an open repository for the scientific community.
Collapse
Affiliation(s)
- Xiao Chen
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences , Beijing 100049, China
| | - Bin Lu
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
| | - Hui-Xian Li
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
| | - Xue-Ying Li
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences , Beijing 100049, China
- Sino-Danish College, University of Chinese Academy of Sciences , Beijing 101408, China
- Sino-Danish Center for Education and Research, Graduate University of Chinese Academy of Sciences , Beijing 101408, China
| | - Yu-Wei Wang
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
| | - Francisco Xavier Castellanos
- Department of Child and Adolescent Psychiatry, NYU Grossman School of Medicine , New York, NY 10016, USA
- Nathan Kline Institute for Psychiatric Research , Orangeburg, New York, NY 10962, USA
| | - Li-Ping Cao
- Affiliated Brain Hospital of Guangzhou Medical University , Guangzhou 510370, China
| | | | - Wei Chen
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine , Hangzhou 310020, Zhejiang, China
| | - Yu-Qi Cheng
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University , Kunming, Yunnan 650032, China
| | - Shi-Xian Cui
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Sino-Danish College, University of Chinese Academy of Sciences , Beijing 101408, China
- Sino-Danish Center for Education and Research, Graduate University of Chinese Academy of Sciences , Beijing 101408, China
| | - Zhao-Yu Deng
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
| | - Yi-Ru Fang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine , Shanghai 200030, China
| | - Qi-Yong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University , Chengdu, Sichuan 610044, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences , Chengdu, Sichuan 610052, China
| | - Wen-Bin Guo
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University , Changsha 410011, Hunan, China
| | - Zheng-Jia-Yi Hu
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
| | - Li Kuang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University , Chongqing 400042, China
| | - Bao-Juan Li
- Xijing Hospital of Air Force Military Medical University , Xi'an, Shaanxi 710032, China
| | - Le Li
- Center for Cognitive Science of Language, Beijing Language and Culture University , Beijing 100083, China
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine , Hangzhou, Zhejiang 310063, China
- Mental Health Center and Psychiatric Laboratory, West China Hospital of Sichuan University , Chengdu, Sichuan 610044, China
| | - Tao Lian
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
| | - Yi-Fan Liao
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
| | - Yan-Song Liu
- Department of Clinical Psychology, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University , Suzhou, Jiangsu 215003, China
| | - Zhe-Ning Liu
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University , Changsha 410011, Hunan, China
| | - Jian-Ping Lu
- Shenzhen Kangning Hospital , Shenzhen, Guangzhou 518020, China
| | - Qing-Hua Luo
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University , Chongqing 400042, China
| | - Hua-Qing Meng
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University , Chongqing 400042, China
| | - Dai-Hui Peng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine , Shanghai 200030, China
| | - Jiang Qiu
- Faculty of Psychology, Southwest University , Chongqing 400715, China
| | - Yue-Di Shen
- Department of Diagnostics, Affiliated Hospital, Hangzhou Normal University Medical School , Hangzhou, Zhejiang 311121, China
| | - Tian-Mei Si
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital) & Key Laboratory of Mental Health, Ministry of Health (Peking University) , Beijing 100191, China
| | - Yan-Qing Tang
- Department of Psychiatry, First Affiliated Hospital, China Medical University , Shenyang, Liaoning 110122, China
| | - Chuan-Yue Wang
- Beijing Anding Hospital, Capital Medical University , Beijing 100120, China
| | - Fei Wang
- Department of Psychiatry, First Affiliated Hospital, China Medical University , Shenyang, Liaoning 110122, China
- Early Intervention Unit, Department of Psychiatry , Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing 210024, China
| | - Hua-Ning Wang
- Xijing Hospital of Air Force Military Medical University , Xi'an, Shaanxi 710032, China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University , Hefei, Anhui 230022, China
| | - Xiang Wang
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University , Changsha 410011, Hunan, China
| | - Ying Wang
- The First Affiliated Hospital of Jinan University , Guangzhou, Guangdong 250024, China
| | - Zi-Han Wang
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
| | - Xiao-Ping Wu
- Xi'an Central Hospital , Xi'an, Shaanxi 710004, China
| | - Chun-Ming Xie
- Department of Neurology, Affiliated ZhongDa Hospital of Southeast University , Nanjing, Jiangsu 210009, China
| | - Guang-Rong Xie
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University , Changsha 410011, Hunan, China
| | - Peng Xie
- Institute of Neuroscience, Chongqing Medical University , Chongqing 400016, China
- Chongqing Key Laboratory of Neurobiology , Chongqing 400000, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University , Chongqing 400042, China
| | - Xiu-Feng Xu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University , Kunming, Yunnan 650032, China
| | - Hong Yang
- Department of Radiology, The First Affiliated Hospital, College of Medicine, Zhejiang University , Hangzhou, Zhejiang 310058, China
| | - Jian Yang
- Chongqing Key Laboratory of Neurobiology , Chongqing 400000, China
| | - Shu-Qiao Yao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University , Changsha 410011, Hunan, China
| | - Yong-Qiang Yu
- The First Affiliated Hospital of Anhui Medical University , Hefei, Anhui 230032, China
| | - Yong-Gui Yuan
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Southeast University , Nanjing, Jiangsu 210009, China
| | - Ke-Rang Zhang
- First Hospital of Shanxi Medical University , Taiyuan, Shanxi 030001, China
| | - Wei Zhang
- West China Hospital of Sichuan University , Chengdu, Sichuan 610044, China
| | - Zhi-Jun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital of Southeast University , Nanjing, Jiangsu 210009, China
| | - Jun-Juan Zhu
- Department of Psychiatry, Shanghai Jiao Tong University School of Medicine , Shanghai 200025, China
| | - Xi-Nian Zuo
- Developmental Population Neuroscience Research Center, IDG/McGovern Institute for Brain Research, Beijing Normal University , Beijing 100091, China
- National Basic Science Data Center , Beijing 100038, China
| | - Jing-Ping Zhao
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University , Changsha 410011, Hunan, China
| | - Yu-Feng Zang
- Center for Cognition and Brain Disorders, The Affiliated Hospital of Hangzhou Normal University , Hangzhou, Zhejiang 310018, China
- Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments , Hangzhou, Zhejiang 310000, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Chao-Gan Yan
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
- International Big-Data Center for Depression Research, Chinese Academy of Sciences , Beijing 100101, China
- Magnetic Resonance Imaging Research Center, Institute of Psychology, Chinese Academy of Sciences , Beijing 100101, China
- Department of Psychology, University of Chinese Academy of Sciences , Beijing 100049, China
- Sino-Danish College, University of Chinese Academy of Sciences , Beijing 101408, China
- Sino-Danish Center for Education and Research, Graduate University of Chinese Academy of Sciences , Beijing 101408, China
| | | |
Collapse
|
14
|
Kwong ASF, Morris TT, Pearson RM, Timpson NJ, Rice F, Stergiakouli E, Tilling K. Polygenic risk for depression, anxiety and neuroticism are associated with the severity and rate of change in depressive symptoms across adolescence. J Child Psychol Psychiatry 2021; 62:1462-1474. [PMID: 33778956 DOI: 10.1111/jcpp.13422] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 01/21/2021] [Accepted: 02/24/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Adolescence marks a period where depression will commonly onset. Twin studies show that genetic influences play a role in how depression develops and changes across adolescence. Recent genome-wide association studies highlight that common genetic variants - which can be combined into polygenic risk scores (PRS) - are also implicated in depression. However, the role of PRS in adolescent depression and changes in adolescent depression is not yet understood. We aimed to examine associations between PRS for five psychiatric traits and depressive symptoms measured across adolescence using cross-sectional and growth-curve models. The five PRS were as follows: depression (DEP), major depressive disorder (MDD), anxiety (ANX), neuroticism (NEU) and schizophrenia (SCZ). METHODS We used data from over 6,000 participants of the Avon Longitudinal Study of Parents and Children (ALSPAC) to examine associations between the five PRS and self-reported depressive symptoms (Short Mood and Feelings Questionnaire) over 9 occasions from 10 to 24 years. The PRS were created from well-powered genome-wide association studies conducted in adult populations. We examined cross-sectional associations between the PRS at each age and then again with longitudinal trajectories of depressive symptoms in a repeated measures framework using multilevel growth-curve analysis to examine the severity and the rate of change. RESULTS There was strong evidence that higher PRS for DEP, MDD and NEU were associated with worse depressive symptoms throughout adolescence and into young adulthood in our cross-sectional analysis, with consistent associations observed across all nine occasions. Growth-curve analyses provided stronger associations (as measured by effect sizes) and additional insights, demonstrating that individuals with higher PRS for DEP, MDD and NEU had steeper trajectories of depressive symptoms across development, all with a greater increasing rate of change during adolescence. Evidence was less consistent for the ANX and SCZ PRS in the cross-sectional analysis, yet there was some evidence for an increasing rate of change in adolescence in the growth-curve analyses with the ANX PRS. CONCLUSIONS These results show that common genetic variants as indexed by varying psychiatric PRS show patterns of specificity that influence both the severity and rate of change in depressive symptoms throughout adolescence and then into young adulthood. Longitudinal data that make use of repeated measures designs have the potential to provide greater insights how genetic factors influence the onset and persistence of adolescent depression.
Collapse
Affiliation(s)
- Alex S F Kwong
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Division of Psychiatry, University of Edinburgh, Edinburgh, UK
| | - Tim T Morris
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Rebecca M Pearson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Frances Rice
- Division of Psychological Medicine & Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics & Genomics, Cardiff University, Cardiff, UK
| | - Evie Stergiakouli
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kate Tilling
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
15
|
Kendall KM, Van Assche E, Andlauer TFM, Choi KW, Luykx JJ, Schulte EC, Lu Y. The genetic basis of major depression. Psychol Med 2021; 51:2217-2230. [PMID: 33682643 DOI: 10.1017/s0033291721000441] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Major depressive disorder (MDD) is a common, debilitating, phenotypically heterogeneous disorder with heritability ranges from 30% to 50%. Compared to other psychiatric disorders, its high prevalence, moderate heritability, and strong polygenicity have posed major challenges for gene-mapping in MDD. Studies of common genetic variation in MDD, driven by large international collaborations such as the Psychiatric Genomics Consortium, have confirmed the highly polygenic nature of the disorder and implicated over 100 genetic risk loci to date. Rare copy number variants associated with MDD risk were also recently identified. The goal of this review is to present a broad picture of our current understanding of the epidemiology, genetic epidemiology, molecular genetics, and gene-environment interplay in MDD. Insights into the impact of genetic factors on the aetiology of this complex disorder hold great promise for improving clinical care.
Collapse
Affiliation(s)
- K M Kendall
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - E Van Assche
- Department of Psychiatry, University of Muenster, Muenster, Germany
| | - T F M Andlauer
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - K W Choi
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA02114, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA02114, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA02115, USA
| | - J J Luykx
- Department of Psychiatry, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Outpatient Second Opinion Clinic, GGNet Mental Health, Warnsveld, The Netherlands
| | - E C Schulte
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Y Lu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
16
|
Zhu X, Ward J, Cullen B, Lyall DM, Strawbridge RJ, Lyall LM, Smith DJ. Phenotypic and genetic associations between anhedonia and brain structure in UK Biobank. Transl Psychiatry 2021; 11:395. [PMID: 34282121 PMCID: PMC8289859 DOI: 10.1038/s41398-021-01522-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
Anhedonia is a core symptom of multiple psychiatric disorders and has been associated with alterations in brain structure. Genome-wide association studies suggest that anhedonia is heritable, with a polygenic architecture, but few studies have explored the association between genetic loading for anhedonia-indexed by polygenic risk scores for anhedonia (PRS-anhedonia)-and structural brain imaging phenotypes. Here, we investigated how anhedonia and PRS-anhedonia were associated with brain structure within the UK Biobank cohort. Brain measures (including total grey/white matter volumes, subcortical volumes, cortical thickness (CT) and white matter integrity) were analysed using linear mixed models in relation to anhedonia and PRS-anhedonia in 19,592 participants (9225 males; mean age = 62.6 years, SD = 7.44). We found that state anhedonia was significantly associated with reduced total grey matter volume (GMV); increased total white matter volume (WMV); smaller volumes in thalamus and nucleus accumbens; reduced CT within the paracentral cortex, the opercular part of inferior frontal gyrus, precentral cortex, insula and rostral anterior cingulate cortex; and poorer integrity of many white matter tracts. PRS-anhedonia was associated with reduced total GMV; increased total WMV; reduced white matter integrity; and reduced CT within the parahippocampal cortex, superior temporal gyrus and insula. Overall, both state anhedonia and PRS-anhedonia were associated with individual differences in multiple brain structures, including within reward-related circuits. These associations may represent vulnerability markers for psychopathology relevant to a range of psychiatric disorders.
