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:

Systematic discovery and perturbation of regulatory genes in human T cells reveals the architecture of immune networks

Abstract

Gene regulatory networks ensure that important genes are expressed at precise levels. When gene expression is sufficiently perturbed, it can lead to disease. To understand how gene expression disruptions percolate through a network, we must first map connections between regulatory genes and their downstream targets. However, we lack comprehensive knowledge of the upstream regulators of most genes. Here, we developed an approach for systematic discovery of upstream regulators of critical immune factors—IL2RA, IL-2 and CTLA4—in primary human T cells. Then, we mapped the network of the target genes of these regulators and putative cis-regulatory elements using CRISPR perturbations, RNA-seq and ATAC-seq. These regulators form densely interconnected networks with extensive feedback loops. Furthermore, this network is enriched for immune-associated disease variants and genes. These results provide insight into how immune-associated disease genes are regulated in T cells and broader principles about the structure of human gene regulatory networks.

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: Approach to map disease gene networks in human T cells.
Fig. 2: Discovery of upstream regulators of IL2RA, IL-2 and CTLA4.
Fig. 3: Arrayed knockouts validate and characterize screen results.
Fig. 4: Individual regulators act at distinct IL2RA CREs.
Fig. 5: IL2RA regulators form highly interconnected gene networks.
Fig. 6: Coregulated gene sets are enriched for immune disease genes.
Fig. 7: IL2RA regulators affect CREs and genes associated with multiple sclerosis.

Similar content being viewed by others

Data availability

The raw sequencing files generated during this study are available at GEO under accession GSE171737. Transcription factor binding motifs used in this study were downloaded from JASPAR2020 (https://doi.org/10.18129/B9.bioc.JASPAR2020), HOCOMOCO v.11 (https://hocomoco11.autosome.org/) and CIS-BP (http://cisbp.ccbr.utoronto.ca/index.php).

Code availability

The code for this paper is available at https://doi.org/10.5281/zenodo.637164689.

References

  1. 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 

  2. Gallagher, M. D. & Chen-Plotkin, A. S. The post-GWAS era: from association to function. Am. J. Hum. Genet. 102, 717–730 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Liu, X., Li, Y. I. & Pritchard, J. K. Trans effects on gene expression can drive omnigenic inheritance. Cell 177, 1022–1034.e6 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Westra, H.-J. et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat. Genet. 45, 1238–1243 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Claussnitzer, M. et al. FTO obesity variant circuitry and adipocyte browning in humans. N. Engl. J. Med. 373, 895–907 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Smemo, S. et al. Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature 507, 371–375 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Small, K. S. et al. Regulatory variants at KLF14 influence type 2 diabetes risk via a female-specific effect on adipocyte size and body composition. Nat. Genet. 50, 572–580 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. The GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369, 1318–1330 (2020).

    Article  PubMed Central  CAS  Google Scholar 

  10. Califano, A., Butte, A. J., Friend, S., Ideker, T. & Schadt, E. Leveraging models of cell regulation and GWAS data in integrative network-based association studies. Nat. Genet. 44, 841–847 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Schumann, K. et al. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc. Natl Acad. Sci. USA 112, 10437–10442 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Shifrut, E. et al. Genome-wide CRISPR screens in primary human T cells reveal key regulators of immune function. Cell 175, 1958–1971.e15 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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 

  14. 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 

  15. Soskic, B. et al. Chromatin activity at GWAS loci identifies T cell states driving complex immune diseases. Nat. Genet. 51, 1486–1493 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. 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 

  17. Adamson, B. et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell 167, 1867–1882.e21 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Dixit, A. et al. Perturb-seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell 167, 1853–1866.e17 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Datlinger, P. et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat. Methods 14, 297–301 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Jaitin, D. A. et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell 167, 1883–1896.e15 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Schumann, K. et al. Functional CRISPR dissection of gene networks controlling human regulatory T cell identity. Nat. Immunol. 21, 1456–1466 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Abbas, A. K., Trotta, E., Simeonov, R. D., Marson, A. & Bluestone, J. A. Revisiting IL-2: biology and therapeutic prospects. Sci. Immunol. 3, eaat1482 (2018).

