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:

Prioritization of autoimmune disease-associated genetic variants that perturb regulatory element activity in T cells

Abstract

Genome-wide association studies (GWASs) have uncovered hundreds of autoimmune disease-associated loci; however, the causal genetic variants within each locus are mostly unknown. Here, we perform high-throughput allele-specific reporter assays to prioritize disease-associated variants for five autoimmune diseases. By examining variants that both promote allele-specific reporter expression and are located in accessible chromatin, we identify 60 putatively causal variants that enrich for statistically fine-mapped variants by up to 57.8-fold. We introduced the risk allele of a prioritized variant (rs72928038) into a human T cell line and deleted the orthologous sequence in mice, both resulting in reduced BACH2 expression. Naive CD8 T cells from mice containing the deletion had reduced expression of genes that suppress activation and maintain stemness and, upon acute viral infection, displayed greater propensity to become effector T cells. Our results represent an example of an effective approach for prioritizing variants and studying their physiologically relevant effects.

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: Prioritizing GWAS variants using high-throughput reporter assays in Jurkat T cells.
Fig. 2: T-GWAS emVars in T cell-accessible chromatin enrich highly for fine-mapped variants.
Fig. 3: Putative causal variants in a BACH2 intron and upstream of TAGAP.
Fig. 4: Base editing of the BACH2 emVar (rs72928038) reduces BACH2 expression.
Fig. 5: Naive T cells from mice with a deletion overlapping orthologous rs72928038 have reduced transcriptional features of stemness.
Fig. 6: Bach218del CD8 T cells are more prone to effector T cell differentiation after acute viral infection.

Similar content being viewed by others

Data availability

Data generated in this study from all manuscript figures are available in NCBI GEO (GSE197539). The 1000 Genomes Phase 3 reference panel was obtained from ftp://ftp.1000genomes.ebi.ac.uk/vol1/ftp/release/20130502/. DHS data across 733 samples were obtained from https://zenodo.org/record/3838751#.X_IA7-lKg6U. Histone chromatin immunoprecipitation sequencing data were downloaded from ENCODE (encodeproject.org); the specific files utilized are listed in Supplementary Table 20. CAGE-based enhancer annotations were downloaded from https://fantom.gsc.riken.jp/5/datafiles/latest/extra/Enhancers/. ChromHMM was obtained from https://egg2.wustl.edu/roadmap/data/byFileType/chromhmmSegmentations/ChmmModels/core_K27ac/jointModel/final/. HOCOMOCO TF position-weighted matrices were obtained from https://hocomoco11.autosome.ru/downloads_v10. ATAC-seq allelic skew data were obtained from Calderon et al.5 (https://www.nature.com/articles/s41588-019-0505-9; Supplementary Table 1, “significant_ASCs” tab). Chromatin accessibility QTLs were downloaded from Gate et al.65 (https://www.nature.com/articles/s41588-018-0156-2; Supplementary Table 6). DeltaSVM precomputed weights for naive CD4 T cells and Jurkat cells were obtained from http://www.beerlab.org/deltasvm_models/downloads/deltasvm_models_e2e.tar.gz. The EMBL GWAS catalog54 (https://www.ebi.ac.uk/gwas/) was accessed on 10 August 2020. T1D GWAS fine-mapping results were obtained from Onengut-Gumuscu et al.15 (https://www.nature.com/articles/ng.3245; Supplementary Table 1). pcHiC data were obtained from Javierre et al.18 (https://osf.io/u8tzp/). The ImmunoSigDB immunologic signatures database (v7.2) was downloaded from http://www.gsea-msigdb.org/gsea/msigdb/. Tscm Bach2 guide RNA perturbed mouse RNA-seq data were obtained from NCBI GEO (GSE152379). The GRCm38 mouse transcriptome index for Kallisto RNA-seq alignments were obtained from https://github.com/pachterlab/kallisto-transcriptome-indices/releases. The Bach218del (stock 35028) mouse strain is available at The Jackson Laboratory.

