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:

Interpreting type 1 diabetes risk with genetics and single-cell epigenomics

Abstract

Genetic risk variants that have been identified in genome-wide association studies of complex diseases are primarily non-coding1. Translating these risk variants into mechanistic insights requires detailed maps of gene regulation in disease-relevant cell types2. Here we combined two approaches: a genome-wide association study of type 1 diabetes (T1D) using 520,580 samples, and the identification of candidate cis-regulatory elements (cCREs) in pancreas and peripheral blood mononuclear cells using single-nucleus assay for transposase-accessible chromatin with sequencing (snATAC–seq) of 131,554 nuclei. Risk variants for T1D were enriched in cCREs that were active in T cells and other cell types, including acinar and ductal cells of the exocrine pancreas. Risk variants at multiple T1D signals overlapped with exocrine-specific cCREs that were linked to genes with exocrine-specific expression. At the CFTR locus, the T1D risk variant rs7795896 mapped to a ductal-specific cCRE that regulated CFTR; the risk allele reduced transcription factor binding, enhancer activity and CFTR expression in ductal cells. These findings support a role for the exocrine pancreas in the pathogenesis of T1D and highlight the power of large-scale genome-wide association studies and single-cell epigenomics for understanding the cellular origins of complex disease.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Genome-wide association and fine mapping identify T1D risk signals.
Fig. 2: Reference map of single-cell chromatin accessibility from T1D-relevant tissues.
Fig. 3: Cell-type-specific enrichment and mechanisms of T1D risk variants.
Fig. 4: Fine-mapped T1D variant regulates CFTR in pancreatic ductal cells.

Similar content being viewed by others

Data availability

Full summary statistics for the T1D GWAS have been deposited into the NHGRI-EBI GWAS catalogue with accession number GCST90014023 and can be downloaded from http://ftp.ebi.ac.uk/pub/databases/gwas/summary_statistics/GCST90014001-GCST90015000/GCST90014023/. Sequencing data for snATAC–seq have been deposited into the NCBI Gene Expression Omnibus (GEO) with accession number GSE163160. Data obtained from the TFClass database are available at http://tfclass.bioinf.med.uni-goettingen.de/ and from the PanglaoDB database at https://panglaodb.se/Source data are provided with this paper.

Code availability

Code used for processing snATAC–seq data sets and clustering cells is available at https://github.com/kjgaulton/pipelines/tree/master/T1D_snATAC_pipeline.

References

  1. Claussnitzer, M. et al. A brief history of human disease genetics. Nature 577, 179–189 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  Google Scholar 

  3. Katsarou, A. et al. Type 1 diabetes mellitus. Nat. Rev. Dis. Primers 3, 17016 (2017).

    Article  PubMed  Google Scholar 

  4. 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 

  5. Barrett, J. C. et al. Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat. Genet. 41, 703–707 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bradfield, J. P. et al. A genome-wide meta-analysis of six type 1 diabetes cohorts identifies multiple associated loci. PLoS Genet. 7, e1002293 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Taliun, D. et al. Sequencing of 53,831 diverse genomes from the NHLBI TOPMed Program. Nature 590, 290–299 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Aylward, A., Chiou, J., Okino, M.-L., Kadakia, N. & Gaulton, K. J. Shared genetic risk contributes to type 1 and type 2 diabetes etiology. Hum. Mol. Genet. ddy314 (2018).

  9. Raeder, H. et al. Mutations in the CEL VNTR cause a syndrome of diabetes and pancreatic exocrine dysfunction. Nat. Genet. 38, 54–62 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Ramos, P. S. et al. A comprehensive analysis of shared loci between systemic lupus erythematosus (SLE) and sixteen autoimmune diseases reveals limited genetic overlap. PLoS Genet. 7, e1002406 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gibson-Corley, K. N., Meyerholz, D. K. & Engelhardt, J. F. Pancreatic pathophysiology in cystic fibrosis. J. Pathol. 238, 311–320 (2016).

    Article  PubMed  Google Scholar 

  12. Sharer, N. et al. Mutations of the cystic fibrosis gene in patients with chronic pancreatitis. N. Engl. J. Med. 339, 645–652 (1998).

