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.

  • Review
  • Published:

Pathway paradigms revealed from the genetics of inflammatory bowel disease

Abstract

Inflammatory bowel disease (IBD) is a complex genetic disease that is instigated and amplified by the confluence of multiple genetic and environmental variables that perturb the immune–microbiome axis. The challenge of dissecting pathological mechanisms underlying IBD has led to the development of transformative approaches in human genetics and functional genomics. Here we describe IBD as a model disease in the context of leveraging human genetics to dissect interactions in cellular and molecular pathways that regulate homeostasis of the mucosal immune system. Finally, we synthesize emerging insights from multiple experimental approaches into pathway paradigms and discuss future prospects for disease-subtype classification and therapeutic intervention.

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: Strategies in human genetics and functional genomics to dissect mechanisms of disease.
Fig. 2: IBD genes and pathways controlling mucosal immunity.
Fig. 3: Pathway paradigms highlighted by IBD genetics.
Fig. 4: Coding variants associated with a phenotypic spectrum across different diseases.

Similar content being viewed by others

References

  1. Xavier, R. J. & Podolsky, D. K. Unravelling the pathogenesis of inflammatory bowel disease. Nature 448, 427–434 (2007).

    ADS  CAS  PubMed  Google Scholar 

  2. Kaplan, G. G. & Ng, S. C. Understanding and preventing the global increase of inflammatory bowel disease. Gastroenterology 152, 313–321.e2 (2017).

    PubMed  Google Scholar 

  3. Lloyd-Price, J. et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. McKinney, E. F., Lee, J. C., Jayne, D. R. W., Lyons, P. A. & Smith, K. G. C. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 523, 612–616 (2015).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hyams, J. S. et al. Clinical and biological predictors of response to standardised paediatric colitis therapy (PROTECT): a multicentre inception cohort study. Lancet 393, 1708–1720 (2019).

    PubMed  PubMed Central  Google Scholar 

  6. Belarif, L. et al. IL-7 receptor influences anti-TNF responsiveness and T cell gut homing in inflammatory bowel disease. J. Clin. Invest. 129, 1910–1925 (2019).

    PubMed  PubMed Central  Google Scholar 

  7. Martins, F. et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat. Rev. Clin. Oncol. 16, 563–580 (2019).

    CAS  PubMed  Google Scholar 

  8. Emond, B., Ellis, L. A., Chakravarty, S. D., Ladouceur, M. & Lefebvre, P. Real-world incidence of inflammatory bowel disease among patients with other chronic inflammatory diseases treated with interleukin-17a or phosphodiesterase 4 inhibitors. Curr. Med. Res. Opin. 35, 1751–1759 (2019).

    CAS  PubMed  Google Scholar 

  9. Jostins, L. et al. Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012). The Immunochip GWAS identified several new loci associated with IBD risk and offered insights into underlying pathways driving disease risk.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Liu, J. Z. et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet. 47, 979–986 (2015). This GWAS analysed individuals across diverse ancestries, with large cohorts, and collectively implicated new loci associated with IBD.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. 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). This GWAS and meta-analysis captured approximately 240 loci associated with IBD.

    PubMed  PubMed Central  Google Scholar 

  12. Huang, H. et al. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature 547, 173–178 (2017). This fine-mapping GWAS implicates putative causal SNPs associated with a number of IBD risk loci.

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dendrou, C. A. et al. Resolving TYK2 locus genotype-to-phenotype differences in autoimmunity. Sci. Transl. Med. 8, 363ra149 (2016).

    PubMed  PubMed Central  Google Scholar 

  14. Momozawa, Y. et al. IBD risk loci are enriched in multigenic regulatory modules encompassing putative causative genes. Nat. Commun. 9, 2427 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Finucane, H. K. et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621–629 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Rivas, M. A. et al. Insights into the genetic epidemiology of Crohn’s and rare diseases in the Ashkenazi Jewish population. PLoS Genet. 14, e1007329 (2018). This exome-sequencing study identified coding variants associated with IBD risk and leveraged ancestry to identify rare strong-acting genetic variants.

    PubMed  PubMed Central  Google Scholar 

  18. Arnadottir, G. A. et al. A homozygous loss-of-function mutation leading to CYBC1 deficiency causes chronic granulomatous disease. Nat. Commun. 9, 4447 (2018).