    Article  PubMed  Google Scholar 

  23. Spolski, R., Li, P. & Leonard, W. J. Biology and regulation of IL-2: from molecular mechanisms to human therapy. Nat. Rev. Immunol. 18, 648–659 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Bayry, J. CTLA-4: a key protein in autoimmunity. Nat. Rev. Rheumatol. 5, 244–245 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Caudy, A. A., Reddy, S. T., Chatila, T., Atkinson, J. P. & Verbsky, J. W. CD25 deficiency causes an immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like syndrome, and defective IL-10 expression from CD4 lymphocytes. J. Allergy Clin. Immunol. 119, 482–487 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Linker-Israeli, M. et al. Defective production of interleukin 1 and interleukin 2 in patients with systemic lupus erythematosus (SLE). J. Immunol. 130, 2651–2655 (1983).

    CAS  PubMed  Google Scholar 

  27. Schubert, D. et al. Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat. Med. 20, 1410–1416 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sharfe, N., Dadi, H. K., Shahar, M. & Roifman, C. M. Human immune disorder arising from mutation of the chain of the interleukin-2 receptor. Proc. Natl Acad. Sci. USA 94, 3168–3171 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Goudy, K. et al. Human IL2RA null mutation mediates immunodeficiency with lymphoproliferation and autoimmunity. Clin. Immunol. 146, 248–261 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bezrodnik, L., Caldirola, M. S., Seminario, A. G., Moreira, I. & Gaillard, M. I. Follicular bronchiolitis as phenotype associated with CD25 deficiency. Clin. Exp. Immunol. 175, 227–234 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kuehn, H. S. et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science 345, 1623–1627 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ochoa, D. et al. Open Targets Platform: supporting systematic drug–target identification and prioritisation. Nucleic Acids Res. 49, D1302–D1310 (2021).

    Article  CAS  PubMed  Google Scholar 

  33. Bousfiha, A. et al. Human inborn errors of immunity: 2019 update of the IUIS phenotypical classification. J. Clin. Immunol. 40, 66–81 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Lambert, S. A. et al. The human transcription factors. Cell 175, 598–599 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Cortez, J. T. et al. CRISPR screen in regulatory T cells reveals modulators of Foxp3. Nature 582, 416–420 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Li, P. et al. STAT5-mediated chromatin interactions in superenhancers activate IL-2 highly inducible genes: functional dissection of the gene locus. Proc. Natl Acad. Sci. USA 114, 12111–12119 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liao, W., Lin, J.-X. & Leonard, W. J. Interleukin-2 at the crossroads of effector responses, tolerance, and immunotherapy. Immunity 38, 13–25 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Simeonov, D. R. et al. Discovery of stimulation-responsive immune enhancers with CRISPR activation. Nature 549, 111–115 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kemmeren, P. et al. Large-scale genetic perturbations reveal regulatory networks and an abundance of gene-specific repressors. Cell 157, 740–752 (2014).

    Article  CAS  PubMed  Google Scholar 

  40. Hughes, T. R. & de Boer, C. G. Mapping yeast transcriptional networks. Genetics 195, 9–36 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Alon, U. Network motifs: theory and experimental approaches. Nat. Rev. Genet. 8, 450–461 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinf. 9, 559 (2008).

    Article  CAS  Google Scholar 

  43. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Fiers, M. W. E. J. et al. Mapping gene regulatory networks from single-cell omics data. Brief. Funct. Genomics 17, 246–254 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. van Dam, S., Võsa, U., van der Graaf, A., Franke, L. & de Magalhães, J. P. Gene co-expression analysis for functional classification and gene-disease predictions. Brief. Bioinform. 19, 575–592 (2018).

    PubMed  Google Scholar 

  46. Margolin, A. A. et al. ARACNE: an algorithm for the reconstruction of gene regulatory networks in a mammalian cellular context. BMC Bioinf. 7, S7 (2006).

    Article  CAS  Google Scholar 

  47. Kanhere, A. et al. T-bet and GATA3 orchestrate Th1 and Th2 differentiation through lineage-specific targeting of distal regulatory elements. Nat. Commun. 3, 1268 (2012).

    Article  PubMed  CAS  Google Scholar 

  48. Schmidl, C. et al. The enhancer and promoter landscape of human regulatory and conventional T-cell subpopulations. Blood 123, e68–e78 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Tangye, S. G. et al. The ever-increasing array of novel inborn errors of immunity: an interim update by the IUIS Committee. J. Clin. Immunol. 41, 666–679 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Tangye, S. G. et al. Human inborn errors of immunity: 2019 update on the classification from the International Union of Immunological Societies Expert Committee. J. Clin. Immunol. 40, 24–64 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Taylor, K. E., Ansel, K. M., Marson, A., Criswell, L. A. & Farh, K. K.-H. PICS2: next-generation fine mapping via probabilistic identification of causal SNPs. Bioinformatics 37, 3004–3007 (2021).

    Article  CAS  PubMed Central  Google Scholar 

  52. Boyle, E. A., Li, Y. I. & Pritchard, J. K. An expanded view of complex traits: from polygenic to omnigenic. Cell 169, 1177–1186 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. 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 

  54. 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 

  55. 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 

  56. Parnas, O. et al. A genome-wide CRISPR screen in primary immune cells to dissect regulatory networks. Cell 162, 675–686 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Henriksson, J. et al. Genome-wide CRISPR screens in T helper cells reveal pervasive crosstalk between activation and differentiation. Cell 176, 882–896.e18 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Brockmann, M. et al. Genetic wiring maps of single-cell protein states reveal an off-switch for GPCR signalling. Nature 546, 307–311 (2017).

    Article  CAS  PubMed  Google Scholar 

  59. Amit, I. et al. Unbiased reconstruction of a mammalian transcriptional network mediating pathogen responses. Science 326, 257–263 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Cusanovich, D. A., Pavlovic, B., Pritchard, J. K. & Gilad, Y. The functional consequences of variation in transcription factor binding. PLoS Genet. 10, e1004226 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Rubin, A. J. et al. Coupled single-cell CRISPR screening and epigenomic profiling reveals causal gene regulatory networks. Cell 176, 361–376.e17 (2019).

    Article  CAS  PubMed  Google Scholar 

  62. Qiu, X. et al. Inferring causal gene regulatory networks from coupled single-cell expression dynamics using Scribe. Cell Syst. 10, 265–274.e11 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Vaquerizas, J. M., Kummerfeld, S. K., Teichmann, S. A. & Luscombe, N. M. A census of human transcription factors: function, expression and evolution. Nat. Rev. Genet. 10, 252–263 (2009).

    Article  CAS  PubMed  Google Scholar 

  64. Mullard, A. Restoring IL-2 to its cancer immunotherapy glory. Nat. Rev. Drug Discov. 20, 163–165 (2021).

    Article  CAS  PubMed  Google Scholar 

  65. Rowshanravan, B., Halliday, N. & Sansom, D. M. CTLA-4: a moving target in immunotherapy. Blood 131, 58–67 (2018).

    Article  CAS  PubMed  Google Scholar 

  66. Ting, P. Y. et al. Guide Swap enables genome-scale pooled CRISPR-Cas9 screening in human primary cells. Nat. Methods 15, 941–946 (2018).

    Article  CAS  PubMed  Google Scholar 

  67. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Hart, T. & Moffat, J. BAGEL: a computational framework for identifying essential genes from pooled library screens. BMC Bioinf. 17, 164 (2016).

    Article  CAS  Google Scholar 

  69. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J. 17, 10–12 (2011).

    Article  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  71. Smith, T., Heger, A. & Sudbery, I. UMI-tools: modeling sequencing errors in unique molecular identifiers to improve quantification accuracy. Genome Res. 27, 491–499 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  73. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Zhang, Y. et al. Model-based analysis of ChIP–Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. 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 

  80. 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 

  81. Wolf, T. et al. Dynamics in protein translation sustaining T cell preparedness. Nat. Immunol. 21, 927–937 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Lynn, R. C. et al. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature 576, 293–300 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Gustafsson, M. et al. A validated gene regulatory network and GWAS identifies early regulators of T cell-associated diseases. Sci. Transl. Med. 7, 313ra178 (2015).

    Article  PubMed  CAS  Google Scholar 

  84. Liao, W., Lin, J.-X., Wang, L., Li, P. & Leonard, W. J. Modulation of cytokine receptors by IL-2 broadly regulates differentiation into helper T cell lineages. Nat. Immunol. 12, 551–559 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Carlson, M. org.Hs.eg.db: Genome wide annotation for Human. R package version 3.8.2 https://doi.org/10.18129/B9.BIOC.ORG.HS.EG.DB (2019).

  86. Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer Science & Business Media, 2009).

  87. Hahne, F. & Ivanek, R. Visualizing genomic data using Gviz and Bioconductor. Methods Mol. Biol. 1418, 335–351 (2016).

    Article  PubMed  Google Scholar 

  88. Lopez-Delisle, L. et al. pyGenomeTracks: reproducible plots for multivariate genomic datasets. Bioinformatics 37, 422–423 (2021).

    Article  CAS  PubMed  Google Scholar 

  89. Jake. jfreimer/immune_network_paper: Release of pipeline for publication. Zenodo https://doi.org/10.5281/zenodo.6371647 (2022).

Download references

Acknowledgements

We thank members of the Marson and Pritchard laboratories for helpful discussions and manuscript feedback. We thank H. Pimentel for advice on analysis. This research was supported by National Institutes of Health (NIH) grants R01HG008140 (J.K.P.) and RM1-HG007735 (W.J.G.). A.M. held a Career Award for Medical Scientists from the Burroughs Wellcome Fund, is a member of the Parker Institute for Cancer Immunotherapy (PICI), was an investigator at the Chan Zuckerberg Biohub and has received funding from the Innovative Genomics Institute (IGI), the American Endowment Foundation, the Cancer Research Institute (CRI) Lloyd J. Old STAR award, a gift from the Jordan Family, a gift from the Byers family and a gift from B. Bakar. O.S. was supported by the NIH grant T32AI125222. S.N. was supported by a Helen Hay Whitney Fellowship. N.S.-A. was supported by a Stanford Graduate Fellowship and CEHG Fellowship. A.F.C. was supported by an NIH F32 postdoctoral fellowship (5F32GM135996-02). Sorting was carried out at the UCSF Flow Cytometry Core (RRID:SCR_018206) supported in part by NIH grant P30 DK063720 and by the NIH S10 instrumentation grant S10 1S10OD021822-01 and the Gladstone Flow Cytometry Core supported by the James B. Pendleton Charitable Trust. RNA-seq was carried out at the DNA Technologies and Expression Analysis Cores at the UC Davis Genome Center, supported by NIH Shared Instrumentation Grant 1S10OD010786-01. Other sequencing was carried out at the UCSF CAT, supported by a PBBR grant. Some of the computing for this project was performed on the Sherlock cluster. We thank Stanford University and the Stanford Research Computing Center for providing computational resources and support that contributed to these research results.

Author information

Authors and Affiliations

Authors

Contributions

J.W.F., O.S., J.K.P. and A.M. conceptualized the study. J.W.F., S.N. and N.S.-A. performed the formal analysis. J.W.F., O.S., A.K., C.M.G., A.F.C. and J.T.C. performed the investigations. W.J.G., J.K.P. and A.M. provided resources. J.W.F. wrote the original draft of the manuscript. J.W.F., O.S., J.K.P. and A.M. reviewed and edited the manuscript. J.W.F. performed visualization. W.J.G., J.K.P. and A.M. supervised the study. W.J.G., J.K.P. and A.M. acquired funding.

Corresponding authors

Correspondence to Jonathan K. Pritchard or Alexander Marson.

Ethics declarations

Competing interests

A.M. is a compensated cofounder, member of the boards of directors and a member of the scientific advisory boards of Spotlight Therapeutics and Arsenal Biosciences. A.M. is a cofounder, member of the boards of directors and a member of the scientific advisory board of Survey Genomics. A.M. is a compensated member of the scientific advisory board of NewLimit. A.M. owns stock in Arsenal Biosciences, Spotlight Therapeutics, NewLimit, Survey Genomics, PACT Pharma and Merck. A.M. has received fees from 23andMe, PACT Pharma, Juno Therapeutics, Trizell, Vertex, Merck, Amgen, Genentech, AlphaSights, Rupert Case Management, Bernstein and ALDA. A.M. is an investor in and informal advisor to Offline Ventures and a client of EPIQ. The Marson laboratory has received research support from Juno Therapeutics, Epinomics, Sanofi, GlaxoSmithKline, Gilead and Anthem. W.J.G. is a consultant for 10x Genomics, which has licensed IP associated with ATAC-seq. W.J.G. has additional affiliations with Guardant Health (consultant) and Protillion Biosciences (cofounder and consultant). J.W.F. is a consultant for NewLimit. J.W.F., O.S., J.K.P. and A.M. are listed as inventors on a patent application related to this work. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Rahul Roychoudhuri, Andrew Wells and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Quality control of the CRISPR screens.

a, Fluorescence activated cell sorting (FACS) gating for IL2RA, IL-2, and CTLA4 screens. Representative example from the IL2RA screen is shown. b, Abundance of sgRNAs targeting GFP in either the starting plasmid or in the GFP + sorted population (n = 3 donors, 1 plasmid pool). c, Differential enrichment between the high- and low-expression bins for sgRNAs targeting genes that are either expressed or not expressed in CD4 + T cells based on RNA-Seq. d, Abundance of sgRNAs targeting essential genes, fitness genes, non-essential genes, or non-targeting guides in the starting plasmid (n = 1) or in the GFP + sorted samples (n = 3 donors). e, Enrichment of sgRNAs between the GFP + sorted population and starting plasmid. Results from Donor 1 and Donor 2 are depicted. Significant hits were identified with MAGeCK and genes with an FDR-adjusted P < 0.05 across all donors are highlighted. f, Comparison of the number of shared significant hits between the different screens and whether those hits have the same direction of effect on their targets. Two-sided sign test P = 0.002, shared direction of effect = 82%, 95% confidence interval 61-95%. All boxplots show the median, first and third quartiles, and 1.5x the interquartile range.

Extended Data Fig. 2 Arrayed knockouts validate IL-2 and CTLA4 screen results.

a,b, Representative flow cytometry density plots for IL-2 (a) or CTLA4 (b) protein levels after knockout of top screen hits. Knockout of hits that decrease target levels are shown in blue, and knockout of hits that increase target levels are shown in red. c,d, Summary of changes in IL-2 (c) or CTLA4 (d) levels measured using flow cytometry. Screen hits selected for validation are displayed on the y-axis ordered by their effect size in the pooled CRISPR screen. For each knockout, bars show the average change in IL-2 or CTLA4 median fluorescence intensity relative to non-targeting controls. Dots show individual data points, and error bars show standard deviation across two guide RNAs and three donors per guide RNA. Concordant changes between the screen and validation that increase or decrease IL-2/CTLA4 levels are shown in red or blue, respectively. Discordant changes are shown in grey. The average insertion/deletion (indel) percentage at the genomic target site across multiple donors for guide RNA 1 (n = 3) and guide RNA 2 (n = 2) is shown to the right. e, Representative flow cytometry density plots for IL2RA protein levels after cells are grown with exogenous IL-2 or without IL-2 + blocking anti-IL-2 antibody. f, Knockout of top regulators of IL2RA in cells cultured with exogenous IL-2 or without IL-2 + blocking anti-IL-2 antibody. IL2RA median fluorescent values are normalized to AAVS1 control knockouts with exogenous IL-2 (black dashed line). Colored dashed lines show the normalized IL2RA median fluorescent intensity averaged across the AAVS1 control knockouts without IL-2 + blocking anti-IL-2 antibody in each donor.

Extended Data Fig. 3 Downstream mapping of genes and chromatin sites controlled by each IL2RA regulator.

a, mRNA fold change for the CRISPR targeted gene in each knockout sample. Data are presented as the effect size from Limma, with error bars showing the 95% confidence interval. b, Comparison of average changes in IL2RA mRNA levels (RNA-Seq) and protein levels (flow cytometry) for each knockout sample collected for RNA-Seq and ATAC-Seq. c, Percent of significantly changed ATAC-Seq peaks in each knockout sample that contain a known motif for the knocked out transcription factor. d,e, The total number of significantly changed genes (d) or peaks (e) detected via RNA-Seq and ATAC-Seq in each knockout sample. For a-e, n = 3 donors for the RNA-Seq and ATAC-Seq data. f, Summary of changes in IL2RA levels measured using flow cytometry. For each knockout, the change in IL2RA median fluorescence intensity is normalized to AAVS1 knockout alone controls. g, The percent of reads containing insertions/deletions (indels) at the genomic target sites for the guide RNAs and samples in f. Solid line indicates the mean indel percentage across different perturbation combinations.

Extended Data Fig. 4 Direct binding of IL2RA regulators at the IL2RA locus.

Chromatin accessibility measured by ATAC-Seq in AAVS1 control knockouts is shown in black. ATAC-Seq data are shown as normalized read coverage; samples were normalized using the size factors from DESeq2. Results from previous CRISPR activation (CRISPRa) screen38 tiling the IL2RA locus in Jurkat cells is shown in pink. CRISPRa tracks show the log2 enrichment of guide RNAs in cells expressing high, mid, or low levels of IL2RA compared to background. Public ChIP-Seq data for IL2RA regulators in various subsets of human CD4 + T cells (STAT5A, STAT5B, ETS1, GATA3, MYB) or engineered bulk T cells (IRF4) are shown in green. ChIP-Seq data are shown as background subtracted binding in reads per million. ATAC-Seq peaks that were significantly differentially accessible in each knockout are shown in blue. The location of a matching binding motif in a significantly differentially accessible peak for each transcription factor is shown in orange. Where available, public ChIP-Seq tracks are from either two independent studies or individual donors: ETS148,81, GATA347, IRF482, MYB83, STAT5A and STAT5B84. chr, chromosome.

Extended Data Fig. 5 Highly co-regulated gene sets are enriched for immune disease genes.

a, Enrichment of heritability for immune traits compared to non-immune traits in significantly differentially accessible ATAC-Seq peaks for each knockout. Only knockouts with at least 1,000 significantly differentially accessible ATAC-Seq peaks are shown. b, Enrichment of heritability for immune traits compared to non-immune traits in a 100-kb window around co-regulated genes. Enrichment for matched background sets for each knockout (a) or each co-regulation bin (b) are shown. Enrichment calculated using stratified LD score regression. Traits were meta-analyzed using inverse-variance weighting; average enrichment and standard error shown. P-values were calculated by first converting the difference in average enrichments to Z-scores, and then converting Z-scores to two-sided P-values (see Methods). For a, Bonferroni-corrected P-values range from 1.8 × 10−2 to 7.5 × 10−16. For b, Bonferroni corrected P-values range from 2.7 × 10−2 to 6.6 × 10−10. NS, not significant. n = 16 immune traits and n = 15 non-immune traits for a and b.

Extended Data Fig. 6 Multiple sclerosis SNPs within CD4 + T cell ATAC-Seq peaks.

a, Enrichment of heritability in accessible ATAC-Seq peaks for different immune traits. Data are presented as estimated enrichment +/− standard error estimated from stratified LD score regression. b, The number of all protein-coding genes and differentially expressed protein-coding genes with a TSS within 100 kb of a multiple sclerosis SNP. Only high confidence multiple sclerosis SNPs (PICS probability greater than 50%) within differentially accessible ATAC-Seq peaks are shown. c, Editing outcomes in CD37 low- and high-expressing cells after using CRISPR/Cas9 and homology-directed repair templates to edit the SNP rs1465697. Editing was performed with guide RNAs targeting the CD37 CRE (CD37 guide RNA) or a control region (AAVS1 guide RNA). d, Length of deletions after CRISPR editing in CD37 low- and high-expressing cells.

Supplementary information

Supplementary Information

Supplementary Note.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–8.

Supplementary Data 1.

RNA-seq results. Results from the RNA-seq experiment following knockout of top IL2RA regulators. Data were analyzed using Limma. The results table includes the Ensembl gene id (ens_id), gene name (gene_name), log2 fold change (logFC) for knockout/control, adjusted P value (adj.P.Val) and the knockout sample that the data corresponds to (sample).

Supplementary Data 2.

ATAC-seq results. Results from the ATAC-seq experiment following knockout of top IL2RA regulators. Data were analyzed using DESeq2. The results table includes the log2 fold change (log2FoldChange) for knockout/control, adjusted P value (padj), the knockout sample that the data corresponds to (sample), the peak name (peakName) and the peak chromosomal coordinates (peak_chr, peak_start, peak_end).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Freimer, J.W., Shaked, O., Naqvi, S. et al. Systematic discovery and perturbation of regulatory genes in human T cells reveals the architecture of immune networks. Nat Genet 54, 1133–1144 (2022). https://doi.org/10.1038/s41588-022-01106-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01106-y

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