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:

Multi-ancestry genome-wide association analyses identify novel genetic mechanisms in rheumatoid arthritis

Abstract

Rheumatoid arthritis (RA) is a highly heritable complex disease with unknown etiology. Multi-ancestry genetic research of RA promises to improve power to detect genetic signals, fine-mapping resolution and performances of polygenic risk scores (PRS). Here, we present a large-scale genome-wide association study (GWAS) of RA, which includes 276,020 samples from five ancestral groups. We conducted a multi-ancestry meta-analysis and identified 124 loci (P < 5 × 10−8), of which 34 are novel. Candidate genes at the novel loci suggest essential roles of the immune system (for example, TNIP2 and TNFRSF11A) and joint tissues (for example, WISP1) in RA etiology. Multi-ancestry fine-mapping identified putatively causal variants with biological insights (for example, LEF1). Moreover, PRS based on multi-ancestry GWAS outperformed PRS based on single-ancestry GWAS and had comparable performance between populations of European and East Asian ancestries. Our study provides several insights into the etiology of RA and improves the genetic predictability of RA.

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

Access options

Buy this article

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

Fig. 1: Diverse ancestral backgrounds of GWAS participants.
Fig. 2: Fine-mapping analysis identified candidate causal variants.
Fig. 3: Splicing and total expression of IL6R jointly contribute to RA risk.
Fig. 4: Splicing of PADI4 contributes to RA risk.
Fig. 5: S-LDSC analysis suggested similar causal variant distributions in EUR-GWAS and EAS-GWAS.
Fig. 6: Fine-mapped variants with high IMPACT scores.
Fig. 7: Functional annotation and multi-ancestry GWAS improved PRS performances.

Similar content being viewed by others

Data availability

We deposited six sets of summary statistics (multi-ancestry, EUR-GWAS and EAS-GWAS for all RA and seropositive RA) to the GWAS Catalog under accession IDs GCST90132222, GCST90132223, GCST90132224, GCST90132225, GCST90132226 and GCST90132227. We deposited the PRS model (multi-ancestry PRS with CD4+ T cell T-bet IMPACT annotation) to the Polygenic Score (PGS) Catalog; the publication ID is PGP000357, and the score ID is PGS002745. Summary statistics and the PRS model are also available at https://data.cyverse.org/dav-anon/iplant/home/kazuyoshiishigaki/ra_gwas/ra_gwas-10-28-2021.tar. The UK Biobank analysis was conducted via application no. 47821.

Code availability

The codes are available at our website (https://github.com/immunogenomics/RA_GWAS) and archived in Zenodo (https://doi.org/10.5281/zenodo.6999289).

References

  1. Ajeganova, S. & Huizinga, T. W. J. Seronegative and seropositive RA: alike but different? Nat. Rev. Rheumatol. 11, 8–9 (2015).

    Article  PubMed  Google Scholar 

  2. Okada, Y. et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506, 376–381 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Ishigaki, K. et al. Large-scale genome-wide association study in a Japanese population identifies novel susceptibility loci across different diseases. Nat. Genet. 52, 669–679 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. MacGregor, A. J. et al. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum. 43, 30–37 (2000).

    Article  CAS  PubMed  Google Scholar 

  5. Ishigaki, K. et al. Polygenic burdens on cell-specific pathways underlie the risk of rheumatoid arthritis. Nat. Genet. 49, 1120–1125 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Westra, H.-J. et al. Fine-mapping and functional studies highlight potential causal variants for rheumatoid arthritis and type 1 diabetes. Nat. Genet. 50, 1366–1374 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Trynka, G. et al. Chromatin marks identify critical cell types for fine mapping complex trait variants. Nat. Genet. 45, 124–130 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Asgari, S. et al. A positively selected FBN1 missense variant reduces height in Peruvian individuals. Nature 582, 234–239 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. SIGMA Type 2 Diabetes Consortium. Association of a low-frequency variant in HNF1A with type 2 diabetes in a Latino population. JAMA 311, 2305–2314 (2014).