    ADS  PubMed  PubMed Central  Google Scholar 

  19. Rivas, M. A. et al. A protein-truncating R179X variant in RNF186 confers protection against ulcerative colitis. Nat. Commun. 7, 12342 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Rivas, M. A. et al. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nat. Genet. 43, 1066–1073 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Beaudoin, M. et al. Deep resequencing of GWAS loci identifies rare variants in CARD9, IL23R and RNF186 that are associated with ulcerative colitis. PLoS Genet. 9, e1003723 (2013). Two papers 20,21 describing exome-sequencing studies identifying risk and protective coding variants associated with IBD.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Glocker, E.-O. et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N. Engl. J. Med. 361, 2033–2045 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Shouval, D. S. et al. Interleukin-10 receptor signaling in innate immune cells regulates mucosal immune tolerance and anti-inflammatory macrophage function. Immunity 40, 706–719 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Zigmond, E. et al. Macrophage-restricted interleukin-10 receptor deficiency, but not IL-10 deficiency, causes severe spontaneous colitis. Immunity 40, 720–733 (2014).

    CAS  PubMed  Google Scholar 

  25. Bernshtein, B. et al. IL-23-producing IL-10Rα-deficient gut macrophages elicit an IL-22-driven proinflammatory epithelial cell response. Sci. Immunol. 4, eaau6571 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Hartono, S., Ippoliti, M. R., Mastroianni, M., Torres, R. & Rider, N. L. Gastrointestinal disorders associated with primary immunodeficiency diseases. Clin. Rev. Allergy Immunol. 57, 145–165 (2019).

    PubMed  Google Scholar 

  27. Frizinsky, S. et al. Novel MALT1 mutation linked to immunodeficiency, immune dysregulation, and an abnormal T cell receptor repertoire. J. Clin. Immunol. 39, 401–413 (2019).

    CAS  PubMed  Google Scholar 

  28. Denson, L. A. et al. Clinical and genomic correlates of neutrophil reactive oxygen species production in pediatric patients with Crohn’s disease. Gastroenterology 154, 2097–2110 (2018).

    CAS  PubMed  Google Scholar 

  29. Schwerd, T. et al. Impaired antibacterial autophagy links granulomatous intestinal inflammation in Niemann–Pick disease type C1 and XIAP deficiency with NOD2 variants in Crohn’s disease. Gut 66, 1060–1073 (2017).

    CAS  PubMed  Google Scholar 

  30. Charbit-Henrion, F. et al. Diagnostic yield of next-generation sequencing in very early-onset inflammatory bowel diseases: a multicentre study. J. Crohn’s Colitis 12, 1104–1112 (2018).

    Google Scholar 

  31. Jardine, S., Dhingani, N. & Muise, A. M. TTC7A: steward of intestinal health. Cell. Mol. Gastroenterol. Hepatol. 7, 555–570 (2019).

    PubMed  Google Scholar 

  32. Holt-Danborg, L. et al. SPINT2 (HAI-2) missense variants identified in congenital sodium diarrhea/tufting enteropathy affect the ability of HAI-2 to inhibit prostasin but not matriptase. Hum. Mol. Genet. 28, 828–841 (2019).

    CAS  PubMed  Google Scholar 

  33. Tronstad, R. R. et al. Genetic and transcriptional analysis of inflammatory bowel disease-associated pathways in patients with GUCY2C-linked familial diarrhea. Scand. J. Gastroenterol. 53, 1264–1273 (2018).

    CAS  PubMed  Google Scholar 

  34. Lassen, K. G. et al. Genetic coding variant in GPR65 alters lysosomal pH and links lysosomal dysfunction with colitis risk. Immunity 44, 1392–1405 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Graham, D. B. et al. Functional genomics identifies negative regulatory nodes controlling phagocyte oxidative burst. Nat. Commun. 6, 7838 (2015).