Code availability

Code supporting this manuscript is available at https://doi.org/10.5281/zenodo.6302248 (ref. 66) (MPRA analysis), https://doi.org/10.5281/zenodo.6299905 (ref. 67) (base-editing analysis) and https://doi.org/10.5281/zenodo.6038725 (ref. 68) (single-cell RNA-seq analysis). Data visualization, exploratory data analysis and processing were performed using R v3.6.2.

References

  1. Farh, K. K.-H. et al. Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 518, 337–343 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Maurano, M. T. et al. Systematic localization of common disease-associated variation in regulatory DNA. Science 337, 1190–1195 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Roychoudhuri, R. et al. BACH2 regulates CD8+ T cell differentiation by controlling access of AP-1 factors to enhancers. Nat. Immunol. 17, 851–860 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Corces, M. R. et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Genet. 48, 1193–1203 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Calderon, D. et al. Landscape of stimulation-responsive chromatin across diverse human immune cells. Nat. Genet. 51, 1494–1505 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ray, J. P. et al. Prioritizing disease and trait causal variants at the TNFAIP3 locus using functional and genomic features. Nat. Commun. 11, 1237 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. Tewhey, R. et al. Direct identification of hundreds of expression-modulating variants using a multiplexed reporter assay. Cell 165, 1519–1529 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ulirsch, J. C. et al. Systematic functional dissection of common genetic variation affecting red blood cell traits. Cell 165, 1530–1545 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Klein, J. C. et al. A systematic evaluation of the design and context dependencies of massively parallel reporter assays. Nat. Methods 17, 1083–1091 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Inoue, F. et al. A systematic comparison reveals substantial differences in chromosomal versus episomal encoding of enhancer activity. Genome Res. 27, 38–52 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Liu, J. Z. et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet. 47, 979–986 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Beecham, A. H. et al. Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat. Genet. 45, 1353–1360 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. International Multiple Sclerosis Genetics Consortium. Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science 365, eaav7188 (2019).

    Article  PubMed Central  CAS  Google Scholar 

  15. Onengut-Gumuscu, S. et al. Fine mapping of type 1 diabetes susceptibility loci and evidence for colocalization of causal variants with lymphoid gene enhancers. Nat. Genet. 47, 381–386 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Tsoi, L. C. et al. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nat. Genet. 44, 1341–1348 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  18. Javierre, B. M. et al. Lineage-specific genome architecture links enhancers and non-coding disease variants to target gene promoters. Cell 167, 1369–1384 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Chen, J. et al. TAGAP instructs Th17 differentiation by bridging Dectin activation to EPHB2 signaling in innate antifungal response. Nat. Commun. 11, 1913 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Tamehiro, N. et al. T‐cell activation RhoGTPase‐activating protein plays an important role in Th17‐cell differentiation. Immunol. Cell Biol. 95, 729–735 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Duke-Cohan, J. S. et al. Regulation of thymocyte trafficking by Tagap, a GAP domain protein linked to human autoimmunity. Sci. Signal. 11, eaan8799 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Yan, J. et al. T cell-intrinsic IRF5 regulates T cell signaling, migration, and differentiation and promotes intestinal inflammation. Cell Rep. 31, 107820 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Daley, S. R. et al. Rasgrp1 mutation increases naive T-cell CD44 expression and drives mTOR-dependent accumulation of Helios+ T cells and autoantibodies. Elife 2, e01020 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Priatel, J. J. et al. RasGRP1 transduces low-grade TCR signals which are critical for T cell development, homeostasis, and differentiation. Immunity 17, 617–627 (2002).