    Article  Google Scholar 

  10. Moltke, I. et al. A common Greenlandic TBC1D4 variant confers muscle insulin resistance and type 2 diabetes. Nature 512, 190–193 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Koyama, S. et al. Population-specific and trans-ancestry genome-wide analyses identify distinct and shared genetic risk loci for coronary artery disease. Nat. Genet. 52, 1169–1177 (2020).

    Article  CAS  PubMed  Google Scholar 

  12. Chen, M.-H. et al. Trans-ethnic and ancestry-specific blood-cell genetics in 746,667 individuals from 5 global populations. Cell 182, 1198–1213.e14 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Huang, H. et al. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature 547, 173–178 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Laufer, V. A. et al. Genetic influences on susceptibility to rheumatoid arthritis in African-Americans. Hum. Mol. Genet. 28, 858–874 (2019).

    Article  CAS  PubMed  Google Scholar 

  15. Martin, A. R. et al. Clinical use of current polygenic risk scores may exacerbate health disparities. Nat. Genet. 51, 584–591 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ruan, Y. et al. Improving polygenic prediction in ancestrally diverse populations. Nat. Genet. 54, 573–580 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Márquez-Luna, C. et al. Multiethnic polygenic risk scores improve risk prediction in diverse populations. Genet. Epidemiol. 41, 811–823 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Leng, R.-X. et al. Identification of new susceptibility loci associated with rheumatoid arthritis. Ann. Rheum. Dis. 79, 1565–1571 (2020).

    Article  CAS  PubMed  Google Scholar 

  19. Kochi, Y. et al. A functional variant in FCRL3, encoding Fc receptor-like 3, is associated with rheumatoid arthritis and several autoimmunities. Nat. Genet. 37, 478–485 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Suzuki, A. et al. Functional haplotypes of PADI4, encoding citrullinating enzyme peptidylarginine deiminase 4, are associated with rheumatoid arthritis. Nat. Genet. 34, 395–402 (2003).

    Article  CAS  PubMed  Google Scholar 

  21. Okada, Y. et al. Meta-analysis identifies nine new loci associated with rheumatoid arthritis in the Japanese population. Nat. Genet. 44, 511–516 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Diogo, D. et al. TYK2 protein-coding variants protect against rheumatoid arthritis and autoimmunity, with no evidence of major pleiotropic effects on non-autoimmune complex traits. PLoS ONE 10, e0122271 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Traylor, M. et al. Genetic associations with radiological damage in rheumatoid arthritis: meta-analysis of seven genome-wide association studies of 2,775 cases. PLoS ONE 14, e0223246 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Márquez, A. et al. Meta-analysis of Immunochip data of four autoimmune diseases reveals novel single-disease and cross-phenotype associations. Genome Med. 10, 97 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Wei, W.-H., Viatte, S., Merriman, T. R., Barton, A. & Worthington, J. Genotypic variability based association identifies novel non-additive loci DHCR7 and IRF4 in sero-negative rheumatoid arthritis. Sci. Rep. 7, 5261 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Márquez, A. et al. A combined large-scale meta-analysis identifies COG6 as a novel shared risk locus for rheumatoid arthritis and systemic lupus erythematosus. Ann. Rheum. Dis. 76, 286–294 (2017).

    Article  PubMed  Google Scholar 

  27. Bossini-Castillo, L. et al. A genome-wide association study of rheumatoid arthritis without antibodies against citrullinated peptides. Ann. Rheum. Dis. 74, e15 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Eyre, S. et al. High-density genetic mapping identifies new susceptibility loci for rheumatoid arthritis. Nat. Genet. 44, 1336–1340 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Acosta-Herrera, M. et al. Genome-wide meta-analysis reveals shared new loci in systemic seropositive rheumatic diseases. Ann. Rheum. Dis. 78, 311–319 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Frisell, T. et al. Familial risks and heritability of rheumatoid arthritis: role of rheumatoid factor/anti-citrullinated protein antibody status, number and type of affected relatives, sex, and age. Arthritis Rheum. 65, 2773–2782 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Padyukov, L. et al. A genome-wide association study suggests contrasting associations in ACPA-positive versus ACPA-negative rheumatoid arthritis. Ann. Rheum. Dis. 70, 259–265 (2011).