Collapse
Affiliation(s)
- Xingxing Zhu
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK.
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Breda Cullen
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Rona J Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Health Data Research (HDR), Glasgow, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
- Division of Psychiatry, Kennedy Tower, Royal Edinburgh Hospital, Edinburgh, UK
| |
Collapse
|
17
|
Lu H, Wang T, Zhang J, Zhang S, Huang S, Zeng P. Evaluating marginal genetic correlation of associated loci for complex diseases and traits between European and East Asian populations. Hum Genet 2021; 140:1285-1297. [PMID: 34091770 DOI: 10.1007/s00439-021-02299-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/31/2021] [Indexed: 12/14/2022]
Abstract
Genome-wide association studies (GWASs) have successfully identified a large amount of single-nucleotide polymorphisms associated with many complex phenotypes in diverse populations. However, a comprehensive understanding of the genetic correlation of associated loci of phenotypes across populations remains lacking and the extent to which associations discovered in one population can be generalized to other populations or can be utilized for trans-ethnic genetic prediction is also unclear. By leveraging summary statistics, we proposed MAGIC to evaluate the trans-ethnic marginal genetic correlation (rm) of per-allele effect sizes for associated SNPs (P < 5E-8) under the framework of measurement error models. We confirmed the methodological advantage of MAGIC over general approaches through simulations and demonstrated its utility by analyzing 34 GWAS summary statistics of phenotypes from the East Asian (Nmax = 254,373) and European (Nmax = 1,220,901) populations. Among these phenotypes, rm was estimated to range from 0.584 (se = 0.140) for breast cancer to 0.949 (se = 0.035) for age of menarche, with an average of 0.835 (se = 0.045). We also uncovered that the trans-ethnic genetic prediction accuracy for phenotypes in the target population would substantially become low when using associated SNPs identified in non-target populations, indicating that associations discovered in the one population cannot be simply generalized to another population and that the accuracy of trans-ethnic phenotype prediction is generally dissatisfactory. Overall, our study provides in-depth insight into trans-ethnic genetic correlation and prediction for complex phenotypes across diverse populations.
Collapse
Affiliation(s)
- Haojie Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ting Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Jinhui Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Shuo Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Shuiping Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.,Center for Medical Statistics and Data Analysis, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ping Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China. .,Center for Medical Statistics and Data Analysis, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
18
|
Amare AT, Schubert KO, Hou L, Clark SR, Papiol S, Cearns M, Heilbronner U, Degenhardt F, Tekola-Ayele F, Hsu YH, Shekhtman T, Adli M, Akula N, Akiyama K, Ardau R, Arias B, Aubry JM, Backlund L, Bhattacharjee AK, Bellivier F, Benabarre A, Bengesser S, Biernacka JM, Birner A, Brichant-Petitjean C, Cervantes P, Chen HC, Chillotti C, Cichon S, Cruceanu C, Czerski PM, Dalkner N, Dayer A, Del Zompo M, DePaulo JR, Étain B, Jamain S, Falkai P, Forstner AJ, Frisen L, Frye MA, Fullerton JM, Gard S, Garnham JS, Goes FS, Grigoroiu-Serbanescu M, Grof P, Hashimoto R, Hauser J, Herms S, Hoffmann P, Hofmann A, Jiménez E, Kahn JP, Kassem L, Kuo PH, Kato T, Kelsoe JR, Kittel-Schneider S, Kliwicki S, König B, Kusumi I, Laje G, Landén M, Lavebratt C, Leboyer M, Leckband SG, Tortorella A, Manchia M, Martinsson L, McCarthy MJ, McElroy SL, Colom F, Mitjans M, Mondimore FM, Monteleone P, Nievergelt CM, Nöthen MM, Novák T, O'Donovan C, Ozaki N, Ösby U, Pfennig A, Potash JB, Reif A, Reininghaus E, Rouleau GA, Rybakowski JK, Schalling M, Schofield PR, Schweizer BW, Severino G, Shilling PD, Shimoda K, Simhandl C, Slaney CM, Squassina A, Stamm T, Stopkova P, Maj M, et alAmare AT, Schubert KO, Hou L, Clark SR, Papiol S, Cearns M, Heilbronner U, Degenhardt F, Tekola-Ayele F, Hsu YH, Shekhtman T, Adli M, Akula N, Akiyama K, Ardau R, Arias B, Aubry JM, Backlund L, Bhattacharjee AK, Bellivier F, Benabarre A, Bengesser S, Biernacka JM, Birner A, Brichant-Petitjean C, Cervantes P, Chen HC, Chillotti C, Cichon S, Cruceanu C, Czerski PM, Dalkner N, Dayer A, Del Zompo M, DePaulo JR, Étain B, Jamain S, Falkai P, Forstner AJ, Frisen L, Frye MA, Fullerton JM, Gard S, Garnham JS, Goes FS, Grigoroiu-Serbanescu M, Grof P, Hashimoto R, Hauser J, Herms S, Hoffmann P, Hofmann A, Jiménez E, Kahn JP, Kassem L, Kuo PH, Kato T, Kelsoe JR, Kittel-Schneider S, Kliwicki S, König B, Kusumi I, Laje G, Landén M, Lavebratt C, Leboyer M, Leckband SG, Tortorella A, Manchia M, Martinsson L, McCarthy MJ, McElroy SL, Colom F, Mitjans M, Mondimore FM, Monteleone P, Nievergelt CM, Nöthen MM, Novák T, O'Donovan C, Ozaki N, Ösby U, Pfennig A, Potash JB, Reif A, Reininghaus E, Rouleau GA, Rybakowski JK, Schalling M, Schofield PR, Schweizer BW, Severino G, Shilling PD, Shimoda K, Simhandl C, Slaney CM, Squassina A, Stamm T, Stopkova P, Maj M, Turecki G, Vieta E, Veeh J, Witt SH, Wright A, Zandi PP, Mitchell PB, Bauer M, Alda M, Rietschel M, McMahon FJ, Schulze TG, Baune BT. Association of polygenic score for major depression with response to lithium in patients with bipolar disorder. Mol Psychiatry 2021; 26:2457-2470. [PMID: 32203155 DOI: 10.1038/s41380-020-0689-5] [Show More Authors] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/28/2020] [Accepted: 02/13/2020] [Indexed: 11/09/2022]
Abstract
Lithium is a first-line medication for bipolar disorder (BD), but only one in three patients respond optimally to the drug. Since evidence shows a strong clinical and genetic overlap between depression and bipolar disorder, we investigated whether a polygenic susceptibility to major depression is associated with response to lithium treatment in patients with BD. Weighted polygenic scores (PGSs) were computed for major depression (MD) at different GWAS p value thresholds using genetic data obtained from 2586 bipolar patients who received lithium treatment and took part in the Consortium on Lithium Genetics (ConLi+Gen) study. Summary statistics from genome-wide association studies in MD (135,458 cases and 344,901 controls) from the Psychiatric Genomics Consortium (PGC) were used for PGS weighting. Response to lithium treatment was defined by continuous scores and categorical outcome (responders versus non-responders) using measurements on the Alda scale. Associations between PGSs of MD and lithium treatment response were assessed using a linear and binary logistic regression modeling for the continuous and categorical outcomes, respectively. The analysis was performed for the entire cohort, and for European and Asian sub-samples. The PGSs for MD were significantly associated with lithium treatment response in multi-ethnic, European or Asian populations, at various p value thresholds. Bipolar patients with a low polygenic load for MD were more likely to respond well to lithium, compared to those patients with high polygenic load [lowest vs highest PGS quartiles, multi-ethnic sample: OR = 1.54 (95% CI: 1.18-2.01) and European sample: OR = 1.75 (95% CI: 1.30-2.36)]. While our analysis in the Asian sample found equivalent effect size in the same direction: OR = 1.71 (95% CI: 0.61-4.90), this was not statistically significant. Using PGS decile comparison, we found a similar trend of association between a high genetic loading for MD and lower response to lithium. Our findings underscore the genetic contribution to lithium response in BD and support the emerging concept of a lithium-responsive biotype in BD.
Collapse
Affiliation(s)
- Azmeraw T Amare
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
- South Australian Academic Health Science and Translation Centre, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Klaus Oliver Schubert
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
- Northern Adelaide Local Health Network, Mental Health Services, Adelaide, SA, Australia
| | - Liping Hou
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Scott R Clark
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Sergi Papiol
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Micah Cearns
- Discipline of Psychiatry, School of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August University Göttingen, Göttingen, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Yi-Hsiang Hsu
- HSL Institute for Aging Research, Harvard Medical School, Boston, MA, USA
- Program for Quantitative Genomics, Harvard School of Public Health, Boston, MA, USA
| | - Tatyana Shekhtman
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Mazda Adli
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Nirmala Akula
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Kazufumi Akiyama
- Department of Biological Psychiatry and Neuroscience, Dokkyo Medical University School of Medicine, Mibu, Tochigi, Japan
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, Hospital University Agency of Cagliari, Cagliari, Italy
| | - Bárbara Arias
- Unitat de Zoologia i Antropologia Biològica (Dpt. Biologia Evolutiva, Ecologia i Ciències Ambientals), Facultat de Biologia and Institut de Biomedicina (IBUB), University of Barcelona, CIBERSAM, Barcelona, Spain
| | - Jean-Michel Aubry
- Department of Psychiatry, Mood Disorders Unit, HUG - Geneva University Hospitals, Geneva, Switzerland
| | - Lena Backlund
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | | | - Frank Bellivier
- INSERM UMR-S 1144, Université Paris Diderot, Département de Psychiatrie et de Médecine Addictologique, AP-HP, Groupe Hospitalier Saint-Louis-Lariboisière-F.Widal, Paris, France
| | - Antonio Benabarre
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Susanne Bengesser
- Department of Psychiatry and Psychotherapeutic Medicine, Research Unit for bipolar affective disorder, Medical University of Graz, Graz, Austria
| | - Joanna M Biernacka
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Armin Birner
- Department of Psychiatry and Psychotherapeutic Medicine, Research Unit for bipolar affective disorder, Medical University of Graz, Graz, Austria
| | - Clara Brichant-Petitjean
- INSERM UMR-S 1144, Université Paris Diderot, Département de Psychiatrie et de Médecine Addictologique, AP-HP, Groupe Hospitalier Saint-Louis-Lariboisière-F.Widal, Paris, France
| | - Pablo Cervantes
- The Neuromodulation Unit, McGill University Health Centre, Montreal, QC, Canada
| | - Hsi-Chung Chen
- Department of Psychiatry & Center of Sleep Disorders, National Taiwan University Hospital, Taipei, Taiwan
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, Hospital University Agency of Cagliari, Cagliari, Italy
| | - Sven Cichon
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
- Human Genomics Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Cristiana Cruceanu
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Piotr M Czerski
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Nina Dalkner
- Department of Psychiatry and Psychotherapeutic Medicine, Research Unit for bipolar affective disorder, Medical University of Graz, Graz, Austria
| | - Alexandre Dayer
- Department of Psychiatry, Mood Disorders Unit, HUG - Geneva University Hospitals, Geneva, Switzerland
| | - Maria Del Zompo
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - J Raymond DePaulo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Bruno Étain
- INSERM UMR-S 1144, Université Paris Diderot, Département de Psychiatrie et de Médecine Addictologique, AP-HP, Groupe Hospitalier Saint-Louis-Lariboisière-F.Widal, Paris, France
| | - Stephane Jamain
- Inserm U955, Translational Psychiatry laboratory, Fondation FondaMental, Créteil, France
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
- Human Genomics Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- Department of Psychiatry (UPK), University of Basel, Basel, Switzerland
| | - Louise Frisen
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mark A Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Janice M Fullerton
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Sébastien Gard
- Service de psychiatrie, Hôpital Charles Perrens, Bordeaux, France
| | - Julie S Garnham
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Fernando S Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Maria Grigoroiu-Serbanescu
- Biometric Psychiatric Genetics Research Unit, Alexandru Obregia Clinical Psychiatric Hospital, Bucharest, Romania
| | - Paul Grof
- Mood Disorders Center of Ottawa, Ottawa, ON, Canada
| | - Ryota Hashimoto
- Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Joanna Hauser
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Stefan Herms
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
- Human Genomics Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
- Human Genomics Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Andrea Hofmann
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
| | - Esther Jiménez
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Jean-Pierre Kahn
- Service de Psychiatrie et Psychologie Clinique, Centre Psychothérapique de Nancy - Université de Lorraine, Nancy, France
| | - Layla Kassem
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Po-Hsiu Kuo
- Department of Public Health & Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Center for Brain Science, Saitama, Japan
| | - John R Kelsoe
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Sebastian Kliwicki
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Barbara König
- Department of Psychiatry and Psychotherapeutic Medicine, Landesklinikum Neunkirchen, Neunkirchen, Austria
| | - Ichiro Kusumi
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Gonzalo Laje
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Mikael Landén
- Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the Gothenburg University, Gothenburg, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Marion Leboyer
- Inserm U955, Translational Psychiatry laboratory, Université Paris-Est-Créteil, Department of Psychiatry and Addictology of Mondor University Hospital, AP-HP, Fondation FondaMental, Créteil, France
| | - Susan G Leckband
- Office of Mental Health, VA San Diego Healthcare System, San Diego, CA, USA
| | | | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Lina Martinsson
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
| | - Michael J McCarthy
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
- Department of Psychiatry, VA San Diego Healthcare System, San Diego, CA, USA
| | - Susan L McElroy
- Department of Psychiatry, Lindner Center of Hope / University of Cincinnati, Mason, OH, USA
| | - Francesc Colom
- Mental Health Research Group, IMIM-Hospital del Mar, Barcelona, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Marina Mitjans
- Unitat de Zoologia i Antropologia Biològica (Dpt. Biologia Evolutiva, Ecologia i Ciències Ambientals), Facultat de Biologia and Institut de Biomedicina (IBUB), University of Barcelona, CIBERSAM, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Francis M Mondimore
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Palmiero Monteleone
- Neurosciences Section, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
- Department of Psychiatry, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn and Department of Genomics, Life & Brain Center, Bonn, Germany
| | - Tomas Novák
- National Institute of Mental Health, Klecany, Czech Republic
| | - Claire O'Donovan
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Norio Ozaki
- Department of Psychiatry & Department of Child and Adolescent Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Urban Ösby
- Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Andrea Pfennig
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - James B Potash
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Eva Reininghaus
- Department of Psychiatry and Psychotherapeutic Medicine, Research Unit for bipolar affective disorder, Medical University of Graz, Graz, Austria
| | - Guy A Rouleau
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Janusz K Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Peter R Schofield
- Neuroscience Research Australia, Sydney, NSW, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Barbara W Schweizer
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Giovanni Severino
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Paul D Shilling
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Katzutaka Shimoda
- Department of Psychiatry, Dokkyo Medical University School of Medicine, Mibu, Tochigi, Japan
| | - Christian Simhandl
- Bipolar Center Wiener Neustadt, Sigmund Freud University, Medical Faculty, Vienna, Austria
| | - Claire M Slaney
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Alessio Squassina
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Thomas Stamm
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany
| | - Pavla Stopkova
- National Institute of Mental Health, Klecany, Czech Republic
| | - Mario Maj
- Department of Psychiatry, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gustavo Turecki
- Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Eduard Vieta
- Bipolar Disorder Program, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, Barcelona, Catalonia, Spain
| | - Julia Veeh
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Adam Wright
- School of Psychiatry, University of New South Wales, and Black Dog Institute, Sydney, NSW, Australia
| | - Peter P Zandi
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Philip B Mitchell
- School of Psychiatry, University of New South Wales, and Black Dog Institute, Sydney, NSW, Australia
| | - Michael Bauer
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
- National Institute of Mental Health, Klecany, Czech Republic
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Francis J McMahon
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
| | - Thomas G Schulze
- Intramural Research Program, National Institute of Mental Health, National Institutes of Health, US Department of Health & Human Services, Bethesda, MD, USA
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center (UMG), Georg-August University Göttingen, Göttingen, Germany
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD, USA
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bernhard T Baune
- Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany.
