دورية أكاديمية

Schizophrenia risk conferred by rare protein-truncating variants is conserved across diverse human populations.

التفاصيل البيبلوغرافية
العنوان: Schizophrenia risk conferred by rare protein-truncating variants is conserved across diverse human populations.
المؤلفون: Liu D; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. dol31@pitt.edu., Meyer D; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Fennessy B; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Feng C; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.; Wellcome Sanger Institute, Hinxton, UK., Cheng E; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Johnson JS; Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Park YJ; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Rieder MK; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Ascolillo S; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., de Pins A; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Dobbyn A; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Lebovitch D; Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Moya E; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Nguyen TH; Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA., Wilkins L; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Hassan A; University of Peshawar, Peshawar, Pakistan., Burdick KE; Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA.; Department of Psychiatry, Harvard Medical School, Boston, MA, USA., Buxbaum JD; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Domenici E; Centre for Computational and Systems Biology, Fondazione The Microsoft Research - University of Trento, Rovereto, Italy.; Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy., Frangou S; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada., Hartmann AM; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria., Laurent-Levinson C; Faculté de Médecine Sorbonne Université, Groupe de Recherche Clinique n°15-Troubles Psychiatriques et Développement, Department of Child and Adolescent Psychiatry, Hôpital Universitaire de la Pitié-Salpêtrière, Paris, France.; Centre de Référence des Maladies Rares à Expression Psychiatrique, Department of Child and Adolescent Psychiatry, AP-HP Sorbonne Université, Hôpital Universitaire de la Pitié-Salpêtrière, Paris, France., Malhotra D; Department of Neuroscience and Rare Diseases, Roche Pharma Research and Early Development, F. Hoffmann-La Roche, Basel, Switzerland., Pato CN; Department of Psychiatry and Behavioral Sciences, SUNY Downstate College of Medicine, New York, NY, USA., Pato MT; Department of Psychiatry and Behavioral Sciences, SUNY Downstate College of Medicine, New York, NY, USA., Ressler K; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.; Division of Depression and Anxiety Disorders, McLean Hospital, Belmont, MA, USA., Roussos P; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.; Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.; Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters VA Medical Center, New York, NY, USA., Rujescu D; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria., Arango C; Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.; Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain., Bertolino A; Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy., Blasi G; Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy., Bocchio-Chiavetto L; Department of Theoretical and Applied Sciences, eCampus University, Novedrate, Italy.; Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy., Campion D; INSERM U1245, Rouen, France.; Centre Hospitalier du Rouvray, Rouen, France., Carr V; Neuroscience Research Australia, Sydney, New South Wales, Australia.; School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia.; Department of Psychiatry, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia., Fullerton JM; Neuroscience Research Australia, Sydney, New South Wales, Australia.; School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia., Gennarelli M; Genetics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.; Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy., González-Peñas J; Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.; Centro de Investigación Biomédica en Red de Salud Mental, Madrid, Spain., Levinson DF; Department of Psychiatry, Stanford University, Stanford, CA, USA., Mowry B; Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.; Queensland Centre for Mental Health Research, The University of Queensland, Brisbane, Queensland, Australia., Nimgaokar VL; Department of Psychiatry, University of Pittsburgh School of Medicine, Western Psychiatric Hospital, Pittsburgh, PA, USA.; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA., Pergola G; Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy., Rampino A; Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy., Cervilla JA; Institute of Neurosciences, Biomedical Research Centre, University of Granada, Granada, Spain.; Department of Psychiatry, San Cecilio University Hospital, University of Granada, Granada, Spain., Rivera M; Institute of Neurosciences, Biomedical Research Centre, University of Granada, Granada, Spain.; Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain., Schwab SG; Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales, Australia., Wildenauer DB; The University of Western Australia, Perth, Western Australia, Australia., Daly M; Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.; Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland., Neale B; Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA., Singh T; Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA., O'Donovan MC; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK., Owen MJ; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK., Walters JT; MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK., Ayub M; University College London, London, UK.; Department of Psychiatry, Queen's University, Kingston, Ontario, Canada., Malhotra AK; Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.; Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA.; Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, New York, NY, USA., Lencz T; Department of Psychiatry, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.; Institute for Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA.; Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, New York, NY, USA., Sullivan PF; Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA.; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden., Sklar P; Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA., Stahl EA; Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.; Regeneron Pharmaceuticals, Tarrytown, NY, USA., Huckins LM; Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. laura.huckins@mssm.edu., Charney AW; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. alexander.charney@mssm.edu.; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA. alexander.charney@mssm.edu.