    Article  PubMed  Google Scholar 

  32. Finucane, H. K. et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet. 47, 1228–1235 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wellcome Trust Case Control Consortium. Bayesian refinement of association signals for 14 loci in 3 common diseases. Nat. Genet. 44, 1294–1301 (2012).

    Article  Google Scholar 

  34. Kichaev, G. & Pasaniuc, B. Leveraging functional-annotation data in trans-ethnic fine-mapping studies. Am. J. Hum. Genet. 97, 260–271 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ulirsch, J. C. et al. Interrogation of human hematopoiesis at single-cell and single-variant resolution. Nat. Genet. 51, 683–693 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Fu, W. et al. A multiply redundant genetic switch ‘locks in’ the transcriptional signature of regulatory T cells. Nat. Immunol. 13, 972–980 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Chen, L. et al. Genetic drivers of epigenetic and transcriptional variation in human immune cells. Cell 167, 1398–1414.e24 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Giambartolomei, C. et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 10, e1004383 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Ferreira, R. C. et al. Functional IL6R 358Ala allele impairs classical IL-6 receptor signaling and influences risk of diverse inflammatory diseases. PLoS Genet. 9, e1003444 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Okada, Y. et al. Significant impact of miRNA–target gene networks on genetics of human complex traits. Sci. Rep. 6, 22223 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Schellekens, G. A., de Jong, B. A. W., van den Hoogen, F. H. J., van de Putte, L. B. A. & van Venrooij, W. J. Citrulline is an essential constituent of antigenic determinants recognized by rheumatoid arthritis–specific autoantibodies. J. Clin. Invest. 101, 273–281 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Suzuki, A. et al. Decreased severity of experimental autoimmune arthritis in peptidylarginine deiminase type 4 knockout mice. BMC Musculoskelet. Disord. 17, 205 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Seri, Y. et al. Peptidylarginine deiminase type 4 deficiency reduced arthritis severity in a glucose-6-phosphate isomerase-induced arthritis model. Sci. Rep. 5, 13041 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Arita, K. et al. Structural basis for Ca2+-induced activation of human PAD4. Nat. Struct. Mol. Biol. 11, 777–783 (2004).

    Article  CAS  PubMed  Google Scholar 

  45. Nanda, S. K. et al. ABIN2 function is required to suppress DSS-induced colitis by a Tpl2-independent mechanism. J. Immunol. 201, 3373–3382 (2018).

    Article  CAS  PubMed  Google Scholar 

  46. Matmati, M. et al. A20 (TNFAIP3) deficiency in myeloid cells triggers erosive polyarthritis resembling rheumatoid arthritis. Nat. Genet. 43, 908–912 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. GTEx Consortium. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

    Article  PubMed Central  Google Scholar 

  48. 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  CAS  PubMed  Google Scholar 

  49. James, E. A. et al. Citrulline-specific Th1 cells are increased in rheumatoid arthritis and their frequency is influenced by disease duration and therapy. Arthritis Rheumatol. 66, 1712–1722 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Takayanagi, H. et al. RANKL maintains bone homeostasis through c-Fos-dependent induction of interferon-β. Nature 416, 744–749 (2002).

    Article  CAS  PubMed  Google Scholar 

  51. Takeuchi, T. et al. Effects of the anti-RANKL antibody denosumab on joint structural damage in patients with rheumatoid arthritis treated with conventional synthetic disease-modifying antirheumatic drugs (DESIRABLE study): a randomised, double-blind, placebo-controlled phase. Ann. Rheum. Dis. 78, 899–907 (2019).

    Article  CAS  PubMed  Google Scholar 

  52. Nakatsuka, K., Nishizawa, Y. & Ralston, S. H. Phenotypic characterization of early onset Paget’s disease of bone caused by a 27-bp duplication in the TNFRSF11A gene. J. Bone Miner. Res. 18, 1381–1385 (2003).

