Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Mapping genomic loci implicates genes and synaptic biology in schizophrenia

Abstract

Schizophrenia has a heritability of 60–80%1, much of which is attributable to common risk alleles. Here, in a two-stage genome-wide association study of up to 76,755 individuals with schizophrenia and 243,649 control individuals, we report common variant associations at 287 distinct genomic loci. Associations were concentrated in genes that are expressed in excitatory and inhibitory neurons of the central nervous system, but not in other tissues or cell types. Using fine-mapping and functional genomic data, we identify 120 genes (106 protein-coding) that are likely to underpin associations at some of these loci, including 16 genes with credible causal non-synonymous or untranslated region variation. We also implicate fundamental processes related to neuronal function, including synaptic organization, differentiation and transmission. Fine-mapped candidates were enriched for genes associated with rare disruptive coding variants in people with schizophrenia, including the glutamate receptor subunit GRIN2A and transcription factor SP4, and were also enriched for genes implicated by such variants in neurodevelopmental disorders. We identify biological processes relevant to schizophrenia pathophysiology; show convergence of common and rare variant associations in schizophrenia and neurodevelopmental disorders; and provide a resource of prioritized genes and variants to advance mechanistic studies.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of GWAS and gene prioritization.
Fig. 2: Gene set enrichment tests at the genome-wide level and for protein-coding genes that contain FINEMAP credible SNPs.
Fig. 3: Mapping of all fine-mapped and SMR-associated genes, and prioritized genes, to synaptic locations using SynGO.
Fig. 4: Associations between schizophrenia and cell types from multiple brain regions in human and mouse.

Similar content being viewed by others

Data availability

Summary statistics for the ‘extended’, ‘core’, ancestry-specific and sex-stratified analyses are available at https://www.med.unc.edu/pgc/download-results/scz/. Genotype data are available for a subset of cohorts, including dbGAP accession numbers and/or restrictions, as described in the ‘Case–control sample descriptions’ section of the Supplementary Information.

Code availability

Core analysis code for RICOPILI can be found at https://sites.google.com/a/broadinstitute.org/ricopili/. This wraps PLINK (https://www.cog-genomics.org/plink2/), EIGENSOFT (https://www.hsph.harvard.edu/alkes-price/software/), Eagle2 (https://alkesgroup.broadinstitute.org/Eagle/), Minimac3 (https://genome.sph.umich.edu/wiki/Minimac3), SHAPEIT3 (https://mathgen.stats.ox.ac.uk/genetics_software/shapeit/shapeit.html), METAL (https://genome.sph.umich.edu/wiki/METAL_Documentation) and LDSR (https://github.com/bulik/ldsc). For downstream analyses, FINEMAP can be found at http://christianbenner.com/, and our utility for meta-analysing cohort-specific LD matrices can be found at https://github.com/Pintaius/LDmergeFM. MAGMA can be found at https://ctg.cncr.nl/software/magma and the GO gene sets and automated curation pipeline are provided in https://github.com/janetcharwood/pgc3-scz_wg-genesets. SMR is available at https://cnsgenomics.com/software/smr/ and SbayesS at https://cnsgenomics.com/software/gctb/.

References

  1. Owen, M. J., Sawa, A. & Mortensen, P. B. Schizophrenia. Lancet 388, 86–97 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Plana-Ripoll, O. et al. A comprehensive analysis of mortality-related health metrics associated with mental disorders: a nationwide, register-based cohort study. Lancet 394, 1827–1835 (2019).

    Article  PubMed  Google Scholar 

  3. Momen, N. C. et al. Association between mental disorders and subsequent medical conditions. N. Engl. J. Med. 382, 1721–1731 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Jääskeläinen, E. et al. A systematic review and meta-analysis of recovery in schizophrenia. Schizophr. Bull. 39, 1296–1306 (2013).

    Article  PubMed  Google Scholar 

  5. International Schizophrenia Consortium. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature 460, 748–752 (2009).