    Article  CAS  PubMed  Google Scholar 

  25. Roychoudhuri, R. et al. BACH2 represses effector programs to stabilize Treg-mediated immune homeostasis. Nature 498, 506–510 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Yao, C. et al. BACH2 enforces the transcriptional and epigenetic programs of stem-like CD8+ T cells. Nat. Immunol. 22, 370–380 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Schmiedel, B. J. et al. Impact of genetic polymorphisms on human immune cell gene expression. Cell 175, 1701–1715 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Thuronyi, B. W. et al. Continuous evolution of base editors with expanded target compatibility and improved activity. Nat. Biotechnol. 37, 1070–1079 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Fulco, C. P. et al. Activity-by-contact model of enhancer–promoter regulation from thousands of CRISPR perturbations. Nat. Genet. 51, 1664–1669 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Oki, S. et al. ChIP-Atlas: a data-mining suite powered by full integration of public ChIP-seq data. EMBO Rep. 19, e46255 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Alpern, D. et al. BRB-seq: ultra-affordable high-throughput transcriptomics enabled by bulk RNA barcoding and sequencing. Genome Biol. 20, 71 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Kakaradov, B. et al. Early transcriptional and epigenetic regulation of CD8 T cell differentiation revealed by single-cell RNA sequencing. Nat. Immunol. 18, 422–432 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gautam, S. et al. The transcription factor c-Myb regulates CD8+ T cell stemness and antitumor immunity. Nat. Immunol. 20, 337–349 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Willinger, T. et al. Human naive CD8 T cells down-regulate expression of the WNT pathway transcription factors lymphoid enhancer binding factor 1 and transcription factor 7 (T cell factor-1) following antigen encounter in vitro and in vivo. J. Immunol. 176, 1439–1446 (2006).

    Article  CAS  PubMed  Google Scholar 

  35. Yamada, T., Park, C. S., Mamonkin, M. & Lacorazza, H. D. Transcription factor ELF4 controls the proliferation and homing of CD8+ T cells via the Krüppel-like factors KLF4 and KLF2. Nat. Immunol. 10, 618–626 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Araki, K. et al. Translation is actively regulated during the differentiation of CD8+ effector T cells. Nat. Immunol. 18, 1046–1057 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Buckler, J. L., Liu, X. & Turka, L. A. Regulation of T-cell responses by PTEN. Immunol. Rev. 224, 239–248 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Liu, Y.-C. The E3 ubiquitin ligase Itch in T cell activation, differentiation, and tolerance. Semin. Immunol. 19, 197–205 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kaech, S. M. & Cui, W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat. Rev. Immunol. 12, 749–761 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Herndler-Brandstetter, D. et al. KLRG1+ effector CD8+ T cells lose KLRG1, differentiate into all memory T cell lineages, and convey enhanced protective immunity. Immunity 48, 716–729 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Joshi, N. S. et al. Inflammation directs memory precursor and short-lived effector CD8+ T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Reilly, S. K. et al. Direct characterization of cis-regulatory elements and functional dissection of complex genetic associations using HCR-FlowFISH. Nat. Genet. 53, 1166–1176 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Yan, J. et al. Systematic analysis of binding of transcription factors to noncoding variants. Nature 591, 147–151 (2021).

    Article  CAS  PubMed  Google Scholar 

  44. Lee, M. N. et al. Common genetic variants modulate pathogen-sensing responses in human dendritic cells. Science 343, 1246980–1246980 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Ye, C. J. et al. Intersection of population variation and autoimmunity genetics in human T cell activation. Science 345, 1254665 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Fairfax, B. P. et al. Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression. Science 343, 1246949 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Ramos-Rodríguez, M. et al. The impact of proinflammatory cytokines on the β-cell regulatory landscape provides insights into the genetics of type 1 diabetes. Nat. Genet. 51, 1588–1595 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Negron, A., Robinson, R. R., Stüve, O. & Forsthuber, T. G. The role of B cells in multiple sclerosis: current and future therapies. Cell. Immunol. 339, 10–23 (2019).

    Article  CAS  PubMed  Google Scholar 

  49. Sahlén, P. et al. Chromatin interactions in differentiating keratinocytes reveal novel atopic dermatitis– and psoriasis-associated genes. J. Allergy Clin. Immunol. 147, 1742–1752 (2021).