    Article  CAS  PubMed  Google Scholar 

  53. Guerrini, M. M. et al. Human osteoclast-poor osteopetrosis with hypogammaglobulinemia due to TNFRSF11A (RANK) mutations. Am. J. Hum. Genet. 83, 64–76 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. French, D. M. et al. WISP-1 is an osteoblastic regulator expressed during skeletal development and fracture repair. Am. J. Pathol. 165, 855–867 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Maeda, A. et al. WNT1-induced secreted protein-1 (WISP1), a novel regulator of bone turnover and Wnt signaling. J. Biol. Chem. 290, 14004–14018 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhang, F. et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat. Immunol. 20, 928–942 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Ramos, M. I. P. et al. Absence of Fms-like tyrosine kinase 3 ligand (Flt3L) signalling protects against collagen-induced arthritis. Ann. Rheum. Dis. 74, 211–219 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Saevarsdottir, S. et al. FLT3 stop mutation increases FLT3 ligand level and risk of autoimmune thyroid disease. Nature 584, 619–623 (2020).

    Article  CAS  PubMed  Google Scholar 

  59. Motegi, T. et al. Identification of rare coding variants in TYK2 protective for rheumatoid arthritis in the Japanese population and their effects on cytokine signalling. Ann. Rheum. Dis. 78, 1062–1069 (2019).

    Article  CAS  PubMed  Google Scholar 

  60. Brown, B. C., Asian Genetic Epidemiology Network Type 2 Diabetes Consortium, Ye, C. J., Price, A. L. & Zaitlen, N. Transethnic genetic-correlation estimates from summary statistics. Am. J. Hum. Genet. 99, 76–88 (2016).

  61. Amariuta, T. et al. Improving the trans-ancestry portability of polygenic risk scores by prioritizing variants in predicted cell-type-specific regulatory elements. Nat. Genet. 52, 1346–1354 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Amariuta, T. et al. IMPACT: genomic annotation of cell-state-specific regulatory elements inferred from the epigenome of bound transcription factors. Am. J. Hum. Genet. 104, 879–895 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Roadmap Epigenomics Consortium. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    Article  PubMed Central  Google Scholar 

  64. 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  CAS  PubMed  PubMed Central  Google Scholar 

  65. Chun, S. et al. Limited statistical evidence for shared genetic effects of eQTLs and autoimmune-disease-associated loci in three major immune-cell types. Nat. Genet. 49, 600–605 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. van der Wijst, M. G. P. et al. Single-cell RNA sequencing identifies celltype-specific cis-eQTLs and co-expression QTLs. Nat. Genet. 50, 493–497 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Kumasaka, N., Knights, A. J. & Gaffney, D. J. High-resolution genetic mapping of putative causal interactions between regions of open chromatin. Nat. Genet. 51, 128–137 (2019).

    Article  CAS  PubMed  Google Scholar 

  68. Gutierrez-Arcelus, M. et al. Allele-specific expression changes dynamically during T cell activation in HLA and other autoimmune loci. Nat. Genet. 52, 247–253 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Baglaenko, Y., Macfarlane, D., Marson, A., Nigrovic, P. A. & Raychaudhuri, S. Genome editing to define the function of risk loci and variants in rheumatic disease. Nat. Rev. Rheumatol. 17, 462–474 (2021).

    Article  CAS  PubMed  Google Scholar 

  70. Arnett, F. C. et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 31, 315–324 (1988).

    Article  CAS  PubMed  Google Scholar 

  71. Aletaha, D. et al. 2010 Rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 62, 2569–2581 (2010).

    Article  PubMed  Google Scholar 

  72. Akiyama, M. et al. Characterizing rare and low-frequency height-associated variants in the Japanese population. Nat. Commun. 10, 4393 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Zhang, W. et al. Whole genome sequencing of 35 individuals provides insights into the genetic architecture of Korean population. BMC Bioinf. 15, S6 (2014).

    Article  Google Scholar 

  74. Lan, T. et al. Deep whole-genome sequencing of 90 Han Chinese genomes. GigaScience 6, gix067 (2017).

    Article  Google Scholar 

  75. Wong, L.-P. et al. Deep whole-genome sequencing of 100 southeast Asian Malays. Am. J. Hum. Genet. 92, 52–66 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Wakefield, J. A Bayesian measure of the probability of false discovery in genetic epidemiology studies. Am. J. Hum. Genet. 81, 208–227 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Rumble, J. M., Fackelman, E. M. & Mobley, J. L. Comparative analyses of PAD expression and activity in myeloid cell lines. J. Immunol. 198, 75.18 (2017).