  6. Pocklington, A. J. et al. Novel findings from CNVs implicate inhibitory and excitatory signaling complexes in schizophrenia. Neuron 86, 1203–1214 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Rees, E. et al. De novo mutations identified by exome sequencing implicate rare missense variants in SLC6A1 in schizophrenia. Nat. Neurosci 23, 179–184 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Lam, M. et al. Comparative genetic architectures of schizophrenia in East Asian and European populations. Nat. Genet. 51, 1670–1678 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. 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).

    Article  PubMed  CAS  Google Scholar 

  11. Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427 (2014).

  12. Räsänen, S., Pakaslahti, A., Syvälahti, E., Jones, P. B. & Isohanni, M. Sex differences in schizophrenia: a review. Nord. J. Psychiatry 54, 37–45 (2000).

    Article  Google Scholar 

  13. Zeng, J. et al. Widespread signatures of natural selection across human complex traits and functional genomic categories. Nat. Commun. 12, 1164 (2021).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  14. Aguet, F. et al. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

    Article  ADS  Google Scholar 

  15. Genome-wide association study identifies five new schizophrenia loci. Nat. Genet. 43, 969–978 (2011).

  16. Skene, N. G. et al. Genetic identification of brain cell types underlying schizophrenia. Nat. Genet. 50, 825–833 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Habib, N. et al. Massively parallel single-nucleus RNA-seq with DroNc-seq. Nat. Methods 14, 955–958 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Zeisel, A. et al. Molecular architecture of the mouse nervous system. Cell 174, 999–1014 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Koopmans, F. et al. SynGO: an evidence-based, expert-curated knowledge base for the synapse. Neuron 103, 217–234 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Benner, C. et al. FINEMAP: efficient variable selection using summary data from genome-wide association studies. Bioinformatics 32, 1493–1501 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Sakuntabhai, A. et al. Mutations in ATP2A2, encoding a Ca2+ pump, cause Darier disease. Nat. Genet. 21, 271–277 (1999).

    Article  PubMed  CAS  Google Scholar 

  22. Cederlöf, M. et al. The association between Darier disease, bipolar disorder, and schizophrenia revisited: a population-based family study. Bipolar Disord. 17, 340–344 (2015).

    Article  PubMed  CAS  Google Scholar 

  23. Pardiñas, A. F. et al. Common schizophrenia alleles are enriched in mutation-intolerant genes and in regions under strong background selection. Nat. Genet. 50, 381–389 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Zhu, Z. et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat. Genet. 48, 481–487 (2016).

    Article  PubMed  CAS  Google Scholar 

  25. Gandal, M. J. et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science 362, eaat8127 (2018).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  26. O’Brien, H. E. et al. Expression quantitative trait loci in the developing human brain and their enrichment in neuropsychiatric disorders. Genome Biol. 19, 194 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Võsa, U. et al. Large-scale cis- and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expression. Nat. Genet. 53, 1300–1310 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Wang, D. et al. Comprehensive functional genomic resource and integrative model for the human brain. Science 362, eaat8464 (2018).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  29. Galvan, L. et al. The striatal kinase DCLK3 produces neuroprotection against mutant huntingtin. Brain 141, 1434–1454 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Singh, T. et al. Rare coding variants in 10 genes confer substantial risk for schizophrenia. Nature https://doi.org/10.1038/s41586-022-04556-w (2022).

  31. Rees, E. et al. Analysis of intellectual disability copy number variants for association with schizophrenia. JAMA Psychiatry 73, 963–969 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Fromer, M. et al. De novo mutations in schizophrenia implicate synaptic networks. Nature 506, 179–184 (2014).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  33. Kaplanis, J. et al. Evidence for 28 genetic disorders discovered by combining healthcare and research data. Nature 586, 757–762 (2020).

  34. 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 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Luo, Y. et al. Exploring the genetic architecture of inflammatory bowel disease by whole-genome sequencing identifies association at ADCY7. Nat. Genet. 49, 186–192 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Cheng, Y. et al. Rare genetic variants affecting urine metabolite levels link population variation to inborn errors of metabolism. Nat. Commun. 12, 964 (2021).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  37. Singh, T., Neale, B. M. & Daly, M. J. Exome sequencing identifies rare coding variants in 10 genes which confer substantial risk for schizophrenia. Preprint at https://doi.org/10.1101/2020.09.18.20192815 (2020).