- Department of Psychiatry, Melbourne Medical School, University of Melbourne, Parkville, VIC, Australia.
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
19
|
Shalimova A, Babasieva V, Chubarev VN, Tarasov VV, Schiöth HB, Mwinyi J. Therapy response prediction in major depressive disorder: current and novel genomic markers influencing pharmacokinetics and pharmacodynamics. Pharmacogenomics 2021; 22:485-503. [PMID: 34018822 DOI: 10.2217/pgs-2020-0157] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Major depressive disorder is connected with high rates of functional disability and mortality. About a third of the patients are at risk of therapy failure. Several pharmacogenetic markers especially located in CYP450 genes such as CYP2D6 or CYP2C19 are of relevance for therapy outcome prediction in major depressive disorder but a further optimization of predictive tools is warranted. The article summarizes the current knowledge on pharmacogenetic variants, therapy effects and side effects of important antidepressive therapeutics, and sheds light on new methodological approaches for therapy response estimation based on genetic markers with relevance for pharmacokinetics, pharmacodynamics and disease pathology identified in genome-wide association study analyses, highlighting polygenic risk score analysis as a tool for further optimization of individualized therapy outcome prediction.
Collapse
Affiliation(s)
- Alena Shalimova
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, 751 24, Sweden.,Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Viktoria Babasieva
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, 751 24, Sweden.,Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Vladimir N Chubarev
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Vadim V Tarasov
- Department of Pharmacology, Institute of Pharmacy, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia.,Institute of Translational Medicine & Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, 751 24, Sweden.,Institute of Translational Medicine & Biotechnology, I. M. Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Jessica Mwinyi
- Department of Neuroscience, Functional Pharmacology, University of Uppsala, Uppsala, 751 24, Sweden
| |
Collapse
|
20
|
Peterson RE. The Genetics of Major Depression: Perspectives on the State of Research and Opportunities for Precision Medicine. Psychiatr Ann 2021; 51:165-169. [PMID: 35291372 DOI: 10.3928/00485713-20210315-01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Major depression (MD) is a leading cause of disability worldwide; it arises from the action and interaction between genetic and environmental factors, and is often comorbid with other psychiatric and medical conditions. To date, upwards of 100 genetic loci have been associated with MD, giving clues to biological underpinnings. Although recent progress has yielded modest insight into the genetic architecture of MD, most studies have been in populations with European ancestry, seriously limiting precision medicine efforts. Broadening diversity of study populations will empower genomic research by expanding discovery and enhancing our understanding of the role of genomic variation in disease etiology. To fully realize the potential of pharmacogenetics and precision medicine, we will need to address the major gaps in our knowledge of the genetic and environmental risk architecture of MD across ancestries, including sex differences, to improve etiologic understanding, diagnosis, prevention, and treatment for all.
Collapse
Affiliation(s)
- Roseann E Peterson
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University
| |
Collapse
|
21
|
Guo J, Bakshi A, Wang Y, Jiang L, Yengo L, Goddard ME, Visscher PM, Yang J. Quantifying genetic heterogeneity between continental populations for human height and body mass index. Sci Rep 2021; 11:5240. [PMID: 33664403 PMCID: PMC7933291 DOI: 10.1038/s41598-021-84739-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/22/2021] [Indexed: 12/26/2022] Open
Abstract
Genome-wide association studies (GWAS) in samples of European ancestry have identified thousands of genetic variants associated with complex traits in humans. However, it remains largely unclear whether these associations can be used in non-European populations. Here, we seek to quantify the proportion of genetic variation for a complex trait shared between continental populations. We estimated the between-population correlation of genetic effects at all SNPs (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$r_{g}$$\end{document}rg) or genome-wide significant SNPs (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$r_{{g\left( {GWS} \right)}}$$\end{document}rgGWS) for height and body mass index (BMI) in samples of European (EUR; \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$n = 49,839$$\end{document}n=49,839) and African (AFR; \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$n = 17,426$$\end{document}n=17,426) ancestry. The \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hat{r}_{g}$$\end{document}r^g between EUR and AFR was 0.75 (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{s}}.{\text{e}}. = 0.035$$\end{document}s.e.=0.035) for height and 0.68 (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{s}}.{\text{e}}. = 0.062$$\end{document}s.e.=0.062) for BMI, and the corresponding \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hat{r}_{{g\left( {GWS} \right)}}$$\end{document}r^gGWS was 0.82 (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{s}}.{\text{e}}. = 0.030$$\end{document}s.e.=0.030) for height and 0.87 (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\text{s}}.{\text{e}}. = 0.064$$\end{document}s.e.=0.064) for BMI, suggesting that a large proportion of GWAS findings discovered in Europeans are likely applicable to non-Europeans for height and BMI. There was no evidence that \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hat{r}_{g}$$\end{document}r^g differs in SNP groups with different levels of between-population difference in allele frequency or linkage disequilibrium, which, however, can be due to the lack of power.
Collapse
Affiliation(s)
- Jing Guo
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.,Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Andrew Bakshi
- Monash Partners Comprehensive Cancer Consortium, Monash Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ying 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
| | - Loic Yengo
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Michael E Goddard
- Faculty of Veterinary and Agricultural Science, University of Melbourne, Parkville, VIC, Australia.,Biosciences Research Division, Department of Economic Development, Jobs, Transport and Resources, Bundoora, VIC, Australia
| | - Peter M Visscher
- 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, 310024, Zhejiang, China. .,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
| |
Collapse
|
22
|
Mullins N, Huang H. Genetic Architecture of Bipolar Disorder in Individuals of Han Chinese and European Ancestries. JAMA Psychiatry 2021; 78:248-249. [PMID: 33263741 DOI: 10.1001/jamapsychiatry.2020.3639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Niamh Mullins
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hailiang Huang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts.,Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly heritable neurodevelopmental disorder that is known to have a polygenic (i.e., many genes of individually small effects) architecture. Polygenic scores (PGS), which characterize this polygenicity as a single score for a given individual, are considered the state-of-the-art in psychiatric genetics research. Despite the proliferation of ADHD studies adopting this approach and its clinical implications, remarkably little is known about the predictive utility of PGS in ADHD research to date, given that there have not yet been any systematic or meta-analytic reviews of this rapidly developing literature. We meta-analyzed 12 unique effect sizes from ADHD PGS studies, yielding an N = 40,088. These studies, which included a mixture of large population-based cohorts and case-control samples of predominantly European ancestry, yielded a pooled ADHD PGS effect size of rrandom = 0.201 (95% CI = [0.144, 0.288]) and an rfixed = 0.190 (95% CI = [0.180, 0.199]) in predicting ADHD. In other words, ADHD PGS reliably account for between 3.6% (in the fixed effects model) to 4.0% (in the random effects model) of the variance in broadly defined phenotypic ADHD. Findings provide important insights into the genetics of psychiatric outcomes and raise several key questions about the impact of PGS on psychiatric research moving forward. Our review concludes by providing recommendations for future research directions in the use of PGS, including new methods to account for comorbidities, integrating bioinformatics to elucidate biological pathways, and leveraging PGS to test mechanistic models of ADHD.
Collapse
Affiliation(s)
- James J Li
- Department of Psychology, University of Wisconsin, Madison, WI, USA.
- Waisman Center, University of Wisconsin, WI, Madison, USA.
- Center for Demography of Health and Aging, University of Wisconsin, WI, Madison, USA.
| | - Quanfa He
- Department of Psychology, University of Wisconsin, Madison, WI, USA
- Waisman Center, University of Wisconsin, WI, Madison, USA
| |
Collapse
|
24
|
Ma M, Zhang Y, Zhang X, Yan H, Zhang D, Yue W. Common and Distinct Alterations of Cognitive Function and Brain Structure in Schizophrenia and Major Depressive Disorder: A Pilot Study. Front Psychiatry 2021; 12:705998. [PMID: 34354618 PMCID: PMC8329352 DOI: 10.3389/fpsyt.2021.705998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/03/2021] [Indexed: 01/10/2023] Open
Abstract
Objective: Numerous studies indicate that schizophrenia (SCZ) and major depressive disorder (MDD) share pathophysiological characteristics. Investigating the neurobiological features of psychiatric-affective disorders may facilitate the diagnosis of psychiatric disorders. Hence, we aimed to explore whether patients with SCZ and patients with MDD had the similar or distinct cognitive impairments and GMV alterations to further understand their underlying pathophysiological mechanisms. Methods: We recruited a total of 52 MDD patients, 64 SCZ patients, and 65 healthy controls (HCs). The Measurement and Treatment Research to Improve Cognition in Schizophrenia (MATRICS) Consensus Cognitive Battery was used to assess cognitive functions. In addition, voxel-based morphometry (VBM) analysis was used to evaluate the gray matter volume (GMV) by using MRI scanning. One-way ANOVA and post-hoc tests were used to find the differences among the MDD, SCZ, and HCs. Finally, we explored the correlation between structural alterations and cognitive functions. Results: Compared with that of HCs, processing speed was impaired in both patients with SCZ and patients with MDD (F = 49.505, p < 0.001). SCZ patients displayed impaired cognitive performance in all dimensions of cognitive functions compared with HCs (p < 0.001, except social cognition, p = 0.043, Bonferroni corrected). Whole-brain VBM analysis showed that both SCZ and MDD groups had reductions of GMV in the medial superior frontal cortex (cluster-level FWE p < 0.05). Patients with SCZ exhibited declining GMV in the anterior cingulate cortex and right middle frontal cortex (MFC) compared with HCs and MDD patients (cluster-level FWE p < 0.05). The mean values of GMV in the right MFC had a positive correlation with the attention/vigilance function in patients with MDD (p = 0.014, partial. r = 0.349, without Bonferroni correction). Conclusions: In total, our study found that MDD and SCZ groups had common cognitive impairments and brain structural alterations, but the SCZ group exhibited more severe impairment than the MDD group in both fields. The above findings may provide a potential support for recognizing the convergent and divergent brain neural pathophysiological mechanisms between MDD and SCZ.