    Google Scholar 

  78. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Director of Health Malaysia for supporting the work described in the South Asian (SAS) population: the Malaysian Epidemiological Investigation of Rheumatoid Arthritis (MyEIRA) study. The MyEIRA study was funded by grants from Ministry of Health Malaysia (NMRR-08-820-1975) and the Swedish National Research Council (DNR-348-2009-6468). The GENRA study and the CARDERA genetics cohort genotyping were funded by Versus Arthritis (grant reference 19739 to I.C.S.). The Nurses’ Health Study (NHS cohort) is funded by the National Institutes of Health (NIH) (R01 AR049880, UM1 CA186107, R01 CA49449, U01 CA176726 and R01 CA67262). The Swedish EIRA study was supported by the Swedish Research Council (to L.K., L.P. and L.A.). S.S. was in part supported by the Mochida Memorial Foundation for Medical and Pharmaceutical Research, Kanae Foundation for the Promotion of Medical Science, Astellas Foundation for Research on Metabolic Disorders, JCR Grant for Promoting Basic Rheumatology, and Manabe Scholarship Grant for Allergic and Rheumatic Diseases. I.C.S. is funded by the National Institute for Health and Care Research (NIHR) Advanced Research Fellowship (grant reference NIHR300826). The views expressed are those of the authors and not necessarily those of the NIHR or the Department of Health and Social Care. K.A.S. is supported by the Sherman Family Chair in Genomic Medicine and by a Canadian Institutes for Health Research Foundation Grant (FDN 148457) and grants from the Ontario Research Fund (RE-09-090) and Canadian Foundation for Innovation (33374). S.-C.B. is supported by the Basic Science Research Program through the NRF funded by the Ministry of Education (NRF-2021R1A6A1A03038899). R.P.K. and J.C.E. are funded by NIH (UL1 TR003096). C.M.L. is partly funded by the NIHR Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London. T. Arayssi was partially supported by the National Priorities Research Program (grant 4-344-3-105 from the Qatar National Research Fund, a member of Qatar Foundation). M. Kerick and J.M. are funded by Rheumatology Cooperative Research Thematic Network program RD16/0012/0013 from the Instituto de Salud Carlos III (Spanish Ministry of Science and Innovation). Y.O. is funded by JSPS KAKENHI (19H01021 and 20K21834), AMED (JP21km0405211, JP21ek0109413, JP21ek0410075, JP21gm4010006 and JP21km0405217), JST Moonshot R&D (JPMJMS2021 and JPMJMS2024), Takeda Science Foundation, and the Bioinformatics Initiative of Osaka University Graduate School of Medicine. Y. Kochi is funded by grants from Nanken-Kyoten, TMDU and Medical Research Center Initiative for High Depth Omics. S.R. is supported by UH2AR067677, U01HG009379, R01AR063759 and U01HG012009.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