  38. Priya, A., Johar, K. & Wong-Riley, M. T. T. Specificity protein 4 functionally regulates the transcription of NMDA receptor subunits GluN1, GluN2A, and GluN2B. Biochim. Biophys. Acta 1833, 2745–2756 (2013).

    Article  PubMed  CAS  Google Scholar 

  39. Ripke, S. et al. Genome-wide association analysis identifies 13 new risk loci for schizophrenia. Nat. Genet. 45, 1150–1159 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Kirov, G. et al. De novo CNV analysis implicates specific abnormalities of postsynaptic signalling complexes in the pathogenesis of schizophrenia. Mol. Psychiatry 17, 142–153 (2012).

    Article  PubMed  CAS  Google Scholar 

  41. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Fagerberg, L. et al. Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol. Cell. Proteomics 13, 397–406 (2014).

    Article  PubMed  CAS  Google Scholar 

  43. Stephens, R. et al. Gene organisation, sequence variation and isochore structure at the centromeric boundary of the human MHC. J. Mol. Biol. 291, 789–799 (1999).

  44. Lam, M. et al. RICOPILI: Rapid Imputation for COnsortias PIpeLIne. Bioinformatics 36, 930–933 (2019).

    Article  PubMed Central  CAS  Google Scholar 

  45. Purcell, S., Neale, B., Todd-Brown, K., Thomas, L. & Ferreira, M. A. PLINK: a toolset for whole-genome association and population-based linkage analysis. Am. J. Hum. Genet. 81, 559–575 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Devlin, B. & Roeder, K. Genomic control for association studies. Biometrics 55, 997–1004 (1999).

    Article  PubMed  CAS  MATH  Google Scholar 

  47. Reference-based phasing using the Haplotype Reference Consortium panel. Nat. Genet. 48, 1443–1448 (2016).

  48. Das, S. et al. Next-generation genotype imputation service and methods. Nat. Genet. 48, 1284–1287 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. The Haplotype Reference Consortium. A reference panel of 64,976 haplotypes for genotype imputation. Nat. Genet. 48, 1279–1283 (2016).

  50. O’Connell, J. et al. Haplotype estimation for biobank-scale data sets. Nat. Genet. 48, 817–820 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Bycroft, C. et al. The UK Biobank resource with deep phenotyping and genomic data. Nature 562, 203–209 (2018).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  52. Chang, C. C. et al. Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience 4, 7 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Lee, J. J. et al. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat. Genet. 50, 1112–1121 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Vittinghoff, E. & McCulloch, C. E. Relaxing the rule of ten events per variable in logistic and cox regression. Am. J. Epidemiol. 165, 710–718 (2007).

    Article  PubMed  Google Scholar 

  55. Heinze, G. & Ploner, M. A SAS macro, S-PLUS library and R package to perform logistic regression without convergence problems. Technical report 2/2004 https://cemsiis.meduniwien.ac.at/fileadmin/user_upload/_imported/fileadmin/msi_akim/CeMSIIS/KB/programme/tr2_2004.pdf (Medical University of Vienna, 2004).

  56. Viechtbauer, W. Conducting meta-analyses in R with the metafor package. J. Stat. Softw. 36, 1–48 (2010).

    Article  Google Scholar 

  57. Lee, S. H., Goddard, M. E., Wray, N. R. & Visscher, P. M. A better coefficient of determination for genetic profile analysis. Genet. Epidemiol. 36, 214–224 (2012).

    Article  PubMed  Google Scholar 

  58. Martínez-Camblor, P. Fully non-parametric receiver operating characteristic curve estimation for random-effects meta-analysis. Stat. Methods Med. Res. 26, 5–20 (2017).

    Article  PubMed  MathSciNet  Google Scholar 

  59. Bryois, J. et al. Genetic identification of cell types underlying brain complex traits yields insights into the etiology of Parkinson’s disease. Nat. Genet. 52, 482–493 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. de Leeuw, C. A., Mooij, J. M., Heskes, T. & Posthuma, D. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput. Biol. 11, e1004219 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Finucane, H. K. et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621–629 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Durinck, S., Spellman, P. T., Birney, E. & Huber, W. Mapping identifiers for the integration of genomic datasets with the R/ Bioconductor package biomaRt. Nat. Protoc. 4, 1184–1191 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Maston, G. A., Evans, S. K. & Green, M. R. Transcriptional regulatory elements in the human genome. Annu. Rev. Genomics Hum. Genet. 7, 29–59 (2006).