Collapse
Affiliation(s)
- Mengying Ma
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China
| | - Yuyanan Zhang
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China
| | - Xiao Zhang
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China
| | - Hao Yan
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China
| | - Dai Zhang
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Weihua Yue
- Institute of Mental Health, The Sixth Hospital, Peking University, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health & National Clinical Research Center for Mental Disorders, Peking University, Beijing, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| |
Collapse
|
25
|
Dueñas HR, Seah C, Johnson JS, Huckins LM. Implicit bias of encoded variables: frameworks for addressing structured bias in EHR-GWAS data. Hum Mol Genet 2020; 29:R33-R41. [PMID: 32879975 PMCID: PMC7530523 DOI: 10.1093/hmg/ddaa192] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022] Open
Abstract
The 'discovery' stage of genome-wide association studies required amassing large, homogeneous cohorts. In order to attain clinically useful insights, we must now consider the presentation of disease within our clinics and, by extension, within our medical records. Large-scale use of electronic health record (EHR) data can help to understand phenotypes in a scalable manner, incorporating lifelong and whole-phenome context. However, extending analyses to incorporate EHR and biobank-based analyses will require careful consideration of phenotype definition. Judgements and clinical decisions that occur 'outside' the system inevitably contain some degree of bias and become encoded in EHR data. Any algorithmic approach to phenotypic characterization that assumes non-biased variables will generate compounded biased conclusions. Here, we discuss and illustrate potential biases inherent within EHR analyses, how these may be compounded across time and suggest frameworks for large-scale phenotypic analysis to minimize and uncover encoded bias.
Collapse
Affiliation(s)
- Hillary R Dueñas
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carina Seah
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jessica S Johnson
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura M Huckins
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mental Illness Research, Education and Clinical Centers, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| |
Collapse
|
26
|
Cai N, Choi KW, Fried EI. Reviewing the genetics of heterogeneity in depression: operationalizations, manifestations and etiologies. Hum Mol Genet 2020; 29:R10-R18. [PMID: 32568380 PMCID: PMC7530517 DOI: 10.1093/hmg/ddaa115] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
With progress in genome-wide association studies of depression, from identifying zero hits in ~16 000 individuals in 2013 to 223 hits in more than a million individuals in 2020, understanding the genetic architecture of this debilitating condition no longer appears to be an impossible task. The pressing question now is whether recently discovered variants describe the etiology of a single disease entity. There are a myriad of ways to measure and operationalize depression severity, and major depressive disorder as defined in the Diagnostic and Statistical Manual of Mental Disorders-5 can manifest in more than 10 000 ways based on symptom profiles alone. Variations in developmental timing, comorbidity and environmental contexts across individuals and samples further add to the heterogeneity. With big data increasingly enabling genomic discovery in psychiatry, it is more timely than ever to explicitly disentangle genetic contributions to what is likely 'depressions' rather than depression. Here, we introduce three sources of heterogeneity: operationalization, manifestation and etiology. We review recent efforts to identify depression subtypes using clinical and data-driven approaches, examine differences in genetic architecture of depression across contexts, and argue that heterogeneity in operationalizations of depression is likely a considerable source of inconsistency. Finally, we offer recommendations and considerations for the field going forward.
Collapse
Affiliation(s)
- Na Cai
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg 85764, Germany
| | - Karmel W Choi
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute, Boston, MA 02142, USA
| | - Eiko I Fried
- Department of Psychology, Leiden University, Leiden 2333 AK, Netherlands
| |
Collapse
|
27
|
Rao S, Shi M, Han X, Lam MHB, Chien WT, Zhou K, Liu G, Wing YK, So HC, Waye MMY. Genome-wide copy number variation-, validation- and screening study implicates a new copy number polymorphism associated with suicide attempts in major depressive disorder. Gene 2020; 755:144901. [PMID: 32554045 DOI: 10.1016/j.gene.2020.144901] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 05/08/2020] [Accepted: 06/10/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND The genetic basis of suicide attempts (SA) remains unclear. Especially the role of copy number variations (CNVs) remains to be elucidated. The present study aimed to identify susceptibility variants associated with SA among Chinese with major depressive disorder (MDD), covering both CNVs and single-nucleotide polymorphisms (SNPs). METHODS We conducted a genome-wide association study (GWAS) on MDD patients with and without SA and top results were tested in a replication study. A genome-wide CNV study was also performed. Subsequently, a validation assay using qRT-PCR technology was performed to confirm any associated CNVs and then applied to the entire cohort to examine the association. RESULTS Although GWAS did not identify any SNPs reaching genome-wide significance, we identified TPH2 as the top susceptibility gene (p-value = 2.75e-05) in gene-based analysis, which is a strong biological candidate for its role in the serotonergic system. As for CNV analysis, we found that the global rate of CNV was higher in SA than that in non-SA subjects (p-value = 0.023). Genome-wide CNV study revealed an SA-associated CNV region that achieved genome-wide significance (corrected p-value = 0.014). The associated CNV was successfully validated with a more rigorous qRT-PCR assay and identified to be a common variant in this cohort. Its deletion rate was higher in SA subjects [OR = 2.05 (1.02-4.12), adjusted p-value = 0.045]. Based on the GTEx database, genetic variants that probed this CNV were significantly associated with the expression level of ZNF33B in two brain regions (p-value < 4.2e-05). In stratified analysis, the CNV showed a significant effect [OR = 2.58 (1.06-6.27), p-value = 0.039] in those with high neuroticism but not in those with average or low neuroticism. CONCLUSIONS We identified a new common CNV likely involved in the etiology of SA. This finding sheds light on an important role of common CNVs in the pathophysiology of SA, suggesting a new promising avenue for investigating its genetic architecture.
Collapse
Affiliation(s)
- Shitao Rao
- The Nethersole School of Nursing, The Croucher Laboratory for Human Genomics, China; Department of Psychiatry, N.T, Hong Kong Special Administrative Region; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, N.T, Hong Kong Special Administrative Region
| | - Mai Shi
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, N.T, Hong Kong Special Administrative Region
| | - Xinyu Han
- College of Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
| | - Marco Ho Bun Lam
- Department of Psychiatry, N.T, Hong Kong Special Administrative Region
| | - Wai Tong Chien
- The Nethersole School of Nursing, The Croucher Laboratory for Human Genomics, China
| | - Keying Zhou
- Shenzhen People's Hospital, The 2nd Clinical Medical College of Jinan University, The 1st Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Guangming Liu
- College of Food and Biological Engineering, Jimei University, Xiamen, Fujian, China
| | - Yun Kwok Wing
- Department of Psychiatry, N.T, Hong Kong Special Administrative Region
| | - Hon-Cheong So
- Department of Psychiatry, N.T, Hong Kong Special Administrative Region; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, N.T, Hong Kong Special Administrative Region; KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology and The Chinese University of Hong Kong, China; CUHK Shenzhen Research Institute, Shenzhen, China; Brain and Mind Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Mary Miu Yee Waye
- The Nethersole School of Nursing, The Croucher Laboratory for Human Genomics, China.
| |
Collapse
|
28
|
Yuan B, Sun X, Xu Z, Pu M, Yuan Y, Zhang Z. Influence of genetic polymorphisms in homocysteine and lipid metabolism systems on antidepressant drug response. BMC Psychiatry 2020; 20:408. [PMID: 32795354 PMCID: PMC7427977 DOI: 10.1186/s12888-020-02798-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/30/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Variation in genes implicated in homocysteine and lipid metabolism systems may influence antidepressant response for patients with major depressive disorder (MDD). This study aimed to investigate whether association of polymorphisms on the MTHFR, ApoE and ApoA4 genes with the treatment response in MDD subjects. METHODS A total of 281 Han Chinese MDD patients received a single antidepressant drug (SSRI or SNRI) for at least 6 weeks, among whom 275 were followed up for 8 weeks. Their response to 6 weeks' treatment and remission to 8 weeks' treatment with antidepressant drugs was determined by changes in the 17-item Hamilton Depression Rating Scale (HARS-17) score. Single SNP and haplotype associations with treatment response were analyzed by UNPHASED 3.0.13. Logistic regression analysis was used to explore the interactions between genotypes and gender or drug type on treatment outcome, only those SNPs that had interactional association with gender or drug type were subjected to further stratified analysis. RESULTS In total group, the haplotype (C-A) in MTHFR (rsl801133 and rs1801131) and the ApoE rs405509 AA genotype were significantly associated with better efficacy of antidepressants; In gender subgroups, only haplotype (C-A) in MTHFR (rsl801133 and rs1801131) was significantly associated with better efficacy of antidepressants in male subgroup; In drug type subgroup, the haplotype (C-A) in MTHFR (rsl801133 and rs1801131) and haplotype (G-C) in ApoE (rs7412 and rs405509) were associated with better efficacy of antidepressants in SNRI treated subgroup; The ApoA4 rs5092 G allele and GG genotype were associated with worse efficacy of antidepressants in SNRI treated subgroup. CONCLUSIONS Genetic polymorphisms in homocysteine and lipid metabolism systems are associated with antidepressant response, particularly for the interactions of the certain genetic with gender or drug type.
Collapse
Affiliation(s)
- Baoyu Yuan
- grid.263826.b0000 0004 1761 0489Department of Neurology, Affiliated ZhongDa Hospital, School of Medical, Southeast University, No.87 Ding Jia Qiao Road, Nanjing, 210009 Jiangsu China
| | - Xiaoyan Sun
- grid.263826.b0000 0004 1761 0489Department of Neurology, Affiliated ZhongDa Hospital, School of Medical, Southeast University, No.87 Ding Jia Qiao Road, Nanjing, 210009 Jiangsu China
| | - Zhi Xu
- grid.263826.b0000 0004 1761 0489Institution of Neuropsychiatry, Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Department of Psychosomatics and Psychiatry, Affiliated ZhongDa Hospital Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009 Jiangsu China
| | - Mengjia Pu
- grid.263826.b0000 0004 1761 0489Department of Neurology, Affiliated ZhongDa Hospital, School of Medical, Southeast University, No.87 Ding Jia Qiao Road, Nanjing, 210009 Jiangsu China
| | - Yonggui Yuan
- grid.263826.b0000 0004 1761 0489Institution of Neuropsychiatry, Southeast University, Nanjing, 210009 Jiangsu China ,grid.263826.b0000 0004 1761 0489Department of Psychosomatics and Psychiatry, Affiliated ZhongDa Hospital Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009 Jiangsu China
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medical, Southeast University, No.87 Ding Jia Qiao Road, Nanjing, 210009, Jiangsu, China. .,Institution of Neuropsychiatry, Southeast University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
29
|
Zhou Z, Shek DT, Zhu X, Dou D. Positive Youth Development and Adolescent Depression: A Longitudinal Study Based on Mainland Chinese High School Students. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E4457. [PMID: 32575856 PMCID: PMC7344806 DOI: 10.3390/ijerph17124457] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/13/2020] [Accepted: 06/19/2020] [Indexed: 12/26/2022]
Abstract
There are several limitations of the scientific literature on the linkage between positive youth development (PYD) attributes and adolescent psychological morbidity. First, longitudinal studies in the field are limited. Second, few studies have used validated PYD measures to explore the related issues. Third, few studies have used large samples. Fourth, limited studies have been conducted in mainland China. In this study, we conducted a longitudinal study using two waves of data collected from 2648 junior high school students in mainland China. In each wave, participants responded to a validated PYD scale (Chinese Positive Youth Development Scale: CPYDS) and other measures of well-being, including the 20-item Centre for Epidemiologic Studies Depression Scale (CES-D). After controlling for the background demographic variables, different measures of CPYDS (cognitive-behavioral competence, prosocial attributes, general positive youth development qualities, positive identity, and overall PYD qualities) were negatively associated with CES-D scores in Wave 1 and Wave 2. Longitudinal analyses also revealed that PYD measures in Wave 1 negatively predicted Wave 2 depression scores and the changes over time. The present findings highlight the protective role of PYD attributes in protecting adolescents from depression.