    Article  CAS  PubMed  Google Scholar 

  13. Diaferia, G. R. et al. Dissection of transcriptional and cis-regulatory control of differentiation in human pancreatic cancer. EMBO J. 35, 595–617 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

  15. McWilliams, R. R. et al. Cystic fibrosis transmembrane conductance regulator (CFTR) gene mutations and risk for pancreatic adenocarcinoma. Cancer 116, 203–209 (2010).

    CAS  PubMed  Google Scholar 

  16. Noone, P. G. et al. Cystic fibrosis gene mutations and pancreatitis risk: relation to epithelial ion transport and trypsin inhibitor gene mutations. Gastroenterology 121, 1310–1319 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Virostko, J. et al. Pancreas volume declines during the first year after diagnosis of type 1 diabetes and exhibits altered diffusion at disease onset. Diabetes Care 42, 248–257 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Campbell-Thompson, M., Rodriguez-Calvo, T. & Battaglia, M. Abnormalities of the exocrine pancreas in type 1 diabetes. Curr. Diab. Rep. 15, 79 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Camunas-Soler, J. et al. Patch-seq links single-cell transcriptomes to human islet dysfunction in diabetes. Cell Metab. 31, 1017–1031 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Fasolino, M. et al. Multiomics single-cell analysis of human pancreatic islets reveals novel cellular states in health and type 1 diabetes. Preprint at https://doi.org/10.1101/2021.01.28.428598 (2021).

  21. Hart, N. J. et al. Cystic fibrosis-related diabetes is caused by islet loss and inflammation. JCI Insight 3, e98240 (2018).

    Article  PubMed Central  Google Scholar 

  22. Valle, A. et al. Reduction of circulating neutrophils precedes and accompanies type 1 diabetes. Diabetes 62, 2072–2077 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Navis, A. & Bagnat, M. Loss of cftr function leads to pancreatic destruction in larval zebrafish. Dev. Biol. 399, 237–248 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lin, Y. et al. Genome-wide association meta-analysis identifies GP2 gene risk variants for pancreatic cancer. Nat. Commun. 11, 3175 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wolpin, B. M. et al. Genome-wide association study identifies multiple susceptibility loci for pancreatic cancer. Nat. Genet. 46, 994–1000 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Johansson, B. B. et al. The role of the carboxyl ester lipase (CEL) gene in pancreatic disease. Pancreatology 18, 12–19 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Bulik-Sullivan, B. et al. An atlas of genetic correlations across human diseases and traits. Nat. Genet. 47, 1236–1241 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. 1000 Genomes Project Consortium A global reference for human genetic variation. Nature 526, 68–74 (2015).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhou, W. et al. Efficiently controlling for case–control imbalance and sample relatedness in large-scale genetic association studies. Nat. Genet. 50, 1335–1341 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bulik-Sullivan, B. K. et al. LD score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat. Genet. 47, 291–295 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Evangelou, M. et al. A method for gene-based pathway analysis using genomewide association study summary statistics reveals nine new type 1 diabetes associations. Genet. Epidemiol. 38, 661–670 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Ji, S.-G. et al. Genome-wide association study of primary sclerosing cholangitis identifies new risk loci and quantifies the genetic relationship with inflammatory bowel disease. Nat. Genet. 49, 269–273 (2017).

    Article  MathSciNet  CAS  PubMed  Google Scholar 

  37. Bentham, J. et al. Genetic association analyses implicate aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus. Nat. Genet. 47, 1457–1464 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Cordell, H. J. et al. International genome-wide meta-analysis identifies new primary biliary cirrhosis risk loci and targetable pathogenic pathways. Nat. Commun. 6, 8019 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  40. de Lange, K. M. et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat. Genet. 49, 256–261 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Dubois, P. C. A. et al. Multiple common variants for celiac disease influencing immune gene expression. Nat. Genet. 42, 295–302 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Jin, Y. et al. Genome-wide association studies of autoimmune vitiligo identify 23 new risk loci and highlight key pathways and regulatory variants. Nat. Genet. 48, 1418–1424 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Paternoster, L. et al. Multi-ancestry genome-wide association study of 21,000 cases and 95,000 controls identifies new risk loci for atopic dermatitis. Nat. Genet. 47, 1449–1456 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  44. López-Isac, E. et al. GWAS for systemic sclerosis identifies multiple risk loci and highlights fibrotic and vasculopathy pathways. Nat. Commun. 10, 4955 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  45. 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  PubMed  PubMed Central  Google Scholar 