    Article  PubMed  CAS  Google Scholar 

  64. A reference panel of 64,976 haplotypes for genotype imputation. Nat. Genet. 48, 1279–1283 (2016).

  65. Genovese, G. et al. Increased burden of ultra-rare protein-altering variants among 4,877 individuals with schizophrenia. Nat. Neurosci. 19, 1433–1441 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Merico, D., Isserlin, R., Stueker, O., Emili, A. & Bader, G. D. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS One 5, e13984 (2010).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  67. Benner, C. et al. Prospects of fine-mapping trait-associated genomic regions by using summary statistics from genome-wide association studies. Am. J. Hum. Genet 101, 539–551 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Võsa, U. et al. Large-scale cis- and trans-eQTL analysis identify thousands of genetic loci and polygenic scores that regulate blood gene expression. Nat. Genet. 53, 1300–1310 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Sonnega, A. et al. Cohort profile: The Health and Retirement Study (HRS). Int. J. Epidemiol. 43, 576–585 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Yang, J., Lee, S. H., Goddard, M. E. & Visscher, P. M. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 88, 76–82 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Yang, J. et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat. Genet. 44, 369–375 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Gusev, A. et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 48, 245–252 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Zhang, W. et al. Integrative transcriptome imputation reveals tissue-specific and shared biological mechanisms mediating susceptibility to complex traits. Nat. Commun. 10, 3834 (2019).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

Download references

Acknowledgements

The National Institute of Mental Health (USA) provides core funding for the PGC under award no. U01MH109514. The content is the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The work of the contributing groups was supported by numerous grants from governmental and charitable bodies as well as philanthropic donation (details in Supplementary Note). We acknowledge a substantial contribution from P. Sklar (deceased) as one of the PGC principal investigators, and E. Scolnick, whose support for this study was vital. We acknowledge the Wellcome Trust Case Control Consortium for the provision of control genotype information. Membership of the Psychosis Endophenotypes International Consortium, the SynGO consortium, the PsychENCODE Consortium, the eQTLGen consortium, the BIOS Consortium and the Indonesia Consortium are provided in the author list. We are grateful to C. Hopkins for illustrations. The work at Cardiff University was additionally supported by Medical Research Council Centre grant no. MR/L010305/1 and program grant no. G0800509. S. Xu also gratefully acknowledges the support of the National Natural Science Foundation of China (NSFC) grants (31525014, 91731303, 31771388, 31961130380 and 32041008), the UK Royal Society-Newton Advanced Fellowship (NAF\R1\191094), the Key Research Program of Frontier Sciences (QYZDJ-SSW-SYS009) and the Strategic Priority Research Program (XDB38000000) of the Chinese Academy of Sciences, and the Shanghai Municipal Science and Technology Major Project (2017SHZDZX01). O. A. Andreassen was supported by the Research Council of Norway (283798, 262656, 248980, 273291, 248828, 248778, 223273); KG Jebsen Stiftelsen, South-East Norway Health Authority, EU H2020 no. 847776. B. Melegh was supported in part by the National Scientific Research Program (NKFIH) K 138669. S. V. Faraone is supported by the European Union’s Seventh Framework Programme for research, technological development and demonstration under grant agreement no. 602805, the European Union’s Horizon 2020 research and innovation programme under grant agreements 667302 and 728018 and NIMH grants 5R01MH101519 and U01 MH109536-01. S. I. Belangero was supported by FAPESP (Fundação de Amparo à Pesquisa do Estado de São Paulo), grant numbers: 2010/08968-6; 2014/07280-1 2011/50740-5 (including R. A. Bressan). The Singapore team (J. Lee, J. Liu, K. Sim, S. A. Chong and M. Subramanian) acknowledges the National Medical Research Council Translational and Clinical Research Flagship Programme (grant no.: NMRC/TCR/003/2008). M. Macek was supported by LM2018132, CZ.02.1.01/0.0/0.0/18_046/0015515 and IP6003 –VZFNM00064203. C. Arango has been funded by the Spanish Ministry of Science and Innovation, Instituto de Salud Carlos III (SAM16PE07CP1, PI16/02012, PI19/024), co-financed by ERDF Funds from the European Commission, ‘A way of making Europe’, CIBERSAM, Madrid Regional Government (B2017/BMD-3740 AGES-CM-2), European Union Structural Funds, European Union Seventh Framework Program and European Union H2020 Program under the Innovative Medicines Initiative 2 Joint Undertaking (grant agreement no 115916, project PRISM; and grant agreement no. 777394, project AIMS-2-TRIALS), Fundación Familia Alonso and Fundación Alicia Koplowitz. E. Bramon acknowledges support from the National Institute of Health Research UK (grant NIHR200756); Mental Health Research UK John Grace QC Scholarship 2018; an ESRC collaborative award 2020; BMA Margaret Temple Fellowship 2016; Medical Research Council New Investigator Award (G0901310); MRC Centenary Award (G1100583); MRC project grant G1100583; National Institute of Health Research UK post-doctoral fellowship (PDA/02/06/016); NARSAD Young Investigator awards 2005 and 2008; Wellcome Trust Research Training Fellowship; Wellcome Trust Case Control Consortium awards (085475/B/08/Z, 085475/Z/08/Z); European Commission Horizon 2020 (747429); NIHR Biomedical Research Centre for Mental Health at the South London and Maudsley NHS Foundation Trust and King’s College London; and NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust and University College London (UCLH BRC - Mental Health Theme). D. Molto is funded by the European Regional Development Fund (ERDF)–Valencian Community 2014–2020, Spain. E. G. Atkinson was supported by the NIMH K01MH121659.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