    ADS  CAS  PubMed  Google Scholar 

  36. Gaublomme, J. T. et al. Single-cell genomics unveils critical regulators of Th17 cell pathogenicity. Cell 163, 1400–1412 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Graham, D. B. et al. Nitric oxide engages an anti-inflammatory feedback loop mediated by peroxiredoxin 5 in phagocytes. Cell Rep. 24, 838–850 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  40. Takeda, H. et al. CRISPR–Cas9-mediated gene knockout in intestinal tumor organoids provides functional validation for colorectal cancer driver genes. Proc. Natl Acad. Sci. USA 116, 15635–15644 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Biton, M. et al. T helper cell cytokines modulate intestinal stem cell renewal and differentiation. Cell 175, 1307–1320.e22 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Bein, A. et al. Microfluidic organ-on-a-chip models of human intestine. Cell. Mol. Gastroenterol. Hepatol. 5, 659–668 (2018).

    PubMed  PubMed Central  Google Scholar 

  43. Parikh, K. et al. Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature 567, 49–55 (2019).

    ADS  CAS  PubMed  Google Scholar 

  44. Kinchen, J. et al. Structural remodeling of the human colonic mesenchyme in inflammatory bowel disease. Cell 175, 372–386.e17 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Smillie, C. S. et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell 178, 714–730.e22 (2019).References 43,45 provide a holistic single-cell transcriptional map of UC.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Martin, J. C. et al. Single-cell analysis of Crohn’s disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy. Cell 178, 1493–1508.e20 (2019). This study provides a single cell transcriptional map of CD.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Haber, A. L. et al. A single-cell survey of the small intestinal epithelium. Nature 551, 333–339 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. West, N. R. et al. Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease. Nat. Med. 23, 579–589 (2017). This study uses transcriptomic signatures of treatment resistance to identify and functionally validate cytokine circuits associated with severe IBD.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Ellinghaus, D. et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat. Genet. 48, 510–518 (2016). This study compares GWAS risk loci across distinct inflammatory diseases to identify common and disease-specific genes.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Metidji, A. et al. The environmental sensor AHR protects from inflammatory damage by maintaining intestinal stem cell homeostasis and barrier integrity. Immunity 49, 353–362.e5 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Pentinmikko, N. et al. Notum produced by Paneth cells attenuates regeneration of aged intestinal epithelium. Nature 571, 398–402 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Serra, D. et al. Self-organization and symmetry breaking in intestinal organoid development. Nature 569, 66–72 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  53. VanDussen, K. L. et al. Abnormal small intestinal epithelial microvilli in patients with Crohn’s disease. Gastroenterology 155, 815–828 (2018).

    PubMed  Google Scholar 

  54. Ramakrishnan, S. K. et al. Intestinal non-canonical NFκB signaling shapes the local and systemic immune response. Nat. Commun. 10, 660 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  55. Schneider, C., O’Leary, C. E. & Locksley, R. M. Regulation of immune responses by tuft cells. Nat. Rev. Immunol. 19, 584–593 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Grün, D. et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature 525, 251–255 (2015).

    ADS  PubMed  Google Scholar 

  57. Beumer, J. et al. Enteroendocrine cells switch hormone expression along the crypt-to-villus BMP signalling gradient. Nat. Cell Biol. 20, 909–916 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Gehart, H. et al. Identification of enteroendocrine regulators by real-time single-cell differentiation mapping. Cell 176, 1158–1173.e16 (2019).

    CAS  PubMed  Google Scholar 

  59. Mohanan, V. et al. C1orf106 is a colitis risk gene that regulates stability of epithelial adherens junctions. Science 359, 1161–1166 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  60. Manzanillo, P. et al. Inflammatory bowel disease susceptibility gene C1ORF106 regulates intestinal epithelial permeability. Immunohorizons 2, 164–171 (2018).

    CAS  PubMed  Google Scholar 

  61. Fujimoto, K. et al. Regulation of intestinal homeostasis by the ulcerative colitis-associated gene RNF186. Mucosal Immunol. 10, 446–459 (2017).

    CAS  PubMed  Google Scholar 

  62. Tong, X. et al. RNF186 impairs insulin sensitivity by inducing ER stress in mouse primary hepatocytes. Cell. Signal. 52, 155–162 (2018).

    CAS  PubMed  Google Scholar 

  63. Sveinbjornsson, G. et al. Rare mutations associating with serum creatinine and chronic kidney disease. Hum. Mol. Genet. 23, 6935–6943 (2014).