مؤلفون مشاركون: Psychiatric Genomics Consortium Phase 3 Targeted Sequencing of Schizophrenia Study Team
المصدر: Nature genetics [Nat Genet] 2023 Mar; Vol. 55 (3), pp. 369-376. Date of Electronic Publication: 2023 Mar 13.
نوع المنشور: Journal Article; Research Support, Non-U.S. Gov't; Research Support, N.I.H., Extramural; Research Support, U.S. Gov't, Non-P.H.S.
اللغة: English
بيانات الدورية: Publisher: Nature Pub. Co Country of Publication: United States NLM ID: 9216904 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1546-1718 (Electronic) Linking ISSN: 10614036 NLM ISO Abbreviation: Nat Genet Subsets: MEDLINE
أسماء مطبوعة: Original Publication: New York, NY : Nature Pub. Co., c1992-
مواضيع طبية MeSH: Schizophrenia*/genetics , Autistic Disorder*/genetics, Humans ; Alleles ; Genetic Predisposition to Disease ; Genome-Wide Association Study/methods
مستخلص: Schizophrenia (SCZ) is a chronic mental illness and among the most debilitating conditions encountered in medical practice. A recent landmark SCZ study of the protein-coding regions of the genome identified a causal role for ten genes and a concentration of rare variant signals in evolutionarily constrained genes 1 . This recent study-and most other large-scale human genetics studies-was mainly composed of individuals of European (EUR) ancestry, and the generalizability of the findings in non-EUR populations remains unclear. To address this gap, we designed a custom sequencing panel of 161 genes selected based on the current knowledge of SCZ genetics and sequenced a new cohort of 11,580 SCZ cases and 10,555 controls of diverse ancestries. Replicating earlier work, we found that cases carried a significantly higher burden of rare protein-truncating variants (PTVs) among evolutionarily constrained genes (odds ratio = 1.48; P = 5.4 × 10 -6 ). In meta-analyses with existing datasets totaling up to 35,828 cases and 107,877 controls, this excess burden was largely consistent across five ancestral populations. Two genes (SRRM2 and AKAP11) were newly implicated as SCZ risk genes, and one gene (PCLO) was identified as shared by individuals with SCZ and those with autism. Overall, our results lend robust support to the rare allelic spectrum of the genetic architecture of SCZ being conserved across diverse human populations.
(© 2023. The Author(s).)
References: Singh, T. et al. Rare coding variants in ten genes confer substantial risk for schizophrenia. Nature 604, 509–516 (2022). (PMID: 35396579980580210.1038/s41586-022-04556-w)
Laursen, T. M., Nordentoft, M. & Mortensen, P. B. Excess early mortality in schizophrenia. Annu. Rev. Clin. Psychol. 10, 425–448 (2014). (PMID: 2431357010.1146/annurev-clinpsy-032813-153657)
Owen, M. J., Sawa, A. & Mortensen, P. B. Schizophrenia. Lancet 388, 86–97 (2016). (PMID: 26777917494021910.1016/S0140-6736(15)01121-6)
Hjorthøj, C., Stürup, A. E., McGrath, J. J. & Nordentoft, M. Years of potential life lost and life expectancy in schizophrenia: a systematic review and meta-analysis. Lancet Psychiatry 4, 295–301 (2017). (PMID: 2823763910.1016/S2215-0366(17)30078-0)
Trubetskoy, V. et al. Mapping genomic loci implicates genes and synaptic biology in schizophrenia. Nature 604, 502–508 (2022). (PMID: 35396580939246610.1038/s41586-022-04434-5)
Marshall, C. R. et al. Contribution of copy number variants to schizophrenia from a genome-wide study of 41,321 subjects. Nat. Genet. 49, 27–35 (2017). (PMID: 2786982910.1038/ng.3725)
Lescai, F. et al. Meta-analysis of Scandinavian schizophrenia exomes. Preprint at bioRxiv https://doi.org/10.1101/836957 (2019).