The management group for this paper was led by M.O.D. and J.T.R.W., with S.R. responsible for primary analytic matters supported by B.M.N. and M.J.D. The management group comprised a subset of the principal investigators of the component studies, bioinformaticians and analysts and was responsible for study design, conduct, management, primary and final interpretation; this group included O.A.A., B.T.B., S.I.B., A.D.B., D.B., E.B., S.C., A. Corvin, D. Curtis, M.J.D., M.D.F., E.D., H.E., A.H.F., P.V.G., M. Gill, S.J.G., K.S.H., H. Huang, N.I., R.S.K., K.S.K., J.A.K., J. Lee, T.L., D.F.L., J. Liu, A. McIntosh, A. McQuillin, V.A.M., D.W.M., B.J.M., B.M.N., M.O.D., R.A.O., M.J.O., A.P., D. Posthuma, S.Q., B.P.R., S.R., D.R., S.G.S., A. Serretti, Y.S., E.A.S., P.F.S., M.T.T., M.P.V., J.T.R.W., D.R.W., T.W., N.R.W., X.Y. and W.Y. GWAS meta-analyses: S.A., G.P., S.R. and V.T. Replication data: S. Magnusson, H.S. and K. Stefansson (deCODE). African American and Latino sample analyses: E.G.A., T.B., G.G., S.R. and V.T. Bioinformatics: J. Bryois, J.C.H., A.F.P., A.J.P., D. Posthuma, P.F.S., K.W. and the SynGO consortium. Comparison of male and female individuals: S.R., J. Sidorenko, V.T. and P.M.V. Heritability and polygenic prediction: O.A.A., O.F., T.G., H. Huang, B.M.N., M.O.D., A.F.P., A.L.R., S.R., V.T., J.T.R.W., N.R.W. and J.Z. Phenotype stratification: C.A.D. and E. Vassos. Cellular and tissue analysis: J. Bryois, M.O.D., D. Posthuma, P.F.S., J.T.R.W. and K.W. Gene Ontology: J.C.H., M.O.D., A.F.P., A.J.P., D. Posthuma, J.T.R.W. and K.W. Fine-mapping: C.B., M.J.D., H. Huang, M. Lam, M.O.D., G.P., A.F.P., M.P., S.R. and J.T.R.W. SMR: L.S.H., M.O.D., T.Q., N.R.W., Y.W. and J.Y. Hi-C: D. Posthuma, A.L.R., P.F.S., J.T.R.W. and K.W. Other transcriptome-wide association studies: M.J.G., L.S.H., M. Kim, P.R., G.V. and W. Zhang. Integration of fine-mapping, gene expression, Hi-C informatics and rare variants: L.S.H., M.O.D., A.F.P., T.Q., A.L.R., P.F.S., J.T.R.W., N.R.W., Y.W. and J.Y. SynGO: F.K., M.O.D., A.F.P., A.B.S., M.V. and J.T.R.W. Additional statistical advice: P.A.H. The remaining authors contributed to the recruitment, phenotyping, genotyping or data processing for the contributing components of the meta-analysis, or provided other forms of functional annotation data. Primary drafting and editing of the manuscript were coordinated by S.R., J.T.R.W. and M.O.D. The primary draft sections were written by J. Bryois, C.Y.C., C.A.D., L.S.H., H. Huang, B.M.N., M.O.D., M.J.O., A.F.P., A.J.P., S.R., A.B.S, P.F.S., V.T., E. Vassos, M.V., J.T.R.W., N.R.W. and J.Y. Additional edits were from O.A.A., M.J.D. and K.S.K. Numerous other authors provided edits, comments and suggestions, and all authors saw and approved the contents of the manuscript. The Chair of the PGC is P.F.S. and the Schizophrenia Working Group of the PGC is led by M.O.D. and J.T.R.W.