Collapse
Affiliation(s)
- Zheng Zhou
- Research Institute of Social Development, Southwestern University of Finance and Economics, Chengdu 611130, China;
| | - Daniel T.L. Shek
- Department of Applied Social Sciences, The Hong Kong Polytechnic University, Hong Kong, China; (X.Z.); (D.D.)
| | - Xiaoqin Zhu
- Department of Applied Social Sciences, The Hong Kong Polytechnic University, Hong Kong, China; (X.Z.); (D.D.)
| | - Diya Dou
- Department of Applied Social Sciences, The Hong Kong Polytechnic University, Hong Kong, China; (X.Z.); (D.D.)
| |
Collapse
|
30
|
Li HJ, Qu N, Hui L, Cai X, Zhang CY, Zhong BL, Zhang SF, Chen J, Xia B, Wang L, Jia QF, Li W, Chang H, Xiao X, Li M, Li Y. Further confirmation of netrin 1 receptor (DCC) as a depression risk gene via integrations of multi-omics data. Transl Psychiatry 2020; 10:98. [PMID: 32184385 PMCID: PMC7078234 DOI: 10.1038/s41398-020-0777-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/21/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022] Open
Abstract
Genome-wide association studies (GWAS) of major depression and its relevant biological phenotypes have been extensively conducted in large samples, and transcriptome-wide analyses in the tissues of brain regions relevant to pathogenesis of depression, e.g., dorsolateral prefrontal cortex (DLPFC), have also been widely performed recently. Integrating these multi-omics data will enable unveiling of depression risk genes and even underlying pathological mechanisms. Here, we employ summary data-based Mendelian randomization (SMR) and integrative risk gene selector (iRIGS) approaches to integrate multi-omics data from GWAS, DLPFC expression quantitative trait loci (eQTL) analyses and enhancer-promoter physical link studies to prioritize high-confidence risk genes for depression, followed by independent replications across distinct populations. These integrative analyses identify multiple high-confidence depression risk genes, and numerous lines of evidence supporting pivotal roles of the netrin 1 receptor (DCC) gene in this illness across different populations. Our subsequent explorative analyses further suggest that DCC significantly predicts neuroticism, well-being spectrum, cognitive function and putamen structure in general populations. Gene expression correlation and pathway analyses in DLPFC further show that DCC potentially participates in the biological processes and pathways underlying synaptic plasticity, axon guidance, circadian entrainment, as well as learning and long-term potentiation. These results are in agreement with the recent findings of this gene in neurodevelopment and psychiatric disorders, and we thus further confirm that DCC is an important susceptibility gene for depression, and might be a potential target for new antidepressants.
Collapse
Affiliation(s)
- Hui-Juan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Na Qu
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Li Hui
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xin Cai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Bao-Liang Zhong
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Shu-Fang Zhang
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Jing Chen
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Bin Xia
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China
| | - Lu Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qiu-Fang Jia
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Wei Li
- Department of Blood Transfusion, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hong Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Yi Li
- Affiliated Wuhan Mental Health Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Research Center for Psychological and Health Sciences, China University of Geosciences, Wuhan, Hubei, China.
| |
Collapse
|
31
|
Xu Q, Guo L, Cheng J, Wang M, Geng Z, Zhu W, Zhang B, Liao W, Qiu S, Zhang H, Xu X, Yu Y, Gao B, Han T, Yao Z, Cui G, Liu F, Qin W, Zhang Q, Li MJ, Liang M, Chen F, Xian J, Li J, Zhang J, Zuo XN, Wang D, Shen W, Miao Y, Yuan F, Lui S, Zhang X, Xu K, Zhang LJ, Ye Z, Yu C. CHIMGEN: a Chinese imaging genetics cohort to enhance cross-ethnic and cross-geographic brain research. Mol Psychiatry 2020; 25:517-529. [PMID: 31827248 PMCID: PMC7042768 DOI: 10.1038/s41380-019-0627-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/21/2019] [Accepted: 11/27/2019] [Indexed: 02/05/2023]
Abstract
The Chinese Imaging Genetics (CHIMGEN) study establishes the largest Chinese neuroimaging genetics cohort and aims to identify genetic and environmental factors and their interactions that are associated with neuroimaging and behavioral phenotypes. This study prospectively collected genomic, neuroimaging, environmental, and behavioral data from more than 7000 healthy Chinese Han participants aged 18-30 years. As a pioneer of large-sample neuroimaging genetics cohorts of non-Caucasian populations, this cohort can provide new insights into ethnic differences in genetic-neuroimaging associations by being compared with Caucasian cohorts. In addition to micro-environmental measurements, this study also collects hundreds of quantitative macro-environmental measurements from remote sensing and national survey databases based on the locations of each participant from birth to present, which will facilitate discoveries of new environmental factors associated with neuroimaging phenotypes. With lifespan environmental measurements, this study can also provide insights on the macro-environmental exposures that affect the human brain as well as their timing and mechanisms of action.
Collapse
Affiliation(s)
- Qiang Xu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, 300052, Tianjin, China
| | - Lining Guo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, 300052, Tianjin, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Meiyun Wang
- Department of Radiology, Zhengzhou University People's Hospital and Henan Provincial People's Hospital, 450003, Zhengzhou, China
- Henan Key Laboratory for Medical Imaging of Neurological Diseases, 450003, Zhengzhou, China
| | - Zuojun Geng
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, 050000, Shijiazhuang, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | - Bing Zhang
- Department of Radiology, Drum Tower Hospital, Medical School of Nanjing University, 210008, Nanjing, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, 410008, Changsha, China
- National Clinical Research Center for Geriatric Disorder, 410008, Changsha, China
| | - Shijun Qiu
- Department of Medical Imaging, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, 510405, Guangzhou, China
| | - Hui Zhang
- Department of Radiology, The First Hospital of Shanxi Medical University, 030001, Taiyuan, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, 310009, Hangzhou, China
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China
| | - Bo Gao
- Department of Radiology, Yantai Yuhuangding Hospital, 264000, Yantai, China
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, 300350, Tianjin, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, 300350, Tianjin, China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hosptial, Fudan University, 200040, Shanghai, China
| | - Guangbin Cui
- Functional and Molecular Imaging Key Lab of Shaanxi Province & Department of Radiology, Tangdu Hospital, The Military Medical University of PLA Airforce (Fourth Military Medical University), 710038, Xi'an, China
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, 300052, Tianjin, China
| | - Wen Qin
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, 300052, Tianjin, China
| | - Quan Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, 300052, Tianjin, China
| | - Mulin Jun Li
- Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Meng Liang
- School of Medical Imaging, Tianjin Medical University, 300203, Tianjin, China
| | - Feng Chen
- Department of Radiology, Hainan General Hospital, 570311, Haikou, China
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, 100730, Beijing, China
| | - Jiance Li
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Jing Zhang
- Department of Magnetic Resonance, Lanzhou University Second Hospital, 730050, Lanzhou, China
| | - Xi-Nian Zuo
- Department of Psychology, University of Chinese Academy of Sciences (CAS), 100049, Beijing, China
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, 100101, Beijing, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, 250012, Jinan, China
| | - Wen Shen
- Department of Radiology, Tianjin First Center Hospital, 300192, Tianjin, China
| | - Yanwei Miao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Fei Yuan
- Department of Radiology, Pingjin Hospital, Logistics University of Chinese People's Armed Police Forces, 300162, Tianjin, China
| | - Su Lui
- Department of Radiology, The Center for Medical Imaging, West China Hospital of Sichuan University, 610041, Chengdu, China
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325000, Wenzhou, China
| | - Xiaochu Zhang
- CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, 230026, Hefei, China
- School of Life Sciences, University of Science & Technology of China, 230026, Hefei, China
| | - Kai Xu
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, 221006, Xuzhou, China
- School of Medical Imaging, Xuzhou Medical University, 221004, Xuzhou, China
| | - Long Jiang Zhang
- Department of Medical Imaging, Jinling Hospital, Medical School of Nanjing University, 210002, Nanjing, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, 300060, Tianjin, China
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, 300052, Tianjin, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
32
|
van den Berg M, Krauskopf J, Ramaekers J, Kleinjans J, Prickaerts J, Briedé J. Circulating microRNAs as potential biomarkers for psychiatric and neurodegenerative disorders. Prog Neurobiol 2020; 185:101732. [DOI: 10.1016/j.pneurobio.2019.101732] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/25/2019] [Accepted: 12/03/2019] [Indexed: 12/21/2022]
|
33
|
So HC, Chau CKL, Lau A, Wong SY, Zhao K. Translating GWAS findings into therapies for depression and anxiety disorders: gene-set analyses reveal enrichment of psychiatric drug classes and implications for drug repositioning. Psychol Med 2019; 49:2692-2708. [PMID: 30569882 DOI: 10.1017/s0033291718003641] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Depression and anxiety disorders (AD) are the first and sixth leading causes of disability worldwide. Despite their high prevalence and significant disability resulted, there are limited advances in new drug development. Recently, genome-wide association studies (GWAS) have greatly advanced our understanding of the genetic basis underlying psychiatric disorders. METHODS Here we employed gene-set analyses of GWAS summary statistics for drug repositioning. We explored five related GWAS datasets, including two on major depressive disorder (MDD2018 and MDD-CONVERGE, with the latter focusing on severe melancholic depression), one on AD, and two on depressive symptoms and neuroticism in the population. We extracted gene-sets associated with each drug from DSigDB and examined their association with each GWAS phenotype. We also performed repositioning analyses on meta-analyzed GWAS data, integrating evidence from all related phenotypes. RESULTS Importantly, we showed that the repositioning hits are generally enriched for known psychiatric medications or those considered in clinical trials. Enrichment was seen for antidepressants and anxiolytics but also for antipsychotics. We also revealed new candidates or drug classes for repositioning, some of which were supported by experimental or clinical studies. For example, the top repositioning hit using meta-analyzed p values was fendiline, which was shown to produce antidepressant-like effects in mouse models by inhibition of acid sphingomyelinase. CONCLUSION Taken together, our findings suggest that human genomic data such as GWAS are useful in guiding drug discoveries for depression and AD.
Collapse
Affiliation(s)
- Hon-Cheong So
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Zoology Institute of Zoology and The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Carlos Kwan-Long Chau
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Alexandria Lau
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Sze-Yung Wong
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kai Zhao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
34
|
Peterson RE, Kuchenbaecker K, Walters RK, Chen CY, Popejoy AB, Periyasamy S, Lam M, Iyegbe C, Strawbridge RJ, Brick L, Carey CE, Martin AR, Meyers JL, Su J, Chen J, Edwards AC, Kalungi A, Koen N, Majara L, Schwarz E, Smoller JW, Stahl EA, Sullivan PF, Vassos E, Mowry B, Prieto ML, Cuellar-Barboza A, Bigdeli TB, Edenberg HJ, Huang H, Duncan LE. Genome-wide Association Studies in Ancestrally Diverse Populations: Opportunities, Methods, Pitfalls, and Recommendations. Cell 2019; 179:589-603. [PMID: 31607513 PMCID: PMC6939869 DOI: 10.1016/j.cell.2019.08.051] [Citation(s) in RCA: 464] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/10/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022]
Abstract
Genome-wide association studies (GWASs) have focused primarily on populations of European descent, but it is essential that diverse populations become better represented. Increasing diversity among study participants will advance our understanding of genetic architecture in all populations and ensure that genetic research is broadly applicable. To facilitate and promote research in multi-ancestry and admixed cohorts, we outline key methodological considerations and highlight opportunities, challenges, solutions, and areas in need of development. Despite the perception that analyzing genetic data from diverse populations is difficult, it is scientifically and ethically imperative, and there is an expanding analytical toolbox to do it well.