    CAS  PubMed  Google Scholar 

  64. Mukherjee, T. et al. NOD1 and NOD2 in inflammation, immunity and disease. Arch. Biochem. Biophys. 670, 69–81 (2019).

    CAS  PubMed  Google Scholar 

  65. Ogura, Y. et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature 411, 603–606 (2001).

    ADS  CAS  PubMed  Google Scholar 

  66. Hsu, L.-C. et al. A NOD2–NALP1 complex mediates caspase-1-dependent IL-1β secretion in response to Bacillus anthracis infection and muramyl dipeptide. Proc. Natl Acad. Sci. USA 105, 7803–7808 (2008).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  67. Martinon, F., Agostini, L., Meylan, E. & Tschopp, J. Identification of bacterial muramyl dipeptide as activator of the NALP3/cryopyrin inflammasome. Curr. Biol. 14, 1929–1934 (2004).

    CAS  PubMed  Google Scholar 

  68. Kim, Y.-G. et al. Cutting edge: Crohn’s disease-associated Nod2 mutation limits production of proinflammatory cytokines to protect the host from Enterococcus faecalis-induced lethality. J. Immunol. 187, 2849–2852 (2011).

    CAS  PubMed  Google Scholar 

  69. Umiker, B. et al. The NLRP3 inflammasome mediates DSS-induced intestinal inflammation in Nod2 knockout mice. Innate Immun. 25, 132–143 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Caruso, R. et al. A specific gene–microbe interaction drives the development of Crohn’s disease-like colitis in mice. Sci. Immunol. 4, eaaw4341 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Chirieleison, S. M. et al. Nucleotide-binding oligomerization domain (NOD) signaling defects and cell death susceptibility cannot be uncoupled in X-linked inhibitor of apoptosis (XIAP)-driven inflammatory disease. J. Biol. Chem. 292, 9666–9679 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Latour, S. & Aguilar, C. XIAP deficiency syndrome in humans. Semin. Cell Dev. Biol. 39, 115–123 (2015).

    CAS  PubMed  Google Scholar 

  73. Li, Q. et al. Variants in TRIM22 that affect NOD2 signaling are associated with very-early-onset inflammatory bowel disease. Gastroenterology 150, 1196–1207 (2016).

    CAS  PubMed  Google Scholar 

  74. Zammit, N. W. et al. Denisovan, modern human and mouse TNFAIP3 alleles tune A20 phosphorylation and immunity. Nat. Immunol. 20, 1299–1310 (2019).

    CAS  PubMed  Google Scholar 

  75. Hrdinka, M. et al. Small molecule inhibitors reveal an indispensable scaffolding role of RIPK2 in NOD2 signaling. EMBO J. 37, e99372 (2018).

    PubMed  PubMed Central  Google Scholar 

  76. Haile, P. A. et al. Discovery of a first-in-class receptor interacting protein 2 (RIP2) kinase specific clinical candidate, 2-((4-(benzo[d]thiazol-5-ylamino)-6-(tert-butylsulfonyl)quinazolin-7-yl)oxy)ethyl dihydrogen phosphate, for the treatment of inflammatory diseases. J. Med. Chem. 62, 6482–6494 (2019).

    CAS  PubMed  Google Scholar 

  77. Hara, H. et al. The adaptor protein CARD9 is essential for the activation of myeloid cells through ITAM-associated and Toll-like receptors. Nat. Immunol. 8, 619–629 (2007).

    CAS  PubMed  Google Scholar 

  78. Xu, X. et al. CARD9S12N facilitates the production of IL-5 by alveolar macrophages for the induction of type 2 immune responses. Nat. Immunol. 19, 547–560 (2018).