Corresponding authors

Correspondence to Stephan Ripke, James T. R. Walters or Michael C. O’Donovan.

Ethics declarations

Competing interests

A. Palotie is a member of Astra Zeneca’s Genomics Advisory Board. V. Salomaa has consulted for Novo Nordisk and Sanofi and has ongoing research collaboration with Bayer (both unrelated to the present study). M. F. Green is a paid consultant for AiCure, Biogen, Lundbeck and Roche, is a member of the Scientific Board of Cadent, and has received research funds from Forum. G. A. Light has consulted to Astellas, Forum, and Neuroverse. K. Nuechterlein has research support from Janssen, Genentech and Brain Plasticity, and has also consulted for Astellas, MedinCell, Takeda, Teva, Genentech, Otsuka, Janssen and Brain Plasticity. D. Cohen has reported past consultation for or the receipt of honoraria from Otsuka, Shire, Lundbeck, Roche and Janssen. M. J. Daly is a founder of Maze Therapeutics and on the scientific advisory board of Neumora Therapeutics. A. K. Malhotra is a consultant to Genomind, InformedDNA and Concert Pharmaceuticals. R. A. Bressan has received research grants from Janssen, has been a forum consultant for Janssen, Sanof and Roche and is on the speakers’ bureau for Ache, Janssen, Sanofi and Torrent. C. Noto was on the speakers’ bureau and/or has acted as a consultant for Janssen and Daiichi-Sankyo in the last 12 months. C. Pantelis has, for the last three years, served on an advisory board for Lundbeck and received honoraria for talks presented at educational meetings organized by Lundbeck. D. A. Collier is a full-time employee and stockholder of Eli Lilly and Company. M. C. O’Donovan is supported by a collaborative research grant from Takeda Pharmaceuticals. M. J. Owen is supported by a collaborative research grant from Takeda Pharmaceuticals. J. T. R. Walters is supported by a collaborative research grant from Takeda Pharmaceuticals. A. J. Pocklington is supported by a collaborative research grant from Takeda Pharmaceuticals. S. R. Marder has consulted for the following companies: Roche, Sunovion, Lundbeck, Boeringer-Ingelheim, Acadia and Merck. S. Gopal is a full time employee and shareholder in Johnson & Johnson (AMEX: JNJ). A. Savitz is an employee of Janssen Research & Development and owns stock or stock options in the company. Q. S. Li is an employee of Janssen Research & Development and owns stock or stock options in the company. T. Kam-Thong is an employee of F. Hoffman-La Roche. A. Rautanen is an employee of F. Hoffman-La Roche. D. Malhotra is an employee of F. Hoffman-La Roche. S. A. Paciga is an employee of Pfizer. O. A. Andreassen is a consultant for HealthLytix, and received speaker’s honorarium from Lundbeck. S. V. Faraone has received income, potential income, travel expenses continuing education support and/or research support from Akili Interactive Labs, Arbor, Genomind, Ironshore, Ondosis, Otsuka, Rhodes, Shire/Takeda, Sunovion, Supernus, Tris and Vallon. With his institution, he has US patent US20130217707 A1 for the use of sodium-hydrogen exchange inhibitors in the treatment of attention deficit hyperactivity disorder. In previous years, he received support from Alcobra, Aveksham, CogCubed, Eli Lilly, Enzymotec, Impact, Janssen, KemPharm, Lundbeck/Takeda, McNeil, Neurolifesciences, Neurovance, Novartis, Pfizer and Vaya. He also receives royalties from books published by Guilford Press: Straight Talk about Your Child’s Mental Health; Oxford University Press: Schizophrenia: The Facts; and Elsevier: ADHD: Non-Pharmacologic Interventions. He is also Program Director of https://adhdinadults.com/. C. Arango has been a consultant to or has received honoraria or grants from Acadia, Angelini, Gedeon Richter, Janssen Cilag, Lundbeck, Minerva, Otsuka, Roche, Sage, Servier, Shire, Schering Plough, Sumitomo Dainippon Pharma, Sunovion and Takeda. K. Alptekin has received grants and honoraria for consulting work, lecturing and research from Abdi İbrahim, Abdi İbrahim Otsuka, Janssen, Ali Raif and TUBITAK.