Collapse
Affiliation(s)
- Roseann E Peterson
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Karoline Kuchenbaecker
- Division of Psychiatry and UCL Genetics Institute, University College London, London W1T 7NF, UK
| | - Raymond K Walters
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Chia-Yen Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alice B Popejoy
- Department of Biomedical Data Science, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sathish Periyasamy
- Queensland Brain Institute and Queensland Centre for Mental Health Research, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Max Lam
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Conrad Iyegbe
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Rona J Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Glasgow G12 8RZ, UK; Department of Medicine Solna, Karolinska Institute, Stockholm, SE 17176, Sweden
| | - Leslie Brick
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, RI 02906, USA
| | - Caitlin E Carey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alicia R Martin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jacquelyn L Meyers
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Jinni Su
- Department of Psychology, Arizona State University, Tempe, AZ 85281, USA
| | - Junfang Chen
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Alexis C Edwards
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Allan Kalungi
- Mental Health Section of MRC/UVRI and LSHTM Uganda Research Unit, P.O. Box 49, Entebbe, Uganda; Department of Psychiatry, Faculty of Medicine & Health Sciences, University of Stellenbosch, Cape Town, South Africa; Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda; Global Initiative for Neuropsychiatric Genetics Education in Research, Harvard T.H. Chan School of Public Health and Broad Institute, Boston, MA 02115, USA
| | - Nastassja Koen
- Department of Psychiatry, Faculty of Medicine & Health Sciences, University of Stellenbosch, Cape Town, South Africa; Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda; Global Initiative for Neuropsychiatric Genetics Education in Research, Harvard T.H. Chan School of Public Health and Broad Institute, Boston, MA 02115, USA
| | - Lerato Majara
- Global Initiative for Neuropsychiatric Genetics Education in Research, Harvard T.H. Chan School of Public Health and Broad Institute, Boston, MA 02115, USA; MRC Human Genetics Research Unit, Division of Human Genetics, Department of Pathology, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, 7925, South Africa
| | - Emanuel Schwarz
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, 68159 Mannheim, Germany
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Eli A Stahl
- Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Patrick F Sullivan
- Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, SE 17176, Sweden; Genetics and Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Evangelos Vassos
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - Bryan Mowry
- Queensland Brain Institute and Queensland Centre for Mental Health Research, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Miguel L Prieto
- Department of Psychiatry, Faculty of Medicine, Universidad de los Andes, Santiago 7620001, Chile; Mental Health Service, Clínica Universidad de los Andes, Santiago 7620001, Chile; Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Alfredo Cuellar-Barboza
- Department of Psychiatry, University Hospital and School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey, Mexico; Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Tim B Bigdeli
- Department of Psychiatry, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hailiang Huang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Laramie E Duncan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
35
|
Gene-environment interactions between HPA-axis genes and stressful life events in depression: a systematic review. Acta Neuropsychiatr 2019; 31:186-192. [PMID: 31106715 DOI: 10.1017/neu.2019.16] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Depression is a disorder caused by genetics and environmental factors. The aim of this study was to perform a review investigating the interaction between genetic variations located in genes involved in hypothalamus-pituitary-adrenal axis (HPA-axis) and stressful life events (SLEs) in depression. METHODS In this systematic review, we selected articles investigating the interaction between genes involved in the HPA-axis, such as Arginine Vasopressin (AVP), Angiotensin Converting Enzyme (ACE), Corticotrophin Releasing Hormone (CRH), Corticotrophin Releasing Hormone Receptor 1 (CRHR1), Corticotrophin Releasing Hormone Receptor 2 (CRHR2), FK506 binding protein (FKBP5), Nuclear Receptor subfamily 3 group C member 1 (NR3C1), Nuclear Receptor subfamily 3 group C member 2 (NR3C2), and SLE. The literature search was conducted using the Pubmed, Embase, and PsychINFO databases in adherence with the PRISMA guidelines. RESULTS The search yielded 48 potentially relevant studies, of which 40 were excluded following screening. Eight studies were included in the final review. A total of 97 single nucleotide polymorphisms (SNPs) were examined in the eight included studies. The most prevalent gene was FKBP5, and the best studied polymorphism was FKBP5:rs1360780. Two of the five studies reported significant gene-environment (G × E) interactions between rs1360780 and SLE. Overall, four studies reported significant G × E interactions between FKBP5, CRH, or CRHR1 and SLE, respectively. No significant G × E interactions were found for the remaining genes. CONCLUSIONS Our results suggest that genetic variation in three genes in the HPA-axis possibly moderate the effects of SLEs in depression.
Collapse
|
36
|
Li H, Chang H, Song X, Liu W, Li L, Wang L, Yang Y, Zhang L, Li W, Zhang Y, Zhou DS, Li X, Zhang C, Fang Y, Sun Y, Dai JP, Luo XJ, Yao YG, Xiao X, Lv L, Li M. Integrative analyses of major histocompatibility complex loci in the genome-wide association studies of major depressive disorder. Neuropsychopharmacology 2019; 44:1552-1561. [PMID: 30771788 PMCID: PMC6785001 DOI: 10.1038/s41386-019-0346-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/01/2019] [Accepted: 02/12/2019] [Indexed: 11/09/2022]
Abstract
Recent European genome-wide association studies (GWAS) have revealed strong statistical correlations between MDD and numerous zero-to-high linked variants in the genomic region containing major histocompatibility complex (MHC) genes (MHC region), but the underlying biological mechanisms are still unclear. To better understand the roles of this genomic region in the neurobiology of MDD, we applied a convergent functional genomics approach to integrate GWAS data of MDD relevant biological phenotypes, gene-expression analyses results obtained from brain samples, and genetic analyses of independent Chinese MDD samples. We observed that independent MDD risk variants in the MHC region were also significantly associated with the relevant biological phenotypes in the predicted directions, including the emotional and cognitive-related phenotypes. Gene-expression analyses further revealed that mRNA expression levels of several MHC region genes in the human brain were associated with MDD risk SNPs and diagnostic status. For instance, a brain-enriched gene ZNF603P consistently showed lower mRNA levels in the individuals carrying MDD risk alleles and in MDD patients. Remarkably, we further found that independent MDD risk SNPs in the MHC region likely converged to affect the mRNA level(s) of the same gene(s), and Europeans and Han Chinese populations have a substantial shared genetic and molecular basis underlying MDD risk associations in the MHC region. These results highlighted several potential pivotal genes at the MHC region in the pathogenesis of MDD. Their common impacts on multiple psychiatric relevant phenotypes also implicated the neurological processes shared by different psychological processes, such as mood and/or cognition, shedding lights on their potential biological mechanisms.
Collapse
Affiliation(s)
- Huijuan Li
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China ,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan China
| | - Hong Chang
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China
| | - Xueqin Song
- grid.412633.1The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Weipeng Liu
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China ,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan China
| | - Lingyi Li
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China
| | - Lu Wang
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China
| | - Yongfeng Yang
- 0000 0004 1808 322Xgrid.412990.7Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China ,0000 0004 1808 322Xgrid.412990.7Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Luwen Zhang
- 0000 0004 1808 322Xgrid.412990.7Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China ,0000 0004 1808 322Xgrid.412990.7Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Wenqiang Li
- 0000 0004 1808 322Xgrid.412990.7Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China ,0000 0004 1808 322Xgrid.412990.7Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Yan Zhang
- 0000 0004 1808 322Xgrid.412990.7Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China ,0000 0004 1808 322Xgrid.412990.7Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Dong-Sheng Zhou
- 0000 0004 1782 599Xgrid.452715.0Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang China
| | - Xingxing Li
- 0000 0004 1782 599Xgrid.452715.0Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang China
| | - Chen Zhang
- 0000 0004 0368 8293grid.16821.3cShanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiru Fang
- 0000 0004 0368 8293grid.16821.3cShanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Sun
- 0000 0000 9147 9053grid.412692.aWuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei China ,Chinese Brain Bank Center, Wuhan, Hubei China
| | - Jia-Pei Dai
- 0000 0000 9147 9053grid.412692.aWuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei China ,Chinese Brain Bank Center, Wuhan, Hubei China
| | - Xiong-Jian Luo
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China ,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan China ,0000000119573309grid.9227.eCenter for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan China
| | - Yong-Gang Yao
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China ,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan China ,0000000119573309grid.9227.eCAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Luxian Lv
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China. .,Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan, China. .,Henan Province People's Hospital, Zhengzhou, Henan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
37
|
Ikeda M, Takahashi A, Kamatani Y, Momozawa Y, Saito T, Kondo K, Shimasaki A, Kawase K, Sakusabe T, Iwayama Y, Toyota T, Wakuda T, Kikuchi M, Kanahara N, Yamamori H, Yasuda Y, Watanabe Y, Hoya S, Aleksic B, Kushima I, Arai H, Takaki M, Hattori K, Kunugi H, Okahisa Y, Ohnuma T, Ozaki N, Someya T, Hashimoto R, Yoshikawa T, Kubo M, Iwata N. Genome-Wide Association Study Detected Novel Susceptibility Genes for Schizophrenia and Shared Trans-Populations/Diseases Genetic Effect. Schizophr Bull 2019; 45:824-834. [PMID: 30285260 PMCID: PMC6581133 DOI: 10.1093/schbul/sby140] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Genome-wide association studies (GWASs) have identified >100 susceptibility loci for schizophrenia (SCZ) and demonstrated that SCZ is a polygenic disorder determined by numerous genetic variants but with small effect size. We conducted a GWAS in the Japanese (JPN) population (a) to detect novel SCZ-susceptibility genes and (b) to examine the shared genetic risk of SCZ across (East Asian [EAS] and European [EUR]) populations and/or that of trans-diseases (SCZ, bipolar disorder [BD], and major depressive disorder [MDD]) within EAS and between EAS and EUR (trans-diseases/populations). Among the discovery GWAS subjects (JPN-SCZ GWAS: 1940 SCZ cases and 7408 controls) and replication dataset (4071 SCZ cases and 54479 controls), both comprising JPN populations, 3 novel susceptibility loci for SCZ were identified: SPHKAP (Pbest = 4.1 × 10-10), SLC38A3 (Pbest = 5.7 × 10-10), and CABP1-ACADS (Pbest = 9.8 × 10-9). Subsequent meta-analysis between our samples and those of the Psychiatric GWAS Consortium (PGC; EUR samples) and another study detected 12 additional susceptibility loci. Polygenic risk score (PRS) prediction revealed a shared genetic risk of SCZ across populations (Pbest = 4.0 × 10-11) and between SCZ and BD in the JPN population (P ~ 10-40); however, a lower variance-explained was noted between JPN-SCZ GWAS and PGC-BD or MDD within/across populations. Genetic correlation analysis supported the PRS results; the genetic correlation between JPN-SCZ and PGC-SCZ was ρ = 0.58, whereas a similar/lower correlation was observed between the trans-diseases (JPN-SCZ vs JPN-BD/EAS-MDD, rg = 0.56/0.29) or trans-diseases/populations (JPN-SCZ vs PGC-BD/MDD, ρ = 0.38/0.12). In conclusion, (a) Fifteen novel loci are possible susceptibility genes for SCZ and (b) SCZ "risk" effect is shared with other psychiatric disorders even across populations.
Collapse
Affiliation(s)
- Masashi Ikeda
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan,Department of Genomic Medicine, Research Institute, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan,Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takeo Saito
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kenji Kondo
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Ayu Shimasaki
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kohei Kawase
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan
| | - Takaya Sakusabe
- Faculty of Clinical Engineering, Fujita Health University, School of Health Sciences, Toyoake, Japan
| | - Yoshimi Iwayama
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Tomoko Toyota
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Tomoyasu Wakuda
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Mitsuru Kikuchi
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Nobuhisa Kanahara
- Department of Psychiatry, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hidenaga Yamamori
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuka Yasuda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuichiro Watanabe
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Satoshi Hoya
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Heii Arai
- Depearmtnt of Psychaitry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Manabu Takaki
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kotaro Hattori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuko Okahisa
- Department of Neuropsychiatry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tohru Ohnuma
- Depearmtnt of Psychaitry, Juntendo University, Faculty of Medicine, Tokyo, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Toshiyuki Someya
- Department of Psychiatry, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ryota Hashimoto
- Department of Pathology of Mental Diseases, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan,Osaka University, Suita, Japan
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan,To whom correspondence should be addressed; Department of Psychiatry, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan; tel: 81-562-93-9250, fax: 81-562-93-1831, e-mail:
| |
Collapse
|
38
|
Abstract
The promise of personalized genomic medicine is that knowledge of a person's gene sequences and activity will facilitate more appropriate medical interventions, particularly drug prescriptions, to reduce the burden of disease. Early successes in oncology and pediatrics have affirmed the power of positive diagnosis and are mostly based on detection of one or a few mutations that drive the specific pathology. However, genetically more complex diseases require the development of polygenic risk scores (PRSs) that have variable accuracy. The rarity of events often means that they have necessarily low precision: many called positives are actually not at risk, and only a fraction of cases are prevented by targeted therapy. In some situations, negative prediction may better define the population at low risk. Here, I review five conditions across a broad spectrum of chronic disease (opioid pain medication, hypertension, type 2 diabetes, major depression, and osteoporotic bone fracture), considering in each case how genetic prediction might be used to target drug prescription. This leads to a call for more research designed to evaluate genetic likelihood of response to therapy and a call for evaluation of PRS, not just in terms of sensitivity and specificity but also with respect to potential clinical efficacy.