    CAS  PubMed  Google Scholar 

  79. Limon, J. J. et al. Malassezia is associated with Crohn’s disease and exacerbates colitis in mouse models. Cell Host Microbe 25, 377–388.e6 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Cao, Z. et al. Ubiquitin ligase TRIM62 regulates CARD9-mediated anti-fungal immunity and intestinal inflammation. Immunity 43, 715–726 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Leshchiner, E. S. et al. Small-molecule inhibitors directly target CARD9 and mimic its protective variant in inflammatory bowel disease. Proc. Natl Acad. Sci. USA 114, 11392–11397 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Bunker, J. J. & Bendelac, A. IgA responses to microbiota. Immunity 49, 211–224 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Castro-Dopico, T. et al. Anti-commensal IgG drives intestinal inflammation and type 17 immunity in ulcerative colitis. Immunity 50, 1099–1114.e10 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Plichta, D. R., Graham, D. B., Subramanian, S. & Xavier, R. J. Therapeutic opportunities in inflammatory bowel disease: mechanistic dissection of host–microbiome relationships. Cell 178, 1041–1056 (2019). This review covers the role of the microbiome and environmental factors that affect IBD.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Goyette, P. et al. High-density mapping of the MHC identifies a shared role for HLA-DRB1*01:03 in inflammatory bowel diseases and heterozygous advantage in ulcerative colitis. Nat. Genet. 47, 172–179 (2015). This fine-mapping GWAS implicated specific HLA alleles in IBD risk.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Lee, J. C. et al. Genome-wide association study identifies distinct genetic contributions to prognosis and susceptibility in Crohn’s disease. Nat. Genet. 49, 262–268 (2017).

    PubMed  PubMed Central  Google Scholar 

  87. Knoop, K. A. et al. Microbial antigen encounter during a preweaning interval is critical for tolerance to gut bacteria. Sci. Immunol. 2, eaao1314 (2017).

    PubMed  PubMed Central  Google Scholar 

  88. Wu, J. et al. Expanded TCRβ CDR3 clonotypes distinguish Crohn’s disease and ulcerative colitis patients. Mucosal Immunol. 11, 1487–1495 (2018).

    CAS  PubMed  Google Scholar 

  89. Christophersen, A. et al. Distinct phenotype of CD4+ T cells driving celiac disease identified in multiple autoimmune conditions. Nat. Med. 25, 734–737 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Vivier, E. et al. Innate lymphoid cells: 10 years on. Cell 174, 1054–1066 (2018).

    CAS  PubMed  Google Scholar 

  91. Zhou, L. et al. Innate lymphoid cells support regulatory T cells in the intestine through interleukin-2. Nature 568, 405–409 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  92. Friedrich, M., Pohin, M. & Powrie, F. Cytokine networks in the pathophysiology of inflammatory bowel disease. Immunity 50, 992–1006 (2019).

    CAS  PubMed  Google Scholar 

  93. Duerr, R. H. et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 314, 1461–1463 (2006).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  94. Bauché, D. et al. LAG3+ regulatory T cells restrain interleukin-23-producing CX3CR1+ gut-resident macrophages during group 3 innate lymphoid cell-driven colitis. Immunity 49, 342–352.e5 (2018).

    PubMed  Google Scholar 

  95. Zhou, L. et al. IL-6 programs TH-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat. Immunol. 8, 967–974 (2007).

    CAS  PubMed  Google Scholar 

  96. Maxwell, J. R. et al. Differential roles for interleukin-23 and interleukin-17 in intestinal immunoregulation. Immunity 43, 739–750 (2015).

    CAS  PubMed  Google Scholar 

  97. Tang, C. et al. Suppression of IL-17F, but not of IL-17A, provides protection against colitis by inducing Treg cells through modification of the intestinal microbiota. Nat. Immunol. 19, 755–765 (2018).

    CAS  PubMed  Google Scholar 

  98. Leppkes, M. et al. RORγ-expressing Th17 cells induce murine chronic intestinal inflammation via redundant effects of IL-17A and IL-17F. Gastroenterology 136, 257–267 (2009).

    CAS  PubMed  Google Scholar 

  99. Malik, A. & Kanneganti, T.-D. Inflammasome activation and assembly at a glance. J. Cell Sci. 130, 3955–3963 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Shouval, D. S. et al. Interleukin 1β mediates intestinal inflammation in mice and patients with interleukin 10 receptor deficiency. Gastroenterology 151, 1100–1104 (2016).

    CAS  PubMed  Google Scholar 

  101. de Luca, A. et al. IL-1 receptor blockade restores autophagy and reduces inflammation in chronic granulomatous disease in mice and in humans. Proc. Natl Acad. Sci. USA 111, 3526–3531 (2014).

    ADS  PubMed  PubMed Central  Google Scholar 

  102. Saitoh, T. et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1β production. Nature 456, 264–268 (2008).

    ADS  CAS  PubMed  Google Scholar 

  103. Lassen, K. G. et al. Atg16L1 T300A variant decreases selective autophagy resulting in altered cytokine signaling and decreased antibacterial defense. Proc. Natl Acad. Sci. USA 111, 7741–7746 (2014).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  104. Murthy, A. et al. A Crohn’s disease variant in Atg16l1 enhances its degradation by caspase 3. Nature 506, 456–462 (2014).