    Article  PubMed  CAS  Google Scholar 

  50. Boyd, M. et al. Characterization of the enhancer and promoter landscape of inflammatory bowel disease from human colon biopsies. Nat. Commun. 9, 1661 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Soemedi, R. et al. Pathogenic variants that alter protein code often disrupt splicing. Nat. Genet. 49, 848–855 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Griesemer, D. et al. Genome-wide functional screen of 3’UTR variants uncovers causal variants for human disease and evolution. Cell 184, 5247–5260 (2021).

    Article  CAS  PubMed  Google Scholar 

  53. Ghoussaini, M. et al. Open Targets Genetics: systematic identification of trait-associated genes using large-scale genetics and functional genomics. Nucleic Acids Res. 49, D1311–D1320 (2021).

    Article  CAS  PubMed  Google Scholar 

  54. Buniello, A. et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 47, D1005–D1012 (2019).

    Article  CAS  PubMed  Google Scholar 

  55. Hollenhorst, P. C. et al. DNA specificity determinants associate with distinct transcription factor functions. PLoS Genet. 5, e1000778 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Qin, W. et al. Efficient CRISPR/Cas9-mediated genome editing in mice by zygote electroporation of nuclease. Genetics 200, 423–430 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. 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 

  59. Kent, W. J., Zweig, A. S., Barber, G., Hinrichs, A. S. & Karolchik, D. BigWig and BigBed: enabling browsing of large distributed datasets. Bioinformatics 26, 2204–2207 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Vaidyanathan, S. et al. Uridine depletion and chemical modification increase Cas9 mRNA activity and reduce immunogenicity without HPLC purification. Mol. Ther. Nucleic Acids 12, 530–542 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Pinello, L. et al. Analyzing CRISPR genome-editing experiments with CRISPResso. Nat. Biotechnol. 34, 695–697 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. de Boer, C. G., Ray, J. P., Hacohen, N. & Regev, A. MAUDE: inferring expression changes in sorting-based CRISPR screens. Genome Biol. 21, 134 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Gate, R. E. et al. Genetic determinants of co-accessible chromatin regions in activated T cells across humans. Nat. Genet. 50, 1140–1150 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Guo, M. H. mhguo1/T_cell_MPRA: Release v1.0.0. Zenodo https://doi.org/10.5281/zenodo.6302248 (2022).

  67. de Boer, C. G. de-Boer-Lab/MAUDE: Release including BACH2 reanalysis code and data. Zenodo https://doi.org/10.5281/zenodo.6299905 (2022).

  68. Pribitzer, S. & Deberg, H. A. Single-cell RNA-seq of Bach2 18del CD8 T cells. Zenodo https://doi.org/10.5281/zenodo.6038725 (2022).

Download references

Acknowledgements

We gratefully acknowledge the contribution of R. Maser and Genetic Engineering Technologies Service, J. Kelmenson and Transgenic Genotyping Service, W. Schott and Flow Cytometry Service and R. Lynch and Genome Technologies Service at The Jackson Laboratory; A. McCaffrey and J. Henderson at Trilink Biotechnologies; the Broad Institute vivarium, Flow Cytometry Core and Genomics Core; and the Benaroya Research Institute vivarium, Flow Cytometry Core, Genomics Core and Bioinformatics Core for expert assistance with the work described in this manuscript. We thank P. Gregersen and B. Diamond for providing genotyped human PBMCs and S. Malkiel for help with processing human PBMCs for ATAC-seq and review of the manuscript. We thank M. Dufort and S. Pribitzer for contributions to single-cell RNA-seq analysis. We thank B. Doughty for discussion on strategies for the base-editing experiments. We thank J.C. Ulirsch and V.M. Green for their critical review of the manuscript. This work is funded by National Institutes of Health grant R25NS065745 (M.H.G.); a Canadian Institutes of Health Research fellowship (C.G.D.) and National Institutes of Health grants K99HG009920. (C.G.D.) and T32AR007108 (M.M.L.); a Helen Hay Whitney postdoctoral fellowship (G.A.N.); EMBO Long-Term Fellowship ALTF486-2018 (M.G.); Cancer Research Institute/Bristol-Myers Squibb Fellow CRI2993 (M.G.); and National Institutes of Health grants U01AI142756 (D.R.L.), RM1HG009490 (D.R.L.), R01AI124693 (D.J.C), NHGRI R01HG008131 (N.H.), P50HG006193 (N.H.), R00HG008179 (R.T.), R35HG011329 (R.T.), R01AI151051 (R.T.), F32AI129249 (J.P.R.) and K22AI153648 (J.P.R.).