  46. Mahajan, A. et al. Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nat. Genet. 50, 1505–1513 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. 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  PubMed  Google Scholar 

  48. Stahl, E. A. et al. Genome-wide association study identifies 30 loci associated with bipolar disorder. Nat. Genet. 51, 793–803 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wray, N. R. et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat. Genet. 50, 668–681 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Grove, J. et al. Identification of common genetic risk variants for autism spectrum disorder. Nat. Genet. 51, 431–444 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Watson, H. J. et al. Genome-wide association study identifies eight risk loci and implicates metabo-psychiatric origins for anorexia nervosa. Nat. Genet. 51, 1207–1214 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature 511, 421–427 (2014).

    Article  ADS  PubMed Central  Google Scholar 

  53. Wuttke, M. et al. A catalog of genetic loci associated with kidney function from analyses of a million individuals. Nat. Genet. 51, 957–972 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Nielsen, J. B. et al. Biobank-driven genomic discovery yields new insight into atrial fibrillation biology. Nat. Genet. 50, 1234–1239 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Tachmazidou, I. et al. Identification of new therapeutic targets for osteoarthritis through genome-wide analyses of UK Biobank data. Nat. Genet. 51, 230–236 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Wheeler, E. et al. Impact of common genetic determinants of hemoglobin A1c on type 2 diabetes risk and diagnosis in ancestrally diverse populations: a transethnic genome-wide meta-analysis. PLoS Med. 14, e1002383 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Horikoshi, M. et al. Genome-wide associations for birth weight and correlations with adult disease. Nature 538, 248–252 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Yengo, L. et al. Meta-analysis of genome-wide association studies for height and body mass index in 700000 individuals of European ancestry. Hum. Mol. Genet. 27, 3641–3649 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Jiang, X. et al. Genome-wide association study in 79,366 European-ancestry individuals informs the genetic architecture of 25-hydroxyvitamin D levels. Nat. Commun. 9, 260 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  60. Manning, A. K. et al. A genome-wide approach accounting for body mass index identifies genetic variants influencing fasting glycemic traits and insulin resistance. Nat. Genet. 44, 659–669 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Day, F. R. et al. Large-scale genomic analyses link reproductive aging to hypothalamic signaling, breast cancer susceptibility and BRCA1-mediated DNA repair. Nat. Genet. 47, 1294–1303 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Day, F. R. et al. Genomic analyses identify hundreds of variants associated with age at menarche and support a role for puberty timing in cancer risk. Nat. Genet. 49, 834–841 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Savage, J. E. et al. Genome-wide association meta-analysis in 269,867 individuals identifies new genetic and functional links to intelligence. Nat. Genet. 50, 912–919 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Strawbridge, R. J. et al. Genome-wide association identifies nine common variants associated with fasting proinsulin levels and provides new insights into the pathophysiology of type 2 diabetes. Diabetes 60, 2624–2634 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Saxena, R. et al. Genetic variation in GIPR influences the glucose and insulin responses to an oral glucose challenge. Nat. Genet. 42, 142–148 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Tobacco and Genetics Consortium. Genome-wide meta-analyses identify multiple loci associated with smoking behavior. Nat. Genet. 42, 441–447 (2010).

    Article  Google Scholar 

  67. Shungin, D. et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature 518, 187–196 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Cousminer, D. L. et al. Genome-wide association and longitudinal analyses reveal genetic loci linking pubertal height growth, pubertal timing and childhood adiposity. Hum. Mol. Genet. 22, 2735–2747 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Taal, H. R. et al. Common variants at 12q15 and 12q24 are associated with infant head circumference. Nat. Genet. 44, 532–538 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Teumer, A. et al. Genome-wide analyses identify a role for SLC17A4 and AADAT in thyroid hormone regulation. Nat. Commun. 9, 4455 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  71. Jansen, P. R. et al. Genome-wide analysis of insomnia in 1,331,010 individuals identifies new risk loci and functional pathways. Nat. Genet. 51, 394–403 (2019).

    Article  CAS  PubMed  Google Scholar 

  72. van der Valk, R. J. P. et al. A novel common variant in DCST2 is associated with length in early life and height in adulthood. Hum. Mol. Genet. 24, 1155–1168 (2015).