    ADS  CAS  PubMed  Google Scholar 

  105. Samie, M. et al. Selective autophagy of the adaptor TRIF regulates innate inflammatory signaling. Nat. Immunol. 19, 246–254 (2018).

    CAS  PubMed  Google Scholar 

  106. Weiss, E. S. et al. Interleukin-18 diagnostically distinguishes and pathogenically promotes human and murine macrophage activation syndrome. Blood 131, 1442–1455 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Xu, H. et al. Innate immune sensing of bacterial modifications of Rho GTPases by the pyrin inflammasome. Nature 513, 237–241 (2014).

    ADS  CAS  PubMed  Google Scholar 

  108. Khare, S. et al. An NLRP7-containing inflammasome mediates recognition of microbial lipopeptides in human macrophages. Immunity 36, 464–476 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Schiering, C. et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 513, 564–568 (2014).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  110. Neill, D. R. et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 464, 1367–1370 (2010).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  111. He, Z. et al. Mast cells are essential intermediaries in regulating IL-33/ST2 signaling for an immune network favorable to mucosal healing in experimentally inflamed colons. Cell Death Dis. 9, 1173 (2018).

    PubMed  PubMed Central  Google Scholar 

  112. Scheibe, K. et al. Inhibiting interleukin 36 receptor signaling reduces fibrosis in mice with chronic intestinal inflammation. Gastroenterology 156, 1082–1097.e11 (2019).

    CAS  PubMed  Google Scholar 

  113. Rioux, J. D. et al. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat. Genet. 39, 596–604 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. McCarroll, S. A. et al. Deletion polymorphism upstream of IRGM associated with altered IRGM expression and Crohn’s disease. Nat. Genet. 40, 1107–1112 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Hampe, J. et al. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat. Genet. 39, 207–211 (2007).

    CAS  PubMed  Google Scholar 

  116. Hui, K. Y. et al. Functional variants in the LRRK2 gene confer shared effects on risk for Crohn’s disease and Parkinson’s disease. Sci. Transl. Med. 10, eaai7795 (2018).

    PubMed  PubMed Central  Google Scholar 

  117. Takagawa, T. et al. An increase in LRRK2 suppresses autophagy and enhances dectin-1-induced immunity in a mouse model of colitis. Sci. Transl. Med. 10, eaan8162 (2018).

    PubMed  PubMed Central  Google Scholar 

  118. Cadwell, K. et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature 456, 259–263 (2008).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  119. VanDussen, K. L. et al. Genetic variants synthesize to produce Paneth cell phenotypes that define subtypes of Crohn’s disease. Gastroenterology 146, 200–209 (2014).

    CAS  PubMed  Google Scholar 

  120. Deuring, J. J. et al. Genomic ATG16L1 risk allele-restricted Paneth cell ER stress in quiescent Crohn’s disease. Gut 63, 1081–1091 (2014).

    CAS  PubMed  Google Scholar 

  121. Grootjans, J. et al. Epithelial endoplasmic reticulum stress orchestrates a protective IgA response. Science 363, 993–998 (2019).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  122. Kaser, A. et al. XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell 134, 743–756 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Adolph, T. E. et al. Paneth cells as a site of origin for intestinal inflammation. Nature 503, 272–276 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  124. Hosomi, S. et al. Intestinal epithelial cell endoplasmic reticulum stress promotes MULT1 up-regulation and NKG2D-mediated inflammation. J. Exp. Med. 214, 2985–2997 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Graham, D. B. et al. TMEM258 is a component of the oligosaccharyltransferase complex controlling ER stress and intestinal inflammation. Cell Rep. 17, 2955–2965 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Park, J. H. et al. SLC39A8 deficiency: a disorder of manganese transport and glycosylation. Am. J. Hum. Genet. 97, 894–903 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Bastaki, F. et al. Single-center experience of N-linked congenital disorders of glycosylation with a summary of molecularly characterized cases in Arabs. Ann. Hum. Genet. 82, 35–47 (2018).