Author information

Authors and Affiliations

Authors

Contributions

J.P.R., R.T., K.M. and M.H.G. conceived the study. J.P.R. performed MPRA, ATAC-seq on human CD4 T cells, base-editing experiments and luciferase, with the help of M.G. K.M. created the Bach218del mouse line and performed RNA-seq on mouse naive CD4 and CD8 T cells. M.M.L and I.A.H., with the help of W.F.P. and D.J.C., performed VSV-OVA in vivo mouse experiments. M.H.G, C.G.D., H.A.D., K.M., R.T. and J.P.R. performed data analysis. G.A.N. and D.R.L. provided essential base-editing reagents and designed base-editing strategies. J.P.R. and M.H.G. wrote the manuscript with the help of K.M., R.T. and N.H. All authors have read and approved the manuscript.

Corresponding authors

Correspondence to Nir Hacohen, Ryan Tewhey or John P. Ray.

Ethics declarations

Competing interests

G.A.N. and D.R.L. have filed patent applications on genome-editing agents. D.R.L. is a consultant and equity owner of Beam Therapeutics, Prime Medicine and Pairwise Plants, companies that use genome editing. N.H. holds equity in BioNTech and consults for Related Sciences. The other authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Patrick Gaffney, Naganari Ohkura, 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

Extended Data Fig. 1 T-GWAS MPRA workflow and quality metrics.

a) Extended MPRA workflow. i) Oligonucleotide synthesis of elements containing variants and 200 bp surrounding genomic region; ii) barcode elements through PCR; iii) sequence barcoded elements to link barcodes to elements; iv) insert minimal promoter and GFP between element and barcode; v) transfect library into Jurkat T cells; vi) harvest RNA and pull down GFP mRNA; vii) create cDNA and plasmid sequencing libraries and sequence, comparing the prevalence of barcodes in cDNA to their prevalence in plasmid libraries; viii) compare alleles for differential reporter expression. b) Correlation of barcode prevalence in separate replicates of plasmid libraries (left), biological replicates of cDNA libraries (middle), and between a cDNA library and a plasmid library (right). c) Plot of MPRA expression fold change (log2 RNA/plasmid; y axis) against normalized plasmid tag counts for elements with putative cis-regulatory activity (active; blue) and inactive elements (black) within the T-GWAS library. d) Plot of MPRA expression fold change (log2 RNA/plasmid; y axis) against normalized plasmid tag counts for positive controls and negative controls compared to T-GWAS elements.

Extended Data Fig. 2 Variant locations relative to cis-regulatory features.