    Article  PubMed  Google Scholar 

  73. Willer, C. J. et al. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 45, 1274–1283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Felix, J. F. et al. Genome-wide association analysis identifies three new susceptibility loci for childhood body mass index. Hum. Mol. Genet. 25, 389–403 (2016).

    Article  CAS  PubMed  Google Scholar 

  75. Chiou, J. et al. Single cell chromatin accessibility identifies pancreatic islet cell type- and state-specific regulatory programs of diabetes risk. Nat. Genet. 53, 455–466 (2021).

    Article  CAS  PubMed  Google Scholar 

  76. Preissl, S. et al. Single-nucleus analysis of accessible chromatin in developing mouse forebrain reveals cell-type-specific transcriptional regulation. Nat. Neurosci. 21, 432–439 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Cusanovich, D. A. et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  80. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  83. Franzén, O., Gan, L.-M. & Björkegren, J. L. M. PanglaoDB: a web server for exploration of mouse and human single-cell RNA sequencing data. Database 2019, baz046 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Xin, Y. et al. Pseudotime ordering of single human β-cells reveals states of insulin production and unfolded protein response. Diabetes 67, 1783–1794 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  86. Amemiya, H. M., Kundaje, A. & Boyle, A. P. The ENCODE blacklist: identification of problematic regions of the genome. Sci. Rep. 9, 9354 (2019).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  87. 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 

  88. Arda, H. E. et al. A chromatin basis for cell lineage and disease risk in the human pancreas. Cell Syst. 7, 310–322 (2018).

    Article  CAS  PubMed  Google Scholar 

  89. 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 

  90. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Khan, A. et al. JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 46, D260–D266 (2018).

    Article  CAS  PubMed  Google Scholar 

  93. Wingender, E., Schoeps, T., Haubrock, M. & Dönitz, J. TFClass: a classification of human transcription factors and their rodent orthologs. Nucleic Acids Res. 43, D97–D102 (2015).

    Article  CAS  PubMed  Google Scholar 

  94. Pliner, H. A. et al. Cicero predicts cis-regulatory DNA interactions from single-cell chromatin accessibility data. Mol. Cell 71, 858–871 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Harrow, J. et al. GENCODE: the reference human genome annotation for The ENCODE Project. Genome Res. 22, 1760–1774 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. 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 

  97. Cusanovich, D. A. et al. A single-cell atlas of in vivo mammalian chromatin accessibility. Cell 174, 1309–1324 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Wakefield, J. Bayes factors for genome-wide association studies: comparison with P-values. Genet. Epidemiol. 33, 79–86 (2009).

    Article  PubMed  Google Scholar 

  99. 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  PubMed  PubMed Central  Google Scholar 

  100. Namkung, W. et al. Ca2+ activates cystic fibrosis transmembrane conductance regulator- and Cl-dependent HCO3− transport in pancreatic duct cells. J. Biol. Chem. 278, 200–207 (2003).

    Article  CAS  PubMed  Google Scholar 

  101. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR–Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Hsu, P. D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Horlbeck, M. A. et al. Compact and highly active next-generation libraries for CRISPR-mediated gene repression and activation. eLife 5, e19760 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  105. Hormozdiari, F. et al. Colocalization of GWAS and eQTL signals detects target genes. Am. J. Hum. Genet. 99, 1245–1260 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Wang, X., Park, J., Susztak, K., Zhang, N. R. & Li, M. Bulk tissue cell type deconvolution with multi-subject single-cell expression reference. Nat. Commun. 10, 380 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH grants DK112155, DK120429 and DK122607 to K.J.G. and M.S., and T32 GM008666 to R.J.G. We thank S. Kuan for assistance with sequencing. Additional acknowledgements for each cohort are listed in the Supplementary Information.

Author information

Authors and Affiliations

Authors

Contributions

K.J.G. and J.C. designed the study and wrote the manuscript. J.C. performed genetic association and single-cell genomics analyses. R.J.G. performed molecular experiments on enhancer function. M.-L.O. and S. Huang performed molecular experiments on variant function. R.M. and E.B. contributed to analyses of single-cell gene expression. J.Y.H. and M.M. generated single-cell accessible chromatin data. P.B. and K.K. contributed to single-cell motif enrichment analysis. D.U.G. and S.P. supervised the generation of single-cell accessible chromatin data and contributed to data interpretation and analyses. M.S. supervised experiments related to enhancer function and contributed to data interpretation. S. Heller and A.K. contributed to the design and interpretation of enhancer experiments.