Collapse
Affiliation(s)
- Greg Gibson
- Center for Integrative Genomics and School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
39
|
Li JJ, Hilton EC, Lu Q, Hong J, Greenberg JS, Mailick MR. Validating psychosocial pathways of risk between neuroticism and late life depression using a polygenic score approach. JOURNAL OF ABNORMAL PSYCHOLOGY 2019; 128:200-211. [PMID: 30829503 PMCID: PMC6462143 DOI: 10.1037/abn0000419] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neuroticism is a stable and heritable personality trait that is strongly linked to depression. Yet, little is known about its association with late life depression, as well as how neuroticism eventuates into depression. This study used data from the Wisconsin Longitudinal Study (WLS; N = 4,877) to examine the direct and indirect effects of neuroticism on late life depression at 3 points in the life course-ages 53, 64, and 71-via stressful life events (i.e., independent and dependent) and social supports measured across adulthood and into later life. Neuroticism was assayed using multiple methods, including self-report measures (phenotypic model) and a polygenic score (polygenic model) informed by a meta-analytic genome-wide association study. Results indicated that the phenotypic model of neuroticism and late life depression was partially mediated via dependent stressful life events experienced after the age of 53 and by age 64 social support. This association was replicated in the polygenic model of neuroticism, providing key evidence that the findings are robust. No indirect effects emerged with respect to age 53 social support, age 71 social support, adult dependent stressful life events (experienced between age 19 and 52), and adult and late life independent stressful life events in either the phenotypic or polygenic models as they pertained to late life depression. Results are consistent with previous findings that individuals with high neuroticism may be vulnerable to late life depression through psychosocial risk factors that are, in part, attributable to their own personality. (PsycINFO Database Record (c) 2019 APA, all rights reserved).
Collapse
Affiliation(s)
- James J. Li
- Department of Psychology, University of Wisconsin-Madison, 1202 West Johnson St., Madison, Wisconsin, 53706, USA
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705, USA
| | - Emily C. Hilton
- Department of Psychology, University of Wisconsin-Madison, 1202 West Johnson St., Madison, Wisconsin, 53706, USA
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705, USA
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI 53792, USA
| | - Jinkuk Hong
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705, USA
| | - Jan S. Greenberg
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705, USA
- School of Social Work, University of Wisconsin-Madison, 1350 University Avenue, Madison, WI 53706, USA
| | - Marsha R. Mailick
- Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave, Madison, WI 53705, USA
| |
Collapse
|
40
|
Li M, Yue W. VRK2, a Candidate Gene for Psychiatric and Neurological Disorders. MOLECULAR NEUROPSYCHIATRY 2018; 4:119-133. [PMID: 30643786 PMCID: PMC6323383 DOI: 10.1159/000493941] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 09/20/2018] [Indexed: 12/20/2022]
Abstract
Recent large-scale genetic approaches, such as genome-wide association studies, have identified multiple genetic variations that contribute to the risk of mental illnesses, among which single nucleotide polymorphisms (SNPs) within or near the vaccinia related kinase 2 (VRK2) gene have gained consistent support for their correlations with multiple psychiatric and neurological disorders including schizophrenia (SCZ), major depressive disorder (MDD), and genetic generalized epilepsy. For instance, the genetic variant rs1518395 in VRK2 showed genome-wide significant associations with SCZ (35,476 cases and 46,839 controls, p = 3.43 × 10-8) and MDD (130,620 cases and 347,620 controls, p = 4.32 × 10-12) in European populations. This SNP was also genome-wide significantly associated with SCZ in Han Chinese population (12,083 cases and 24,097 controls, p = 3.78 × 10-13), and all associations were in the same direction of allelic effects. These studies highlight the potential roles of VRK2 in the central nervous system, and this gene therefore might be a good candidate to investigate the shared genetic and molecular basis between SCZ and MDD, as it is one of the few genes known to show genome-wide significant associations with both illnesses. Furthermore, the VRK2 gene was found to be involved in multiple other congenital deficits related to the malfunction of neurodevelopment, adding further support for the involvement of this gene in the pathogenesis of these neurological and psychiatric illnesses. While the precise function of VRK2 in these conditions remains unclear, preliminary evidence suggests that it may affect neuronal proliferation and migration via interacting with multiple essential signaling pathways involving other susceptibility genes/proteins for psychiatric disorders. Here, we have reviewed the recent progress of genetic and molecular studies of VRK2, with an emphasis on its role in psychiatric illnesses and neurological functions. We believe that attention to this important gene is necessary, and further investigations of VRK2 may provide hints into the underlying mechanisms of SCZ and MDD.
Collapse
Affiliation(s)
- Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Weihua Yue
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China
- Key Laboratory of Mental Health, Ministry of Health (Peking University) and National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| |
Collapse
|
41
|
Abstract
Major depression is a common illness that severely limits psychosocial functioning and diminishes quality of life. In 2008, WHO ranked major depression as the third cause of burden of disease worldwide and projected that the disease will rank first by 2030.1 In practice, its detection, diagnosis, and management often pose challenges for clinicians because of its various presentations, unpredictable course and prognosis, and variable response to treatment.
Collapse
Affiliation(s)
- Gin S Malhi
- Department of Academic Psychiatry, Sydney Medical School Northern, University of Sydney, Sydney, NSW, Australia; CADE Clinic, Royal North Shore Hospital, Sydney, NSW, Australia.
| | - J John Mann
- Molecular Imaging and Neuropathology Division, Department of Psychiatry, Columbia University, New York, NY, USA
| |
Collapse
|
42
|
Common variants on 6q16.2, 12q24.31 and 16p13.3 are associated with major depressive disorder. Neuropsychopharmacology 2018; 43:2146-2153. [PMID: 29728651 PMCID: PMC6098070 DOI: 10.1038/s41386-018-0078-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 04/10/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022]
Abstract
Accumulating evidence suggests that genetic factors have a role in major depressive disorder (MDD). However, only limited MDD risk loci have been identified so far. Here we perform a meta-analysis (a total of 90,150 MDD cases and 246,603 controls) through combing three genome-wide association studies of MDD, including 23andMe (cases were self-reported with a clinical diagnosis or treatment of depression), CONVERGE (cases were diagnosed using the Composite International Diagnostic Interview) and PGC (cases were diagnosed using direct structured diagnostic interview (by trained interviewers) or clinician-administered DSM-IV checklists). Genetic variants from two previously unreported loci (rs10457592 on 6q16.2 and rs2004910 on 12q24.31) showed significant associations with MDD (P < 5 × 10-8) in a total of 336,753 subjects. SNPs (a total of 171) with a P < 1 × 10-7 in the meta-analysis were further replicated in an independent sample (GS:SFHS, 2,659 MDD cases (diagnosed with DSM-IV) and 17,237 controls) and one additional risk locus (rs3785234 on 16p13.3, P = 1.57 × 10-8) was identified in the combined samples (a total of 92,809 cases and 263,840 controls). Risk variants on the identified risk loci were associated with gene expression in human brain tissues and mRNA expression analysis showed that FBXL4 and RSRC1 were significantly upregulated in brains of MDD cases compared with controls, suggesting that genetic variants may confer risk of MDD through regulating the expression of these two genes. Our study identified three novel risk loci (6q16.2, 12q24.31, and 16p13.3) for MDD and suggested that FBXL4 and RSRC1 may play a role in MDD. Further functional characterization of the identified risk genes may provide new insights for MDD pathogenesis.
Collapse
|
43
|
Peterson RE, Cai N, Dahl AW, Bigdeli TB, Edwards AC, Webb BT, Bacanu SA, Zaitlen N, Flint J, Kendler KS. Molecular Genetic Analysis Subdivided by Adversity Exposure Suggests Etiologic Heterogeneity in Major Depression. Am J Psychiatry 2018; 175:545-554. [PMID: 29495898 PMCID: PMC5988935 DOI: 10.1176/appi.ajp.2017.17060621] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The extent to which major depression is the outcome of a single biological mechanism or represents a final common pathway of multiple disease processes remains uncertain. Genetic approaches can potentially identify etiologic heterogeneity in major depression by classifying patients on the basis of their experience of major adverse events. METHOD Data are from the China, Oxford, and VCU Experimental Research on Genetic Epidemiology (CONVERGE) project, a study of Han Chinese women with recurrent major depression aimed at identifying genetic risk factors for major depression in a rigorously ascertained cohort carefully assessed for key environmental risk factors (N=9,599). To detect etiologic heterogeneity, genome-wide association studies, heritability analyses, and gene-by-environment interaction analyses were performed. RESULTS Genome-wide association studies stratified by exposure to adversity revealed three novel loci associated with major depression only in study participants with no history of adversity. Significant gene-by-environment interactions were seen between adversity and genotype at all three loci, and 13.2% of major depression liability can be attributed to genome-wide interaction with adversity exposure. The genetic risk in major depression for participants who reported major adverse life events (27%) was partially shared with that in participants who did not (73%; genetic correlation=+0.64). Together with results from simulation studies, these findings suggest etiologic heterogeneity within major depression as a function of environmental exposures. CONCLUSIONS The genetic contributions to major depression may differ between women with and those without major adverse life events. These results have implications for the molecular dissection of major depression and other complex psychiatric and biomedical diseases.
Collapse
Affiliation(s)
- Roseann E. Peterson
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia
| | - Na Cai
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, CB10 1SA Hinxton, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, CB10 1SD Hinxton, Cambridge, UK
| | - Andy W. Dahl
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Tim B. Bigdeli
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia
- State University of New York Downstate Medical Center, Brooklyn, New York
| | - Alexis C. Edwards
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia
| | - Bradley T. Webb
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia
| | - Silviu-Alin Bacanu
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, Virginia
| | - Noah Zaitlen
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - Jonathan Flint
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, California
| | | |
Collapse
|
44
|
Bogdan R, Baranger DAA, Agrawal A. Polygenic Risk Scores in Clinical Psychology: Bridging Genomic Risk to Individual Differences. Annu Rev Clin Psychol 2018; 14:119-157. [PMID: 29579395 PMCID: PMC7772939 DOI: 10.1146/annurev-clinpsy-050817-084847] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genomewide association studies (GWASs) across psychiatric phenotypes have shown that common genetic variants generally confer risk with small effect sizes (odds ratio < 1.1) that additively contribute to polygenic risk. Summary statistics derived from large discovery GWASs can be used to generate polygenic risk scores (PRS) in independent, target data sets to examine correlates of polygenic disorder liability (e.g., does genetic liability to schizophrenia predict cognition?). The intuitive appeal and generalizability of PRS have led to their widespread use and new insights into mechanisms of polygenic liability. However, when currently applied across traits they account for small amounts of variance (<3%), are relatively uninformative for clinical treatment, and, in isolation, provide no insight into molecular mechanisms. Larger GWASs are needed to increase the precision of PRS, and novel approaches integrating various data sources (e.g., multitrait analysis of GWASs) may improve the utility of current PRS.
Collapse
Affiliation(s)
- Ryan Bogdan
- BRAINLab, Department of Psychological and Brain Sciences, and Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA;
| | - David A A Baranger
- BRAINLab, Department of Psychological and Brain Sciences, and Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA;
| | - Arpana Agrawal
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
45
|
Edwards AC, Docherty AR, Moscati A, Bigdeli TB, Peterson RE, Webb BT, Bacanu SA, Hettema JM, Flint J, Kendler KS. Polygenic risk for severe psychopathology among Europeans is associated with major depressive disorder in Han Chinese women. Psychol Med 2018; 48:777-789. [PMID: 28969721 PMCID: PMC5843532 DOI: 10.1017/s0033291717002148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Previous studies have demonstrated that several major psychiatric disorders are influenced by shared genetic factors. This shared liability may influence clinical features of a given disorder (e.g. severity, age at onset). However, findings have largely been limited to European samples; little is known about the consistency of shared genetic liability across ethnicities. METHOD The relationship between polygenic risk for several major psychiatric diagnoses and major depressive disorder (MDD) was examined in a sample of unrelated Han Chinese women. Polygenic risk scores (PRSs) were generated using European discovery samples and tested in the China, Oxford, and VCU Experimental Research on Genetic Epidemiology [CONVERGE (maximum N = 10 502)], a sample ascertained for recurrent MDD. Genetic correlations between discovery phenotypes and MDD were also assessed. In addition, within-case characteristics were examined. RESULTS European-based polygenic risk for several major psychiatric disorder phenotypes was significantly associated with the MDD case status in CONVERGE. Risk for clinically significant indicators (neuroticism and subjective well-being) was also associated with case-control status. The variance accounted for by PRS for both psychopathology and for well-being was similar to estimates reported for within-ethnicity comparisons in European samples. However, European-based PRS were largely unassociated with CONVERGE family history, clinical characteristics, or comorbidity. CONCLUSIONS The shared genetic liability across severe forms of psychopathology is largely consistent across European and Han Chinese ethnicities, with little attenuation of genetic signal relative to within-ethnicity analyses. The overall absence of associations between PRS for other disorders and within-MDD variation suggests that clinical characteristics of MDD may arise due to contributions from ethnicity-specific factors and/or pathoplasticity.