a) Location relative to TSSs of all MPRA tested variants, active elements (pCRE), and emVars. b) Enrichment of variants within pCREs (light blue) and emVars (dark blue) within chromHMM-defined genomic regions in human T cells. * for P < 0.05; *** P < 1.4 ×10−3 (Bonferroni-corrected for 36 independent tests). c) Functional enrichment of variants within pCREs and emVars. *** for P < 6.3 ×10−3 (nominal p-value threshold of 0.05 Bonferroni-corrected for 8 independent tests). d) Proportion of inactive element and pCRE variants and emVars that have allelic bias in ATAC-seq. e) Scatter plot comparing MPRA log2 allelic bias (y axis) with allelic bias in ATAC-seq from hematopoietic cells (x axis)5. Red dots are emVars (n = 5) and gray dots are pCRE variants (n = 45). f) Proportion of MPRA inactive and pCRE variants, and emVars that are chromatin accessibility QTLs (caQTLs) from T cells65. g) Scatter plot comparing caQTL effect size (beta; x axis) and MPRA log2 allelic bias (y axis). Red dots are emVars (n = 6) and gray dots are pCREs (n = 22). h) Scatter plot comparing deltaSVM score (x axis) with MPRA log2 allelic bias (y axis) (n = 278). i) Proportion of MPRA inactive and pCRE variants and emVars that overlap TF motifs. j) Scatter plot comparing allele-specific TF binding scores (y axis) and MPRA allelic bias (x axis) for emVars predicted to perturb TF binding (n = 11). Enrichment of emVars for TF ChIP-seq (-log10P on y axis). Calculations for (b and c) are risk ratios (see Methods) with Fisher’s exact test P values and Bonferroni correction (see Supplementary Tables 5 and 6 for exact P values). (d, f, and i) P values calculated using two-sided two proportions z test with no multiple comparisons adjustment. (e, g, h and j) P values are from linear regression F statistic. (k) P values are from a two-sided binomial test.

Extended Data Fig. 3 MPRA prioritizes variants in hundreds of loci.

a) Total number of GWAS loci tested (green) and number of loci with at least one emVar identified (orange) for each disease GWAS. b) Histogram of the number of emVars within each GWAS locus. c) Bar plot showing enrichment (from all loci tested) of DHS alone (left), emVars (middle), and emVars in T cell DHS (right) for PICS fine-mapped variants, with the minimum posterior probability threshold indicated on the x axis and fold enrichment shown on the y axis and bars with darker shades of blue as probability increases. Details of PICS enrichment results are shown in Supplementary Table 10. d) Bar plot showing enrichment of emVars for fine-mapped T1D GWAS loci from Onengut-Gumuscu et al. Statistical fine-mapping posterior probability threshold is shown on the x axis and fold enrichment shown on the y axis and with darker shades of blue as probability increases. For both c) and d), gray numbers below each bar show the number of emVars that are statistically fine-mapped at a given PICS probability threshold. Purple numbers above each bar show the -log10 of the enrichment P value. Enrichment in (c) and (d) were calculated as a risk ratio (see Methods), and P values were determined through a two-sided Fisher’s exact test.

Extended Data Fig. 4 Putative causal variants in the promoters of IRF5 and RASGRP.

a, b) Dotplots showing DHS signal (DHS score) and statistical significance of allelic bias (log10FDR of MPRA allelic bias) for MPRA variants in the region; all tested variants on haplotype (black), significant emVars in DHS (red) (top). Position of variants that are emVars, pCREs, variants tested in MPRA, and disease-associated variants for CD, MS, psoriasis, RA, T1D, and UC from the GWAS Catalog54 (middle). Genes in the locus are shown along with chromatin accessibility profiles (from in Jurkat and specific T cell subsets) and T cell pcHiC loops anchored on the region containing the emVar. Gray line depicts position of the prioritized emVar position with respect to all data types. Statistical significance of allelic biases in (a) and (b) were calculated using a paired Student’s two-sided t-test as described in Methods.

Extended Data Fig. 5 rs72928038 reduces luciferase reporter expression and contacts the BACH2 promoter.

a) Luciferase reporter activity of rs72928038 alleles (n = 3, two independent experiments). b) Promoter capture HiC (pcHiC18) conducted in naïve T cells anchored on the region containing the rs72928038. For (a), statistical significance was calculated using a Student’s two-sided t-test, central tendency is shown as median, and all points are plotted to show dispersion with error bars representing standard deviation. (c) i) Schematic of installing rs72928038 using the evoCDAmax cytosine base editor, achieving 95% base editing. We also created a second condition, separately combining the 95% base-edited cells with WT base-edited cells (combined 50/50) post-nucleofection. (ii) We performed PrimeFlow, staining BACH2 mRNA, and sorted cells based on high and low BACH2 expression. iii) We sequenced the amplicon containing rs72928038 in all sorted populations. iv) Mock data of expected ratios of risk vs. nonrisk alleles in high and low bins of BACH2 expression. If rs72928038 reduces BACH2 expression, one would anticipate the edited risk allele to enrich in low BACH2 expression bins.