Corresponding authors

Correspondence to Joshua Chiou or Kyle J. Gaulton.

Ethics declarations

Competing interests

K.J.G is a consultant for Genentech and holds stock in Vertex Pharmaceuticals; neither is related to the work in this study. The other authors declare no competing interests.

Additional information

Peer review information Nature thanks Jason Torres, Anna Hutchinson, Chris Wallace and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data figures and tables

Extended Data Fig. 1 Independent association signals at T1D risk loci.

Bayes factors (natural log-transformed) for independent association signals at the known PTPN2 locus (left) and the novel BCL11A locus (right). Variants are coloured by linkage disequilibrium (r2) with the index variant for each signal.

Extended Data Fig. 2 Rare variants with large effects on T1D risk.

a, The relationship between minor allele frequency and T1D odds ratios (OR) for index variants at 136 T1D signals. Signals with common index variants and larger effect size estimates (PTPN22 1:114,377,568 A:G and INS 11:2,182,224 A:T) or rare index variants (MAF < 0.005) are labelled. Points and lines represent estimates for OR and 95% CI. b, Comparison of OR across cohorts for rare variants. Missing values indicate that the variant was not tested in the cohort. Points and lines represent estimates for OR and 95% CI.

Extended Data Fig. 3 Genetic correlations between T1D and other traits.

Genetic correlations between T1D and immune-related diseases (left), other diseases (middle), and non-disease traits (right); adj., adjusted; circ., circumference. Two-sided P values are adjusted for multiple comparisons with false discovery rate (FDR). Colours indicate significance: red, correlation is significant after FDR correction (FDR < 0.1); black, correlation is nominally significant (P < 0.05) but not significant after FDR correction; grey, correlation is not significant. Points and lines represent genetic correlation estimates and 95% CI.

Extended Data Fig. 4 Annotations derived from single-cell chromatin accessibility of T1D-relevant tissues.

a, Relative gene accessibility (column-normalized chromatin accessibility reads in gene bodies) showing examples of marker genes used to identify cluster labels. Aggregated chromatin accessibility profiles in a 50-kb window around selected marker genes (bottom). b, Single-cell motif enrichment z-scores (left) and expression of motif subfamily members (right) for examples of TFs with lineage-, cell-type-, or cell-state-specific motif enrichment and expression. TFs with matching motif enrichment and expression are highlighted. c, Co-accessibility between AQP1 and cCREs in ductal cells (left) and CEL and cCREs in acinar cells (right).

Extended Data Fig. 5 Single-cell RNA-seq reference map of PBMCs and pancreatic islets.

a, Clustering of 90,495 expression profiles from scRNA-seq experiments of peripheral blood mononuclear cells and pancreatic islets from published studies. Cells are plotted on the first two UMAP components and coloured according to cluster assignment. The number of cells in each cluster is shown next to its corresponding label. HSC, haematopoietic stem cell; γδ T, gamma delta T cell; pDC, plasmacytoid dendritic cell. b, Relative gene expression (average expression for all cells within a cluster and scaled from 0–100 across clusters) showing examples of marker genes used to assign cluster labels. c, Pearson correlation coefficient between gene expression and promoter accessibility specificity scores using a list containing the top 100 most specific genes for each scRNA-seq cluster found in snATAC–seq.

Extended Data Fig. 6 GWAS enrichment for T1D compared to other diseases and traits.

Stratified LD score regression coefficient z-scores for autoimmune and inflammatory diseases (top), other diseases (middle), and non-disease quantitative endophenotypes (bottom) for cCREs that are active in immune and pancreatic cell types. Two sided P values were calculated from z-scores and multiple test correction was performed using FDR. ***FDR < 0.001, **FDR < 0.01, *FDR < 0.1.

Source data

Extended Data Fig. 7 Fine-mapped variants linked to exocrine-specific genes.

a, The GP2 locus contains three variants in a distal cCRE that is co-accessible with the GP2 promoter in acinar cells, which account for the majority of the causal probability (cPPA = 0.98). Chromatin accessibility at both the distal cCRE and the GP2 promoter is highly specific to acinar cells. b, Variant rs72802342 at the CTRB1/2/BCAR1 locus overlaps a distal cCRE that is co-accessible with the CTRB2 and CTRB1 promoters in acinar cells. Chromatin accessibility at the CTRB1 and CTRB2 promoters is highly specific to acinar cells. Variants contained in the 99% credible set are circled in black.