K. Ishigaki, S.S., C.T., Y.O. and S.R. conceived and designed the study. K. Ishigaki wrote the manuscript with critical input from S.S., C.T., Y.L., Y.O. and S.R. K. Ishigaki conducted meta-analysis and all GWAS downstream analyses with the help of S.S., C.T., T. Amariuta, Y.L., Y.O. and S.R. K. Yamaguchi and Y. Kochi conducted PADI4 long-read sequencing and PADI4 sQTL analysis. M. Koido, K.T., Y. Kamatani and C.T. contributed to construction of the population-specific reference panel. K. Ishigaki, S.S., C.T., K.S., V.A.L., I.C.S., S.V., D.P., J.B., G.X., J.Z., C.I.A., E.K., R.J.C., K.A.S., M. Kerick, F.M., M. Traylor, C.M.L., H.X., R.S., T. Arayssi, J.M., L.K., Y.O. and S.R. conducted GWAS analyses. C.T., C.L.T., V.A.L., S.V., M. Takahashi, X.W., L.L., T.L., D.P., A.B., G.O., J.B., S.M., K.P.L., R.J.C., E.W.K., K. Matsuo, F.M., S.E., H.X., K. Ikari, P.K.G., L.P., Y.O. and S.R. contributed to genotyping experiments. C.T., K.S., C.L.T., V.A.L., I.C.S., S.V., K.O., A.M., M.H., H.I., M. Hammoudeh, S.A.E., B.K.M., H.H., H.B., I.W.U., X.W., L.L., T.L., D.P., A.B., G.O., S.M.M.V., A.J.M., S.H., H.T., E.T., A.S., Y.M., Kenichi Yamamoto, S.M., G.X., J.Z., C.I.A., E.K., G.W., I.v.d.H.-B., J.C., K.P.L., R.J.C., H.-S.L., S.-Y.B., K.A.S., N.d.V., L.A., S.R.-D., E.W.K., S.-C.B., R.P.K., J.C.E., X.M., T.H., P.D., M.S., M. Kerick, J.C.D., The BioBank Japan Project, K. Matsuda, K. Matsuo, T.M., F.M., K.F., Y.T., A.K., C.M.L., S.E., H.X., R.S., T. Arayssi, K. Ikari, M. Harigai, P.K.G., Kazuhiko Yamamoto, S.L.B., L.P., J.M., L.K., Y.O. and S.R. contributed to the collection of samples and management of genotype data and clinical information. J.C.D.’s involvement in this project was primarily as faculty at Vanderbilt University Medical Center prior to joining the NIH.

Corresponding authors

Correspondence to Yukinori Okada or Soumya Raychaudhuri.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks the anonymous reviewers 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

Extended Data Fig. 1 PCA and UMAP plot of 1KG Phase 3.

a, We projected each individual’s imputed genotype into a PC space, which was calculated using all individuals in 1KG Phase 3. b, We further conducted UMAP analysis using the top 20 PC scores of all GWAS samples and all individuals in 1KG Phase 3. We plotted all samples in 1KG Phase 3 or plotted samples in each ancestry separately. UMAP plot of all GWAS samples is provided in Fig. 1c.

Extended Data Fig. 2 Effect size heterogeneity between seropositive and seronegative RA.

a, Odds ratio and its 95% confidence interval of seropositive and seronegative RA are plotted. Among the lead variants at 122 significant autosomal loci, we plotted 118 variants common in EUR of 1KG Phase 3 (MAF > 0.01). We present the results from multi-ancestry GWAS and EUR-GWAS. Effect size heterogeneity was assessed in the multi-ancestry GWAS results by Cochran’s Q test (Phet). Pearson’s correlation data are provided. b, Differences (delta) of effect size magnitude between seropositive and seronegative RA. A positive delta indicates a larger magnitude of effect size in seropositive RA than seronegative RA. We defined the standard error (s.e.) of delta by the following formula: \(\sqrt {S.E._{seropositive}^2 + S.E._{seronegative}^2}\). We provided 2 × s.e. of delta as error bars. In seropositive RA GWAS, we had 27,448 cases and 240,149 controls; in seronegative RA GWAS, we had 4,515 cases and 240,149 controls.

Extended Data Fig. 3 Enrichment of high PIP variants within open chromatin regions of 18 blood cell populations.

Enrichment of high PIP variants within open chromatin regions of 18 blood cell populations was analyzed by gchromVAR software. The horizontal dashed line indicates Bonferroni corrected P value threshold (0.05/18 = 0.0027). P values were estimated by the enrichment test implemented in gchromVAR.

Extended Data Fig. 4 Conditional analysis results at the IL2RA and TNFAIP3 loci.

a,b, Conditional analysis was conducted in each cohort, and the results were meta-analyzed using the inverse-variance weighted fixed effect model (a, IL2RA locus; b, TNFAIP3 locus). We used multi-ancestry GWAS results. Results at the TYK2 locus are provided in Extended Data Fig. 6. Variants in LD with the lead variant (r2 > 0.6 both in EUR and EAS ancestries) in each round of conditional analysis are highlighted by different colors.

Extended Data Fig. 5 Haplotype-level association test at the IL6R locus.