Collapse
Affiliation(s)
- A. C. Edwards
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - A. R. Docherty
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, University Neuropsychiatric Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - A. Moscati
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - T. B. Bigdeli
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - R. E. Peterson
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - B. T. Webb
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - S.-A. Bacanu
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - J. M. Hettema
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - J. Flint
- Department of Psychiatry and Biobehavioral Sciences, UCLA; David Geffen School of Medicine, Center for Neurobehavioral Genetics, UCLA; and Semel Institute for Neuroscience and Human Behavior at UCLA; Los Angeles, CA, USA
| | - K. S. Kendler
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
46
|
Dunn EC, Sofer T, Wang MJ, Soare TW, Gallo LC, Gogarten SM, Kerr KF, Chen CY, Stein MB, Ursano RJ, Guo X, Jia Y, Yao J, Rotter JI, Argos M, Cai J, Perreira K, Wassertheil-Smoller S, Smoller JW. Genome-wide association study of depressive symptoms in the Hispanic Community Health Study/Study of Latinos. J Psychiatr Res 2018; 99:167-176. [PMID: 29505938 PMCID: PMC6192675 DOI: 10.1016/j.jpsychires.2017.12.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 11/28/2017] [Accepted: 12/14/2017] [Indexed: 12/12/2022]
Abstract
Although genome-wide association studies (GWAS) have identified several variants linked to depression, few GWAS of non-European populations have been performed. We conducted a genome-wide analysis of depression in a large, population-based sample of Hispanics/Latinos. Data came from 12,310 adults in the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). Past-week depressive symptoms were assessed using the 10-item Center for Epidemiological Studies of Depression Scale. Three phenotypes were examined: a total depression score, a total score modified to account for psychiatric medication use, and a score excluding anti-depressant medication users. We estimated heritability due to common variants (h2SNP), and performed a GWAS of the three phenotypes. Replication was attempted in three independent Hispanic/Latino cohorts. We also performed sex-stratified analyses, analyzed a binary trait indicating probable depression, and conducted three trans-ethnic analyses. The three phenotypes exhibited significant heritability (h2SNP = 6.3-6.9%; p = .002) in the total sample. No SNPs were genome-wide significant in analyses of the three phenotypes or the binary indicator of probable depression. In sex-stratified analyses, seven genome-wide significant SNPs (one in females; six in males) were identified, though none were supported through replication. Four out of 24 loci identified in prior GWAS were nominally associated in HCHS/SOL. There was no evidence of overlap in genetic risk factors across ancestry groups, though this may have been due to low power. We conducted the largest GWAS of depression-related phenotypes in Hispanic/Latino adults. Results underscore the genetic complexity of depressive symptoms as a phenotype in this population and suggest the need for much larger samples.
Collapse
Affiliation(s)
- Erin C Dunn
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, United States; Department of Psychiatry, Harvard Medical School, Boston, MA, United States; Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, United States.
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Min-Jung Wang
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Thomas W Soare
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, United States; Department of Psychiatry, Harvard Medical School, Boston, MA, United States; Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, United States
| | - Linda C Gallo
- Department of Psychology, San Diego State University, San Diego, CA, United States
| | - Stephanie M Gogarten
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Kathleen F Kerr
- Department of Biostatistics, University of Washington, Seattle, WA, United States
| | - Chia-Yen Chen
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, United States; Department of Psychiatry, Harvard Medical School, Boston, MA, United States; Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, United States; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Murray B Stein
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Robert J Ursano
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Yucheng Jia
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Maria Argos
- School of Public Health, University of Illinois at Chicago, Chicago, IL, United States
| | - Jianwen Cai
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Krista Perreira
- College of Arts and Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, United States; Department of Psychiatry, Harvard Medical School, Boston, MA, United States; Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, United States
| |
Collapse
|
47
|
Xiao X, Zheng F, Chang H, Ma Y, Yao YG, Luo XJ, Li M. The Gene Encoding Protocadherin 9 (PCDH9), a Novel Risk Factor for Major Depressive Disorder. Neuropsychopharmacology 2018; 43:1128-1137. [PMID: 28990594 PMCID: PMC5854803 DOI: 10.1038/npp.2017.241] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/13/2017] [Accepted: 09/27/2017] [Indexed: 12/11/2022]
Abstract
Genomic analyses have identified only a handful of robust risk loci for major depressive disorder (MDD). In addition to the published genome-wide significant genes, it is believed that there are undiscovered 'treasures' underlying the current MDD genome-wide association studies (GWASs) and gene expression data sets, and digging into these data will allow better understanding of the illness and development of new therapeutic approaches. For this purpose, we performed a meta-analytic study combining three MDD GWAS data sets (23andMe, CONVERGE, and PGC), and then conducted independent replications of significant loci in two additional samples. The genome-wide significant variants then underwent explorative analyses on MDD-related phenotypes, cognitive function alterations, and gene expression in brains. In the discovery meta-analysis, a previously unidentified single-nucleotide polymorphism (SNP) rs9540720 in the PCDH9 gene was genome-wide significantly associated with MDD (p=1.69 × 10-8 in a total of 89 610 cases and 246 603 controls), and the association was further strengthened when additional replication samples were included (p=1.20 × 10-8 in a total of 136 115 cases and 355 275 controls). The risk SNP was also associated with multiple MDD-related phenotypes and cognitive function impairment in diverse samples. Intriguingly, the risk allele of rs9540720 predicted lower PCDH9 expression, consistent with the diagnostic analysis results that PCDH9 mRNA expression levels in the brain and peripheral blood tissues were reduced in MDD patients compared with healthy controls. These convergent lines of evidence suggest that PCDH9 is likely a novel risk gene for MDD. Our study highlights the necessity and importance of excavating the public data sets to explore risk genes for MDD, and this approach is also applicable to other complex diseases.
Collapse
Affiliation(s)
- Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Fanfan Zheng
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Hong Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Yina Ma
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China,Kunming Institute of Zoology, Chinese Academy of Sciences, No. 32 Jiao-Chang Donglu, Kunming, Yunnan 650223, China, Tel: +86 871 65190162, Fax: +86 871 65190162, E-mail:
| |
Collapse
|
48
|
Wedow R, Masters RK, Mollborn S, Schnabel L, Boardman JD. Body size reference norms and subjective weight status: A gender and life course approach. SOCIAL FORCES; A SCIENTIFIC MEDIUM OF SOCIAL STUDY AND INTERPRETATION 2018; 96:1377-1409. [PMID: 29681662 PMCID: PMC5905672 DOI: 10.1093/sf/sox073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This paper uses data from The National Longitudinal Study of Adolescent to Adult Health (Add Health) to describe county-level variation in norms regarding physical weight among adolescents in the United States. We demonstrate that regardless of one's physical size, those residing in counties with a heavier weight norm are significantly less likely to see themselves as overweight than those residing in counties with a light weight norm. We further show that the local weight norm during adolescence (Wave 1) is associated with individuals' weight perceptions through adolescence and into young adulthood (Wave 4), though these associations attenuate in strength as respondents age. Our results suggest that weight norms have a stronger influence on weight perceptions among women compared to men and that the role of gender is particularly important during adolescence. We encourage life course researchers to consider the normative health environment during adolescence as an important context for understanding disparities in health and health lifestyles as people age.
Collapse
Affiliation(s)
- Robbee Wedow
- Department of Sociology, University of Colorado, Boulder, Colorado
- Health and Society Program and Population Center, Institute of Behavioral Science, University of Colorado, Boulder, Colorado
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado
| | - Ryan K. Masters
- Department of Sociology, University of Colorado, Boulder, Colorado
- Health and Society Program and Population Center, Institute of Behavioral Science, University of Colorado, Boulder, Colorado
| | - Stefanie Mollborn
- Department of Sociology, University of Colorado, Boulder, Colorado
- Health and Society Program and Population Center, Institute of Behavioral Science, University of Colorado, Boulder, Colorado
| | - Landon Schnabel
- Department of Sociology, Indiana University, Bloomington, Indiana
| | - Jason D. Boardman
- Department of Sociology, University of Colorado, Boulder, Colorado
- Health and Society Program and Population Center, Institute of Behavioral Science, University of Colorado, Boulder, Colorado
- Institute for Behavioral Genetics, University of Colorado, Boulder, Colorado
| |
Collapse
|
49
|
Shadrina M, Bondarenko EA, Slominsky PA. Genetics Factors in Major Depression Disease. Front Psychiatry 2018; 9:334. [PMID: 30083112 PMCID: PMC6065213 DOI: 10.3389/fpsyt.2018.00334] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/02/2018] [Indexed: 12/22/2022] Open
Abstract
Depressive disorders (DDs) are one of the most widespread forms of psychiatric pathology. According to the World Health Organization, about 350 million people in the world are affected by this condition. Family and twin studies have demonstrated that the contribution of genetic factors to the risk of the onset of DDs is quite large. Various methodological approaches (analysis of candidate genes, genome-wide association analysis, genome-wide sequencing) have been used, and a large number of the associations between genes and different clinical DD variants and DD subphenotypes have been published. However, in most cases, these associations have not been confirmed in replication studies, and only a small number of genes have been proven to be associated with DD development risk. To ascertain the role of genetic factors in DD pathogenesis, further investigations of the relevant conditions are required. Special consideration should be given to the polygenic characteristics noted in whole-genome studies of the heritability of the disorder without a pronounced effect of the major gene. These observations accentuate the relevance of the analysis of gene-interaction roles in DD development and progression. It is important that association studies of the inherited variants of the genome should be supported by analysis of dynamic changes during DD progression. Epigenetic changes that cause modifications of a gene's functional state without changing its coding sequence are of primary interest. However, the opportunities for studying changes in the epigenome, transcriptome, and proteome during DD are limited by the nature of the disease and the need for brain tissue analysis, which is possible only postmortem. Therefore, any association studies between DD pathogenesis and epigenetic factors must be supplemented through the use of different animal models of depression. A threefold approach comprising the combination of gene association studies, assessment of the epigenetic state in DD patients, and analysis of different "omic" changes in animal depression models will make it possible to evaluate the contribution of genetic, epigenetic, and environmental factors to the development of different forms of depression and to help develop ways to decrease the risk of depression and improve the treatment of DD.
Collapse
Affiliation(s)
- Maria Shadrina
- Laboratory of Molecular Genetics of Hereditary Diseases, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Elena A Bondarenko
- Laboratory of Molecular Genetics of Hereditary Diseases, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Petr A Slominsky
- Laboratory of Molecular Genetics of Hereditary Diseases, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
50
|
Vadodaria KC, Stern S, Marchetto MC, Gage FH. Serotonin in psychiatry: in vitro disease modeling using patient-derived neurons. Cell Tissue Res 2017; 371:161-170. [PMID: 28812143 DOI: 10.1007/s00441-017-2670-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/01/2017] [Indexed: 01/18/2023]
Abstract
Several lines of evidence implicate serotonin in the etiology of multiple psychiatric disorders, especially mood disorders, such as major depressive disorder (MDD) and bipolar disorder (BD). Much of our current understanding of biological mechanisms underlying serotonergic alterations in mood disorders comes from animal studies. Innovation in induced pluripotent stem cell and transdifferentiation technologies for deriving neurons from adult humans has enabled the study of disease-relevant cellular phenotypes in vitro. In this context, human serotonergic neurons can now be generated using three recently published methodologies. In this mini-review, we broadly discuss evidence linking altered serotonergic neurotransmission in MDD and BD and focus on recently published methods for generating human serotonergic neurons in vitro.
Collapse
Affiliation(s)
- Krishna C Vadodaria
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA
| | - Shani Stern
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA
| | - Maria C Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA.
| |
Collapse
|