Extended Data Fig. 8 rs7795896 has allelic effects on ductal enhancer activity.

a, Relative luciferase units (RLU) for reporter containing 594-bp sequence surrounding rs7795896 in Capan-1 cells (n = 6; 2 batches × 3 transfections). Centre line, median; box limits, 25th and 75th percentiles; whiskers extend to 1.5 × the interquartile range from the 25th and 75th percentiles. P value by two-sided, two-way ANOVA. b, Luciferase reporter assay in Capan-1 cells transfected with pGL4.23 minimal promoter plasmids containing rs7795896 in the forward orientation. Relative luciferase units (RLU) represent Firefly:Renilla ratios normalized to control cells transfected with the empty pGL4.23 vector. P value by two-sided Student’s t-test. c, Electrophoretic mobility shift assay with nuclear extract from Capan-1 cells using probes for rs7795896 alleles, with or without 200× unlabelled competitor probe (200× comp.). Quantification of the bound fraction (specific binding/free probe). Data are from n = 1 experiment. d, rs7795896 overlaps histone marks of active enhancers (H3K4me1, H3K27ac; region: chr7:117,050,000–117,125,000, hg19) but not promoters (H3K4me3) in pancreatic ductal adenocarcinoma (PDAC) cell lines (Capan-1, Capan-2, and CFPAC-1). rs7795896 overlaps a ChIP–seq peak for the transcription factor HNF1B in CFPAC-1 cells and a predicted HNF1B motif. e, Relative expression for genes in a 2-Mb window around rs7795896 with non-zero expression and the puromycin resistance and dCas9 genes. Ctrl, control, n = 3 biological replicates; Enh, enhancer, n = 9, 3 sgRNAs × 3 biological replicates; Prom, promoter, n = 3 biological replicates. Data are mean ± 95% CI. P values by two-sided Student’s t-test (Prom versus Ctrl) or two-sided ANOVA (Enh versus Ctrl); NS, not significant.

Source data

Extended Data Fig. 9 rs7795896 affects CFTR expression levels in ductal cells.

a, Bayesian colocalization of T1D signal and CFTR pancreas eQTL. Variants in the T1D credible set are circled. b, Expression of pancreatic cell type marker genes from scRNA-seq. c, Proportions of selected pancreatic cell types estimated by MuSiC for 220 bulk pancreas RNA-seq samples from the GTEx v7 release using single-cell expression profiles. d, −log10-transformed two-sided uncorrected P values from linear regression interaction between dosage and cell-type proportion for the CFTR pancreas eQTL.

Extended Data Fig. 10 Relationship between T1D and other pancreatic diseases.

a, rs7795896 GWAS association for T1D (from full meta-analysis), pancreatic disease, and autoimmune disease. Points and lines represent OR estimates and 95% CI. Two-sided P values from GWAS meta-analysis are unadjusted for multiple comparisons. b, Variants that regulate genes with specialized exocrine pancreas function influence T1D risk, and we hypothesize that these effects are mediated through inflammation and immune infiltration.

Supplementary information

Supplementary Information

This file contains Supplementary Figs. 1-9 and the Supplementary Note.

Reporting Summary

Supplementary Tables

This file contains Supplementary Tables 1-11.

Supplementary Data 1

This file contains the 99% credible sets for 136 independent T1D risk signals.

Supplementary Data 2

This file contains cell type-resolved TPM-normalized gene expression for pancreatic and immune cell types.

Supplementary Data 3

This file contains the catalog of 448,142 cCREs.

Supplementary Data 4

This file contains the subset of 25,436 cCREs elements with the most cell type-specific accessibility.

Supplementary Data 5

This file contains cell type-resolved chromVAR motif enrichment z-scores.

Supplementary Data 6

This file contains co-accessibility links between distal cCREs and gene promoters.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chiou, J., Geusz, R.J., Okino, ML. et al. Interpreting type 1 diabetes risk with genetics and single-cell epigenomics. Nature 594, 398–402 (2021). https://doi.org/10.1038/s41586-021-03552-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-03552-w

This article is cited by

Comments

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

Search

Quick links

Nature Briefing

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

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