Haplotype-level association results at the IL6R locus. We defined haplotypes as shown in the table, and we estimated the dosage of four haplotypes using phased imputed genotype data. We conducted multivariate logistic regression tests in each of EUR cohorts (n = 97,173 in total), and the results were meta-analyzed using the inverse-variance weighted fixed effect model. The effect size estimate and 2 x s.e. are shown in the right panel.

Extended Data Fig. 6 Three ancestry-specific signals at the TYK2 locus.

a,b, Conditional analysis was conducted in each cohort, and the results were meta-analyzed using the inverse-variance weighted fixed effect model (a, EUR-GWAS; b, EAS-GWAS). Variants in LD with the lead variant (r2 > 0.6 in EUR or EAS ancestries) in each round of conditional analysis are highlighted by different colors.

Extended Data Fig. 7 S-LDSC analysis using histone mark annotations.

a, The estimate and its 95% confidence interval of the heritability proportion explained by 396 histone mark annotations are provided. Confidence intervals and the P values indicating non-negative tau were estimated via block-jackknife implemented in the LDSC software (one-sided test). When a heritability enrichment is significant (P < 0.05/396 = 1.3 ×10−4), that annotation is colored by the type of GWAS. b, The best histone mark annotation (H3K4me1 in PMA-I stimulated primary CD4+ T cells) and the best IMPACT annotation (CD4+ T cell T-bet) were jointly modeled in S-LDSC analysis. Tau estimate (per variant heritability in each annotation) normalized by total per variant heritability are provided as the centre, and its 95% confidence interval are provided as error bars. EUR-GWAS results were used. c, P values were calculated by block-jackknife implemented in the LDSC software and indicate the significance of non-negative tau (per variant heritability) of each annotation (one-sided test). Each histone mark annotation is colored by its cell type category. Horizontal dashed line indicates Bonferroni-corrected P-value threshold (0.05/396 = 1.3 ×10−4). Top panel shows the results without controlling the effect of the best IMPACT annotation (CD4+ T cell T-bet), and the bottom panel shows the results with controlling for this effect.

Extended Data Fig. 8 PRS performance comparison between the previous RA GWAS and this GWAS.

The liability scale R2 in the 15 cohorts not included in the previous RA GWAS. We used the multi-ancestry GWAS results reported in Okada et al. and this GWAS to develop PRS models. We used the LOCO approach to evaluate the PRS model based on this GWAS. The differences were assessed by two-sided paired Wilcoxon text (n = 15). Within each boxplot, the horizontal lines reflect the median, the top and bottom of each box reflect the interquartile range (IQR), and the whiskers reflect the maximum and minimum values within each grouping no further than 1.5 × IQR from the hinge.

Extended Data Fig. 9 PRS performances in different ancestral groups.

PRS performances (liability scale R2) are shown for each combination of PRS models and cohort groups for which the PRS was applied (n = 25, 8, and 4, respectively, from the left). The differences between the group with the best performance and the second best were analyzed by two-sided paired Wilcoxon test. Within each boxplot, the horizontal lines reflect the median, the top and bottom of each box reflect the interquartile range (IQR), and the whiskers reflect the maximum and minimum values within each grouping no further than 1.5 × IQR from the hinge. We used the LOCO approach where applicable.

Extended Data Fig. 10 PRS distribution differences between cases and controls.

PRS distribution differences between cases and controls. Multi-ancestry PRS with CD4+ T cell T-bet IMPACT annotation was used. We used the LOCO approach. In each cohort, PRS was scaled using mean and s.d. of the control samples, and individual level data were merged across cohorts in an ancestry group.

Supplementary information

Supplementary Information

Supplementary Note, Supplementary Figs. 1–11 and Supplementary Data.

Reporting Summary

Peer Review File

Supplementary Table

Supplementary Tables 1–14.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ishigaki, K., Sakaue, S., Terao, C. et al. Multi-ancestry genome-wide association analyses identify novel genetic mechanisms in rheumatoid arthritis. Nat Genet 54, 1640–1651 (2022). https://doi.org/10.1038/s41588-022-01213-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01213-w

This article is cited by

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