Steinberg, S. et al. Truncating mutations in RBM12 are associated with psychosis. Nat. Genet. 49, 1251–1254 (2017). (PMID: 2862810910.1038/ng.3894)
Singh, T. et al. The contribution of rare variants to risk of schizophrenia in individuals with and without intellectual disability. Nat. Genet. 49, 1167–1173 (2017). (PMID: 28650482553321910.1038/ng.3903)
Genovese, G. et al. Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia. Nat. Neurosci. 19, 1433–1441 (2016). (PMID: 27694994510419210.1038/nn.4402)
Singh, T. et al. Rare loss-of-function variants in SETD1A are associated with schizophrenia and developmental disorders. Nat. Neurosci. 19, 571–577 (2016). (PMID: 26974950668926810.1038/nn.4267)
Takata, A. et al. Loss-of-function variants in schizophrenia risk and SETD1A as a candidate susceptibility gene. Neuron 82, 773–780 (2014). (PMID: 24853937438788310.1016/j.neuron.2014.04.043)
Fromer, M. et al. De novo mutations in schizophrenia implicate synaptic networks. Nature 506, 179–184 (2014). (PMID: 24463507423700210.1038/nature12929)
Purcell, S. M. et al. A polygenic burden of rare disruptive mutations in schizophrenia. Nature 506, 185–190 (2014). (PMID: 24463508413649410.1038/nature12975)
World Population Prospects 2019: Highlights (United Nations, 2019).
Popejoy, A. B. & Fullerton, S. M. Genomics is failing on diversity. Nature 538, 161–164 (2016). (PMID: 27734877508970310.1038/538161a)
Peterson, R. E. et al. Genome-wide association studies in ancestrally diverse populations: opportunities, methods, pitfalls, and recommendations. Cell 179, 589–603 (2019). (PMID: 31607513693986910.1016/j.cell.2019.08.051)
Bigdeli, T. B. et al. Contributions of common genetic variants to risk of schizophrenia among individuals of African and Latino ancestry. Mol. Psychiatry 25, 2455–2467 (2020). (PMID: 3159146510.1038/s41380-019-0517-y)
Gulsuner, S. et al. Genetics of schizophrenia in the South African Xhosa. Science 367, 569–573 (2020). (PMID: 32001654955832110.1126/science.aay8833)
Lam, M. et al. Comparative genetic architectures of schizophrenia in East Asian and European populations. Nat. Genet. 51, 1670–1678 (2019). (PMID: 31740837688512110.1038/s41588-019-0512-x)
Li, Z. et al. Genome-wide association analysis identifies 30 new susceptibility loci for schizophrenia. Nat. Genet. 49, 1576–1583 (2017). (PMID: 2899125610.1038/ng.3973)
Li, Z. et al. Genome-wide analysis of the role of copy number variation in schizophrenia risk in Chinese. Biol. Psychiatry 80, 331–337 (2016). (PMID: 2679544210.1016/j.biopsych.2015.11.012)
De Candia, T. R. et al. Additive genetic variation in schizophrenia risk is shared by populations of African and European descent. Am. J. Hum. Genet. 93, 463–470 (2013). (PMID: 23954163384587210.1016/j.ajhg.2013.07.007)
Mensah-Ablorh, A. et al. Meta-analysis of rare variant association tests in multiethnic populations. Genet. Epidemiol. 40, 57–65 (2016). (PMID: 2663901010.1002/gepi.21939)
Hindy, G. et al. Rare coding variants in 35 genes associate with circulating lipid levels—a multi-ancestry analysis of 170,000 exomes. Am. J. Hum. Genet. 109, 81–96 (2022). (PMID: 3493293810.1016/j.ajhg.2021.11.021)
Ulirsch, J. C. et al. The genetic landscape of Diamond–Blackfan anemia. Am. J. Hum. Genet. 104, 356 (2019). (PMID: 30735661637225910.1016/j.ajhg.2018.12.011)
Guo, M. H., Plummer, L., Chan, Y.-M., Hirschhorn, J. N. & Lippincott, M. F. Burden testing of rare variants identified through exome sequencing via publicly available control data. Am. J. Hum. Genet. 103, 522–534 (2018). (PMID: 30269813617428810.1016/j.ajhg.2018.08.016)
Shaw, N. D. et al. SMCHD1 mutations associated with a rare muscular dystrophy can also cause isolated arhinia and Bosma arhinia microphthalmia syndrome. Nat. Genet. 49, 238–248 (2017). (PMID: 28067909547342810.1038/ng.3743)
Zhao, J. et al. A burden of rare variants associated with extremes of gene expression in human peripheral blood. Am. J. Hum. Genet. 98, 299–309 (2016). (PMID: 26849112474636910.1016/j.ajhg.2015.12.023)
Huffman, J. E. et al. Rare and low-frequency variants and their association with plasma levels of fibrinogen, FVII, FVIII, and vWF. Blood 126, e19–e29 (2015). (PMID: 26105150456681310.1182/blood-2015-02-624551)
Singh, T., Neale, B., Daly, M. J. & SCHEMA Consortium Exome sequencing of 23,851 cases implicates novel risk genes and provides insights into the genetic architecture of schizophrenia.Eur. Neuropsychopharmacol. 29, S1098 (2019). (PMID: 10.1016/j.euroneuro.2018.08.061)
Singh, T., Neale, B., Daly, M. J. & SCHEMA Consortium Initial results from the meta-analysis of the whole-exomes of over 20,000 schizophrenia cases and 45,000 controls.Eur. Neuropsychopharmacol. 29, S813–S814 (2019). (PMID: 10.1016/j.euroneuro.2017.08.057)
Nguyen, H. T. et al. Integrative analysis of rare variants and pathway information shows convergent results between immune pathways, drug targets and epilepsy genes. Preprint at bioRxiv https://doi.org/10.1101/410100 (2018).