    CAS  PubMed  Google Scholar 

  128. Taylor, C. T. & Colgan, S. P. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat. Rev. Immunol. 17, 774–785 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Campbell, E. L. et al. Transmigrating neutrophils shape the mucosal microenvironment through localized oxygen depletion to influence resolution of inflammation. Immunity 40, 66–77 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Curtis, V. F. et al. Stabilization of HIF through inhibition of cullin-2 neddylation is protective in mucosal inflammatory responses. FASEB J. 29, 208–215 (2015).

    CAS  PubMed  Google Scholar 

  131. Xue, X. et al. Endothelial PAS domain protein 1 activates the inflammatory response in the intestinal epithelium to promote colitis in mice. Gastroenterology 145, 831–841 (2013).

    CAS  PubMed  Google Scholar 

  132. Solanki, S., Devenport, S. N., Ramakrishnan, S. K. & Shah, Y. M. Temporal induction of intestinal epithelial hypoxia-inducible factor-2α is sufficient to drive colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 317, G98–G107 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Glover, L. E. et al. Control of creatine metabolism by HIF is an endogenous mechanism of barrier regulation in colitis. Proc. Natl Acad. Sci. USA 110, 19820–19825 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  134. Tannahill, G. M. et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 496, 238–242 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  135. Nadjsombati, M. S. et al. Detection of succinate by intestinal tuft cells triggers a type 2 innate immune circuit. Immunity 49, 33–41.e7 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Nair, S. et al. Irg1 expression in myeloid cells prevents immunopathology during M. tuberculosis infection. J. Exp. Med. 215, 1035–1045 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Mills, E. L. et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature 556, 113–117 (2018).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  138. Skon-Hegg, C. et al. LACC1 regulates TNF and IL-17 in mouse models of arthritis and inflammation. J. Immunol. 202, 183–193 (2019).

    CAS  PubMed  Google Scholar 

  139. Lahiri, A., Hedl, M., Yan, J. & Abraham, C. Human LACC1 increases innate receptor-induced responses and a LACC1 disease-risk variant modulates these outcomes. Nat. Commun. 8, 15614 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  140. Cader, M. Z. et al. C13orf31 (FAMIN) is a central regulator of immunometabolic function. Nat. Immunol. 17, 1046–1056 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Molofsky, A. B., Savage, A. K. & Locksley, R. M. Interleukin-33 in tissue homeostasis, injury, and inflammation. Immunity 42, 1005–1019 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Schafer, S. et al. IL-11 is a crucial determinant of cardiovascular fibrosis. Nature 552, 110–115 (2017).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  143. Kotlarz, D. et al. Human TGF-β1 deficiency causes severe inflammatory bowel disease and encephalopathy. Nat. Genet. 50, 344–348 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Flynn, R. S. et al. Endogenous IGFBP-3 regulates excess collagen expression in intestinal smooth muscle cells of Crohn’s disease strictures. Inflamm. Bowel Dis. 17, 193–201 (2011).

    PubMed  Google Scholar 

  145. Geesala, R. et al. Loss of RHBDF2 results in an early-onset spontaneous murine colitis. J. Leukoc. Biol. 105, 767–781 (2019).

    CAS  PubMed  Google Scholar 

  146. Li, Y. et al. Human RIPK1 deficiency causes combined immunodeficiency and inflammatory bowel diseases. Proc. Natl Acad. Sci. USA 116, 970–975 (2019).

    CAS  PubMed  Google Scholar 

  147. Pagnini, C., Pizarro, T. T. & Cominelli, F. Novel pharmacological therapy in inflammatory bowel diseases: beyond anti-tumor necrosis factor. Front. Pharmacol. 10, 671 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Ma, C. et al. Innovations in oral therapies for inflammatory bowel disease. Drugs 79, 1321–1335 (2019).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is supported by grants from the Helmsley Charitable Trust and NIH (to R.J.X.). We thank H. Kang for valuable scientific input, editorial assistance and illustrative design.

Author information

Authors and Affiliations

Authors

Contributions

D.B.G. and R.J.X. conceived and wrote the article, and reviewed and edited the manuscript before submission.

Corresponding authors

Correspondence to Daniel B. Graham or Ramnik J. Xavier.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Jeff Barrett 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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Graham, D.B., Xavier, R.J. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature 578, 527–539 (2020). https://doi.org/10.1038/s41586-020-2025-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2025-2

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: 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