Extended Data Fig. 6 Orthologous rs72928038 region binds STATs and ETS1 and deletion of the region in CD8 T cells partially recapitulates transcriptional phenotypes of Bach2-deficient Tscms.

a) STAT and ETS1 TF ChIP-seq peaks30 overlapping mouse rs72928038 ortholog. b) PCA on RNA-seq of naïve CD4 T cells from WT and Bach218del mice. c) Bach2 expression in WT and Bach218del naïve CD4 T cells from RNA-seq normalized counts. d) GSEA enrichment of Bach218del vs. WT naïve CD8 (left) and CD4 (right) T cells. Depicted GSEA results for a gene set derived from genes upregulated in empty vector vs. Bach2 sgRNA-transduced Tscms (d left and right) and genes upregulated in Bach2 sgRNA-transduced Tscms vs. empty vector (d middle). Full GSEA results are shown in Supplementary Table 16. e) Expression of genes in Tscms that have been transduced with empty vector or a Bach2 sgRNA (same experiment as in d) for differentially expressed genes in Bach218del vs. WT naïve CD8 T cells. Genes upregulated in Bach218del T cells as compared to WT are on the left and downregulated are on the right. Normalized enrichment score (NES) in (d) was calculated based on observed enrichment as compared to enrichments from permuted data as previously described and statistical significance shown as the false discovery rate (q). P value in (b) was determined by a two-sided Wald test from normalized counts and adjusted P value was determined using Benjamini Hochberg adjustment. For (b-e), n = 3 independent animals.

Extended Data Fig. 7 Bach218del CD8 T cells have reduced memory-precursor and enhancer effector phenotypes.

(ae) Flow cytometry histograms depicting WT (orange) and Bach218del (turquoise) expression of CD62L (a), Eomes (b), CD127 (c), KLRG1 (d), and CX3CR1 (e). For (ae), n = 10 independent animals per experiment analyzed over 2 experiments, and statistical significance was calculated using a Student’s paired two-sided t-test with no adjustments for multiple testing.

Extended Data Fig. 8 Single-cell RNA-seq of WT and Bach218del CD8 T cells at 8 dpi with VSV-OVA.

a) UMAP plots depicting the composition of cells from different pools (left) and between mice (right). b) Heatmap depicting the top genes representing each cluster in Fig. 6f. c) UMAP plot indicating regions from Fig. 6h with significant enrichment of WT or Bach218del cells. d) Line plots depicting the relative frequencies of WT and Bach218del cells, for each of 5 replicates/group, within each cluster depicted in Fig. 6f. Statistical significance for (c) was assessed using a two-sided permutation test with N = 5,000 permutations, identifying cellular enrichment outside of the 99% confidence interval (see methods). (d) was calculated using a Student’s paired two-sided t-test with no adjustments for multiple testing.

Extended Data Fig. 9 Flow cytometry gating strategy for Bach218del and WT OTI cotransfer VSV-OVA experiment.

Cells are gated on the lymphocyte population and single cells, followed by gating out dead cells, gating on activated CD8 T cells, and identifying cells from each genotype using CD45.1.2 (WT) and CD45.2 (Bach218del). Cells were further assessed for their prevalence in effector (KLRG1 + ) or memory precursor (CD127 + ) populations.

Supplementary information

Supplementary Information

Supplementary Note, Methods, Tables 1–20 and References.

Reporting Summary

Peer Review File

Supplementary Table 1

All tables.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mouri, K., Guo, M.H., de Boer, C.G. et al. Prioritization of autoimmune disease-associated genetic variants that perturb regulatory element activity in T cells. Nat Genet 54, 603–612 (2022). https://doi.org/10.1038/s41588-022-01056-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01056-5

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research