Nguyen, H. T. et al. Integrated Bayesian analysis of rare exonic variants to identify risk genes for schizophrenia and neurodevelopmental disorders. Genome Med. 9, 114 (2017). (PMID: 29262854573815310.1186/s13073-017-0497-y)
Samocha, K. E. et al. Regional missense constraint improves variant deleteriousness prediction. Preprint at bioRxiv https://doi.org/10.1101/148353 (2017).
McMillan, P. J. et al. Pathological tau drives ectopic nuclear speckle scaffold protein SRRM2 accumulation in neuron cytoplasm in Alzheimer’s disease. Acta Neuropathol. Commun. 9, 117 (2021). (PMID: 34187600824389010.1186/s40478-021-01219-1)
Lester, E. et al. Tau aggregates are RNA–protein assemblies that mislocalize multiple nuclear speckle components. Neuron 109, 1675–1691.e9 (2021). (PMID: 33848474814103110.1016/j.neuron.2021.03.026)
Fazeli, S. et al. A compound downregulation of SRRM2 and miR-27a-3p with upregulation of miR-27b-3p in PBMCs of Parkinson’s patients is associated with the early stage onset of disease. PLoS ONE 15, e0240855 (2020). (PMID: 33171483765476810.1371/journal.pone.0240855)
Kaplanis, J. et al. Evidence for 28 genetic disorders discovered by combining healthcare and research data. Nature 586, 757–762 (2020). (PMID: 33057194711682610.1038/s41586-020-2832-5)
Liu, S. et al. Illuminating links between cis-regulators and trans-acting variants in the human prefrontal cortex. Genome Med. 14, 133 (2022). (PMID: 36424644968587610.1186/s13073-022-01133-8)
Palmer, D. S. et al. Exome sequencing in bipolar disorder identifies AKAP11 as a risk gene shared with schizophrenia. Nat. Genet. 54, 541–547 (2022). (PMID: 35410376911746710.1038/s41588-022-01034-x)
Satterstrom, F. K. et al. Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism. Cell 180, 568–584.e23 (2020). (PMID: 31981491725048510.1016/j.cell.2019.12.036)
Bigdeli, T. B. et al. Genome-wide association studies of schizophrenia and bipolar disorder in a diverse cohort of US veterans. Schizophr. Bull. 47, 517–529 (2021). (PMID: 3316915510.1093/schbul/sbaa133)
Cross-Disorder Group of the Psychiatric Genomics Consortium Genomic relationships, novel loci, and pleiotropic mechanisms across eight psychiatric disorders. Cell 179, 1469–1482.e11 (2019). (PMID: 707703210.1016/j.cell.2019.11.020)
Minelli, A. et al. PCLO gene: its role in vulnerability to major depressive disorder. J. Affect. Disord. 139, 250–255 (2012). (PMID: 2238604910.1016/j.jad.2012.01.028)
Choi, K. H. et al. Gene expression and genetic variation data implicate PCLO in bipolar disorder. Biol. Psychiatry 69, 353–359 (2011). (PMID: 2118501110.1016/j.biopsych.2010.09.042)
Sullivan, P. F. et al. Genome-wide association for major depressive disorder: a possible role for the presynaptic protein piccolo. Mol. Psychiatry 14, 359–375 (2009). (PMID: 1906514410.1038/mp.2008.125)
Lu, H. et al. Evaluating marginal genetic correlation of associated loci for complex diseases and traits between European and East Asian populations. Hum. Genet. 140, 1285–1297 (2021). (PMID: 3409177010.1007/s00439-021-02299-8)
Guo, J. et al. Quantifying genetic heterogeneity between continental populations for human height and body mass index. Sci. Rep. 11, 5240 (2021). (PMID: 33664403793329110.1038/s41598-021-84739-z)
Martin, A. R. et al. Clinical use of current polygenic risk scores may exacerbate health disparities. Nat. Genet. 51, 584–591 (2019). (PMID: 30926966656383810.1038/s41588-019-0379-x)
Curtis, D. Polygenic risk score for schizophrenia is more strongly associated with ancestry than with schizophrenia. Psychiatr. Genet. 28, 85–89 (2018). (PMID: 3016065910.1097/YPG.0000000000000206)
Brown, B. C. et al. Transethnic genetic-correlation estimates from summary statistics. Am. J. Hum. Genet. 99, 76–88 (2016). (PMID: 27321947500543410.1016/j.ajhg.2016.05.001)
Bragg, L. M., Stone, G., Butler, M. K., Hugenholtz, P. & Tyson, G. W. Shining a light on dark sequencing: characterising errors in Ion Torrent PGM data. PLoS Comput. Biol. 9, e1003031 (2013). (PMID: 23592973362371910.1371/journal.pcbi.1003031)
Chen, T. & Guestrin, C. XGBoost: a scalable tree boosting system. Preprint at https://doi.org/10.48550/arXiv.1603.02754 (2016).
Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007). (PMID: 17701901195083810.1086/519795)
McLaren, W. et al. Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics 26, 2069–2070 (2010). (PMID: 20562413291672010.1093/bioinformatics/btq330)
Ma, C., Blackwell, T., Boehnke, M., Scott, L. J. & GoT2D investigators Recommended joint and meta-analysis strategies for case-control association testing of single low-count variants. Genet. Epidemiol. 37, 539–550 2013). (PMID: 23788246404932410.1002/gepi.21742)
Do, R., Kathiresan, S. & Abecasis, G. R. Exome sequencing and complex disease: practical aspects of rare variant association studies. Hum. Mol. Genet. 21, R1–R9 (2012). (PMID: 22983955345964110.1093/hmg/dds387)
معلومات مُعتمدة: MR/L010305/1 United Kingdom MRC_ Medical Research Council; R01 MH104964 United States MH NIMH NIH HHS; R01 MH113619 United States MH NIMH NIH HHS; R01 MH123451 United States MH NIMH NIH HHS
فهرسة مساهمة: Investigator: HS Aghanwa; M Ansari; A Asif; R Aslam; JL Ayuso; T Bigdeli; S Bignotti; J Bobes; B Bradley; P Buckley; MJ Cairns; SV Catts; AR Chaudhry; D Cohen; BL Collins; A Consoli; J Costas; B Crespo-Facorro; NP Daskalakis; M Davidson; KL Davis; F Dickerson; IA Dogar; E Drapeau; L Fañanás; A Fanous; W Fatima; M Fatjo; C Filippich; J Friedman; JF Fullard; P Georgakopoulos; M Giannitelli; I Giegling; MJ Green; O Guillin; B Gutierrez; HY Handoko; SK Hansen; M Haroon; V Haroutunian; FA Henskens; F Hussain; AV Jablensky; J Junejo; BJ Kelly; SA Khan; MNS Khan; A Khan; HR Khawaja; B Khizar; SP Kleopoulos; J Knowles; B Konte; AAAA Kusumawardhani; N Leghari; X Liu; A Lori; CM Loughland; K Mahmood; S Mahmood; D Malaspina; D Malik; A McNaughton; PT Michie; V Michopolous; E Molina; MD Molto; A Munir; G Muntané; F Naeem; DJ Nancarrow; A Nasar; T Nasr; JU Ohaeri; J Ott; C Pantelis; S Periyasamy; AG Pinto; A Powers; B Ramos; NH Rana; M Rapaport; A Reichenberg; S Saker-Delye; U Schall; PR Schofield; RJ Scott; M Shanahan; CS Weickert; C Sjaarda; HJ Smith; JJ Suárez-Rama; M Tariq; F Thibaut; PA Tooney; M Umar; E Vilella; M Weiser; JQ Wu; R Yolken
تواريخ الأحداث: Date Created: 20230314 Date Completed: 20230315 Latest Revision: 20240221
رمز التحديث: 20240221
مُعرف محوري في PubMed: PMC10011128
DOI: 10.1038/s41588-023-01305-1
PMID: 36914870
قاعدة البيانات: MEDLINE
الوصف
تدمد:1546-1718
DOI:10.1038/s41588-023-01305-1