Peer review

Peer review information

Nature thanks Paul O’Reilly and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Primary GWAS Manhattan plot.

The x axis indicates chromosomal position and the y axis is the significance of association (−log10(P)). The red line represents genome-wide significance level (5 × 10−8). SNPs in green are in linkage disequilibrium (LD; r2 > 0.1) with index SNPs (diamonds) which represent LD-independent genome-wide significant associations.

Extended Data Fig. 2 Polygenic risk prediction.

a, Distributions of liability scale R2 across 98 left-out-cohorts for polygenic risk scores built from SNPs with different p-value thresholds. Distributions of liability R2 (assuming schizophrenia life-time risk of 1%) are shown for each p-value threshold, with point size representing size of the left-out cohort and colour representing ancestry. The median liability R2 is represented as a horizontal black line. b, Liability R2 of predicted and observed phenotypes in left-out cohorts using variants with p-value threshold p = 0.05, from the fixed effect meta-analysis of variant effects, unadjusted for multiple comparisons. The polygenic risk scores are derived from two separate sets of leave-one-out GWAS meta-analyses: y axis R2 based on the results of primary GWAS including all ancestries; x axis R2 based on cohorts of the same ancestry as the test samples. Circles denote core PGC samples. Triangles denote African American and Latino samples processed external to PGC by the providing author.

Extended Data Fig. 3 Association between 37 human tissues and schizophrenia.

The mean of the evidence (-log10P) obtained from two methods (MAGMA, LDSC) for testing GWAS data for enrichment of association in genes with high expression in each tissue as determined from bulk RNA-seq14. The bar colour indicates whether gene expression in the tissue is significantly associated with both methods, one method or none. The black vertical line represents the significance threshold corrected for the total number of tissues tested in this experiment. We also analysed previous waves of PGC schizophrenia GWAS11,15 for comparison.

Extended Data Table 1 List of prioritized genes

Supplementary information

Supplementary Information

The main supplementary information document, containing the Supplementary Note; all supplementary figure legends; supplementary figures 3–5, 8, 9, 11, 12; and cohort descriptions.

Reporting Summary

Peer Review File

Supplementary Figure 1a

Supplementary Figure 1b

Supplementary Figure 2a

Supplementary Figure 2b

Supplementary Figure 6

Supplementary Figure 7

Supplementary Figure 10

Supplementary Tables

This zip folder contains excel workbooks for all Supplementary Tables (1–29).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Trubetskoy, V., Pardiñas, A.F., Qi, T. et al. Mapping genomic loci implicates genes and synaptic biology in schizophrenia. Nature 604, 502–508 (2022). https://doi.org/10.1038/s41586-022-04434-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04434-5

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing