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:

Chromatin activity at GWAS loci identifies T cell states driving complex immune diseases

Abstract

Immune-disease-associated variants are enriched in active chromatin regions of T cells and macrophages. However, whether these variants function in specific cell states is unknown. Here we stimulated T cells and macrophages in the presence of 13 cytokines and profiled active and open chromatin regions. T cell activation induced major chromatin remodeling, while the presence of cytokines fine-tuned the magnitude of changes. We developed a statistical method that accounts for subtle changes in the chromatin landscape to identify SNP enrichment across cell states. Our results point towards the role of immune-disease-associated variants in early rather than late activation of memory CD4+ T cells, with modest differences across cytokines. Furthermore, variants associated with inflammatory bowel disease are enriched in type 1 T helper (TH1) cells, whereas variants associated with Alzheimer’s disease are enriched in different macrophage cell states. Our results represent an in-depth analysis of immune-disease-associated variants across a comprehensive panel of activation states of T cells and macrophages.

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: Quantitative changes in chromatin activity distinguish immune cell states.
Fig. 2: Overview of the CHEERS method.
Fig. 3: Enrichment of disease-associated SNPs in H3K27ac regions in cytokine-induced cell states.
Fig. 4: Example loci that drive the enrichment of SNPs associated with immune diseases in cytokine-induced cell states.

Similar content being viewed by others

Data availability

All of the raw-sequencing files are deposited in the EGA. ATAC-seq data are available at EGAS00001003501. H3K27ac data are available at EGAS00001002749.

Code availability

CHEERS code is available through GitHub (https://github.com/trynkaLab/CHEERS).

References

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

    Article  CAS  Google Scholar 

  2. ENCODE Project Consortium An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  5. 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  Google Scholar 

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

    Article  Google Scholar 

  7. Trynka, G. et al. Disentangling the effects of colocalizing genomic annotations to functionally prioritize non-coding variants within complex-trait loci. Am. J. Hum. Genet. 97, 139–152 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. Alasoo, K. et al. Shared genetic effects on chromatin and gene expression indicate a role for enhancer priming in immune response. Nat. Genet. 50, 424–431 (2018).

    Article  CAS  Google Scholar 

  10. Martinez, F. O. & Gordon, S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 6, 13 (2014).

    Article  Google Scholar 

  11. Khor, B., Gardet, A. & Xavier, R. J. Genetics and pathogenesis of inflammatory bowel disease. Nature 474, 307–317 (2011).

    Article  CAS  Google Scholar 

  12. Yoshizaki, A. et al. Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions. Nature 491, 264–268 (2012).

    Article  CAS  Google Scholar 

  13. Mascanfroni, I. D. et al. IL-27 acts on DCs to suppress the T cell response and autoimmunity by inducing expression of the immunoregulatory molecule CD39. Nat. Immunol. 14, 1054–1063 (2013).

    Article  CAS  Google Scholar 

  14. Feldmann, M. Development of anti-TNF therapy for rheumatoid arthritis. Nat. Rev. Immunol. 2, 364–371 (2002).

    Article  CAS  Google Scholar 

  15. Hall, J. C. & Rosen, A. Type I interferons: crucial participants in disease amplification in autoimmunity. Nat. Rev. Rheumatol. 6, 40–49 (2010).

    Article  CAS  Google Scholar 

  16. Glinos, D. A., Soskic, B., Jostins, L., Sansom, D. M. & Trynka, G. Genomic profiling of T cell activation reveals dependency of memory T cells on CD28 costimulation. Preprint at bioRxiv https://doi.org/10.1101/421099 (2018).

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

    Article  Google Scholar 

  18. Minoux, M. et al. Gene bivalency at Polycomb domains regulates cranial neural crest positional identity. Science 355, eaal2913 (2017).

    Article  Google Scholar 

  19. Mueller, B. et al. Widespread changes in nucleosome accessibility without changes in nucleosome occupancy during a rapid transcriptional induction. Genes Dev. 31, 451–462 (2017).

    Article  CAS  Google Scholar 

  20. Pickrell, J. K. Joint analysis of functional genomic data and genome-wide association studies of 18 human traits. Am. J. Hum. Genet. 94, 559–573 (2014).

    Article  CAS  Google Scholar 

  21. Hu, X. et al. Integrating autoimmune risk loci with gene-expression data identifies specific pathogenic immune cell subsets. Am. J. Hum. Genet. 89, 496–506 (2011).

    Article  CAS  Google Scholar 

  22. Calderon, D. et al. Inferring relevant cell types for complex traits by using single-cell gene expression. Am. J. Hum. Genet. 101, 686–699 (2017).

    Article  CAS  Google Scholar 

  23. Tansey, K. E., Cameron, D. & Hill, M. J. Genetic risk for Alzheimer’s disease is concentrated in specific macrophage and microglial transcriptional networks. Genome Med. 10, 14 (2018).

    Article  Google Scholar 

  24. Sénécal, V. et al. Production of IL-27 in multiple sclerosis lesions by astrocytes and myeloid cells: modulation of local immune responses. Glia 64, 553–569 (2016).

    Article  Google Scholar 

  25. Lalive, P. H. et al. Increased interleukin-27 cytokine expression in the central nervous system of multiple sclerosis patients. J. Neuroinflammation 14, 144 (2017).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  27. Calderon, D. et al. Landscape of stimulation-responsive chromatin across diverse human immune cells. Preprint at bioRxiv https://doi.org/10.1101/409722 (2018).

  28. Schett, G., Elewaut, D., McInnes, I. B., Dayer, J.-M. & Neurath, M. F. How cytokine networks fuel inflammation: toward a cytokine-based disease taxonomy. Nat. Med. 19, 822–824 (2013).

    Article  CAS  Google Scholar 

  29. McInnes, I. B., Buckley, C. D. & Isaacs, J. D. Cytokines in rheumatoid arthritis — shaping the immunological landscape. Nat. Rev. Rheumatol. 12, 63–68 (2016).

    Article  CAS  Google Scholar 

  30. Babaloo, Z. et al. Increased IL-17A but decreased IL-27 serum levels in patients with multiple sclerosis. Iran. J. Immunol. 10, 47–54 (2013).

    CAS  PubMed  Google Scholar 

  31. Jamshidian, A., Shaygannejad, V., Pourazar, A., Zarkesh-Esfahani, S.-H. & Gharagozloo, M. Biased Treg/Th17 balance away from regulatory toward inflammatory phenotype in relapsed multiple sclerosis and its correlation with severity of symptoms. J. Neuroimmunol. 262, 106–112 (2013).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  33. Lo, B. et al. Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 349, 436–440 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  35. Tivol, E. A. et al. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 3, 541–547 (1995).

    Article  CAS  Google Scholar 

  36. Neurath, M. F. et al. The transcription factor T-bet regulates mucosal T cell activation in experimental colitis and Crohn’s disease. J. Exp. Med. 195, 1129–1143 (2002).

    Article  CAS  Google Scholar 

  37. Gosselin, D. et al. An environment-dependent transcriptional network specifies human microglia identity. Science 356, eaal3222 (2017).

    Article  Google Scholar 

  38. Schmidl, C., Rendeiro, A. F., Sheffield, N. C. & Bock, C. ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors. Nat. Methods 12, 963–965 (2015).

    Article  CAS  Google Scholar 

  39. 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  Google Scholar 

  40. Jiang, H., Lei, R., Ding, S.-W. & Zhu, S. Skewer: a fast and accurate adapter trimmer for next-generation sequencing paired-end reads. BMC Bioinformatics 15, 182 (2014).

    Article  Google Scholar 

  41. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595 (2010).

    Article  Google Scholar 

  42. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  Google Scholar 

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

    Article  Google Scholar 

  44. 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  Google Scholar 

  45. Ferreira, M. A. et al. Shared genetic origin of asthma, hay fever and eczema elucidates allergic disease biology. Nat. Genet. 49, 1752–1757 (2017).

    Article  CAS  Google Scholar 

  46. Jansen, I. E. et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat. Genet. 51, 404–413 (2019).

    Article  CAS  Google Scholar 

  47. Demenais, F. et al. Multiancestry association study identifies new asthma risk loci that colocalize with immune-cell enhancer marks. Nat. Genet. 50, 42–53 (2018).

    Article  CAS  Google Scholar 

  48. Trynka, G. et al. Dense genotyping identifies and localizes multiple common and rare variant association signals in celiac disease. Nat. Genet. 43, 1193–1201 (2011).

    Article  CAS  Google Scholar 

  49. Nelson, C. P. et al. Association analyses based on false discovery rate implicate new loci for coronary artery disease. Nat. Genet. 49, 1385–1391 (2017).

    Article  CAS  Google Scholar 

  50. Hoffmann, T. J. et al. A large electronic-health-record-based genome-wide study of serum lipids. Nat. Genet. 50, 401–413 (2018).

    Article  CAS  Google Scholar 

  51. Jostins, L. et al. Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012).

    Article  CAS  Google Scholar 

  52. International Multiple Sclerosis Genetics Consortium (IMSGC) Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat. Genet. 45, 1353–1360 (2013).

    Article  Google Scholar 

  53. Chang, D. et al. A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nat. Genet. 49, 1511–1516 (2017).

    Article  CAS  Google Scholar 

  54. Tsoi, L. C. et al. Large scale meta-analysis characterizes genetic architecture for common psoriasis associated variants. Nat. Commun. 8, 15382 (2017).

    Article  CAS  Google Scholar 

  55. Langefeld, C. D. et al. Transancestral mapping and genetic load in systemic lupus erythematosus. Nat. Commun. 8, 16021 (2017).

    Article  CAS  Google Scholar 

  56. Bossini-Castillo, L., López-Isac, E., Mayes, M. D. & Martín, J. Genetics of systemic sclerosis. Semin. Immunopathol. 37, 443–451 (2015).

    Article  CAS  Google Scholar 

  57. 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  Google Scholar 

  58. 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  Google Scholar 

Download references

Acknowledgements

This work was funded by the Open Targets grant (OTAR040) awarded to G.T. G.T. is supported by the Wellcome Trust (grant WT206194). E.C.-G. is supported by a Gates Cambridge Scholarship (OPP1144). L.B.-C. is supported by the MRC Skills Development Fellowship (MR/N014995/1).

Author information

Authors and Affiliations

Authors

Contributions

G.T. and B.S. conceived and designed the project. B.S., E.C.-G. and D.J.S. carried out the experimental work. B.S. and E.C.-G. performed the data analysis. B.S., E.C.-G., G.T., W.C.R., N.N., J.E.-G., D.F.T., C.G.C.L., D.W., L.B.-C. and P.G.B. interpreted the results. B.S., D.W., E.C.-G. and G.T. developed the CHEERS method. L.B.-C. calculated LD blocks. G.T. supervised the analysis. G.T., B.S., E.C.-G., W.C.R., N.N., J.E.-G., D.F.T., C.G.C.L., D.W. and P.G.B. wrote the manuscript.

Corresponding author

Correspondence to Gosia Trynka.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Fig. 1 Quality control of ATAC and H3K27ac data.

a) Purity of naive T cells (CD4+CD45RA+), memory T cells (CD4+CD45RO+) and macrophages. Representative plots were from three biologically independent samples in T cells and two biologically independent samples in macrophages. b) Number of reads, number of peaks, fraction of reads in peaks (FRiP) and mean q-value of H3K27ac and ATAC peaks. Peak q-values were determined using MACS2. Number of samples in H3K27ac is: 38 (naive_16h), 35 (naive_5d), 32 (memory_16h), 35 (memory_5d), 16 (macrophage_6h) and 17 (macrophage_24h). Number of samples in ATAC is: 36 (naive_16h), 31 (naive_5d), 34 (memory_16h), 28 (memory_5d), 19 (macrophage_6h) and 20 (macrophage_24h). Minima, maxima, centre, and percentiles are reported in Supplementary Table 2. c) Number of peaks in resting and stimulated naive and memory T cells. Stimulated cells were cultured with anti-CD3/anti-CD28 beads. Number of samples in H3K27ac is: 15 (resting) and 125 (stimulated). Number of samples in ATAC is: 10 (resting) and 119 (stimulated). We performed two tailed t-test (ChM: t = 4.58, df = 25.1, p-value = 0.0001; ATAC: t = 0.87, df = 10.55, p-value = 0.41). Minima, maxima, centre, and percentiles are reported in Supplementary table 2. d) Percentage of reproducible peaks per condition. Reproducible peaks are peaks that were observed in at least two biological replicates of the same condition. Number of samples in H3K27ac and ATAC is the same as in panel B. Minima, maxima, centre, and percentiles are reported in Supplementary table 2. e) Comparison of read counts in peaks between biologically independent samples (between individuals) and 15 technical replicates (within an individual) included in our study. The red line represents the mean R2.

Supplementary Fig. 2 Chromatin regulatory landscape in cytokine induced immune cell states captured by H3K27ac ChM-seq and ATAC-seq.

a) Distribution of ATAC and H3K27ac peak length. b) Distribution of ATAC and H3K27ac peaks relative to TSS. c) Proportion of ATAC and H3K27ac in different genomic annotations. d) Overlap between ATAC and H3K27ac peaks (upper panel) and of the genes in proximity to peaks (bottom panel). e) Pathway enrichment analysis using genes in proximity to the top and bottom 1% variable H3K27ac peaks. P-values were calculated using g:ProfileR with default parameters. The number of genes used for the enrichment analysis was 1,156 (nearby top variable peaks) and 1,253 (nearby bottom variable peaks).

Supplementary Fig. 3 PCA of H3K27ac and ATAC data.

a) PCA using H3K27ac and b) ATAC-seq data across all cell types and cell states within the study (left), and T cells only (right). Each sample is colored by broad cell lineage, while the color shade represents duration of activation. Circles represent resting cells and triangles correspond to stimulated cells. PCA plots were derived using 73 naive T cells, 67 memory T cells and 33 macrophage states for H3K27ac (a) and 67 naive T cells, 62 memory T cells and 39 macrophage states for ATAC (b). c) PCA of T cells using reads in H3K27ac peaks within cell type and stimulation time point. PCA plots were derived using 31 naive D5 T cells, 29 memory D5 T cells, 14 macrophages stimulated for 6h and 15 macrophages stimulated for 24h cell states.

Supplementary Fig. 4 ATAC-seq read pile-ups at hallmark loci for cytokine induced cell states.

ATAC-seq read pile ups in regions proximal to condition specific hallmark genes: CD3 (T cells), CD14 (macrophages), KI67 (T cell activation), TBX21 (Th1), GATA3 (Th2), IL35B (that is EBI3, iTreg), IFIT2 (IFN-induced). Genomic coordinates (GRCh38) for each gene are labeled. ATAC-seq tracks were generated after merging three biologically independent samples per cell state.

Supplementary Fig. 5 Binary SNP-peak overlap does not discriminate between closely related cell states.

We used GoShifter to test for SNP enrichment across cytokine induced cell states. GoShifter was performed after merging three biologically independent samples per cell state. P-values were calculated using 10,000 permutations implemented in GoShifter.

Supplementary Fig. 6 Power calculations for CHEERS.

Colors represent percentile of top cell state peak specificity ranks which were sampled to compute power of detecting significant enrichment under different percentages (x-axis) of SNPs mapping in these top peaks.

Supplementary Fig. 7 Disease SNP enrichment in H3K27ac regions in blood cell types from the BLUEPRINT and ROADMAP projects.

a) One-sided p-values are reported from a discrete uniform distribution. CHEERS was performed after merging one to six biological replicates per cell type (sample sizes are shown in Supplementary Table 2) resulting in 236,222 peaks. GWAS data and the statistical methods are described in the “GWAS data processing” and “CHEERS” sections of the Methods. The dotted gray line marks the nominal p-value threshold of 0.05, while the solid gray line represents the Bonferroni-corrected significance threshold (p-value ≤ 0.0026). b) Correlation of -log10(p-values) across diseases. Pearson’s correlations between 13 diseases were calculated using 20 cell types. c) Proportion of peaks shared by multiple immune cell types (barplot) and coefficient of variation of read counts per peak between cell types in the BLUEPRINT data. Number of peaks is 236,222. d) One-sided p-values are reported from a discrete uniform distribution. CHEERS was performed on one sample resulting in 174,296 peaks. GWAS data and the statistical method are described in the “GWAS data processing” and “CHEERS” sections of the Methods.

Supplementary Fig. 8 Disease SNP enrichment in TSS proximal and distal H3K27ac regions.

One-sided p-values are reported from a discrete uniform distribution. CHEERS was performed after merging three biologically independent samples. GWAS data and the statistical method are described in the “GWAS data processing” and “CHEERS” sections of the Methods. The dotted gray line marks the nominal p-value threshold of 0.05, while the solid gray line represents the Bonferroni-corrected significance threshold (p-value ≤ 9 x 10-4). A) Enrichment using proximal (≤5kb from TSS, 79,419 peaks) and B) distal peaks (>5kb from TSS, 48,305 peaks).

Supplementary Fig. 9 Clustering by cell state specificity score of peaks overlapping SNPs associated with Crohn’s disease, rheumatoid arthritis and Alzheimer’s disease.

a) Each row corresponds to a H3K27ac peak, while each column corresponds to a cytokine induced cell state. Shades of blue represent specificity of a peak (specificity rank of a peak normalized to the mean specificity rank of all peaks). b,c,d) Genomic coordinates (GRCh38) for each gene are labeled. H3K27ac tracks are generated after merging three biologically independent samples per cell state. b) Read pileups of H3K27ac (blue) and ATAC-seq (red) peaks at loci driving the enrichment of allergy variants in T cell activation. c) Read pileups of H3K27ac (blue) and ATAC-seq (red) peaks at loci driving the enrichment of RA variants in T cell activation. d) Read pileups of H3K27ac (blue) and ATAC-seq (red) peaks at loci driving the enrichment of IBD variants in Th1 cells.

Supplementary Fig. 10 CHEERS SNP enrichment in ATAC-seq defined regions in cytokine induced cell states.

a) One-sided p-values are reported from a discrete uniform distribution. CHEERS was performed after merging three biologically independent samples resulting in 136,692 peaks. GWAS data and the statistical method are described in the “GWAS data processing” and “CHEERS” sections of the Methods. The dotted gray line marks the nominal p-value threshold of 0.05, while the solid gray line represents the Bonferroni-corrected significance threshold (p-value ≤ 9 x 10-4). b) Correlation of 55 p-value ranks from CHEERS SNP enrichments obtained using H3K27ac and ATAC-seq. The grey line depicts the identity line. Pearson’s correlation coefficient (PCC) is provided below each disease label.

Supplementary Fig. 11 Disease SNP enrichment in TSS proximal and distal ATAC peaks.

One-sided p-values are reported from a discrete uniform distribution. CHEERS was performed after merging three biologically independent samples. GWAS data and the statistical method are described in the “GWAS data processing” and “CHEERS” sections of the Methods. The dotted gray line marks the nominal p-value threshold of 0.05, while the solid gray line represents the Bonferroni-corrected significance threshold (p-value ≤ 9 x 10-4). A) CHEERS was performed on proximal ATAC peaks (≤ 5kb from TSS, 88,150 peaks). B) CHEERS was performed on distal peaks (> 5kb from TSS, 48,543 peaks).

Supplementary information

Supplementary Text and Figures

Supplementary Figs. 1–11

Reporting Summary

Supplementary Table 1

Cytokine concentrations.

Supplementary Table 2

Sample sizes and violin plot details.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Soskic, B., Cano-Gamez, E., Smyth, D.J. et al. Chromatin activity at GWAS loci identifies T cell states driving complex immune diseases. Nat Genet 51, 1486–1493 (2019). https://doi.org/10.1038/s41588-019-0493-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-019-0493-9

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