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 Article
  • Published:

Redefining the IBDs using genome-scale molecular phenotyping

Abstract

The IBDs, Crohn’s disease and ulcerative colitis, are chronic inflammatory conditions of the gastrointestinal tract resulting from an aberrant immune response to enteric microbiota in genetically susceptible individuals. Disease presentation and progression within and across IBDs, especially Crohn’s disease, are highly heterogeneous in location, severity of inflammation and other phenotypes. Current clinical classifications fail to accurately predict disease course and response to therapies. Genome-wide association studies have identified >240 loci that confer risk of IBD, but the clinical utility of these findings remains unclear, and mechanisms by which the genetic variants contribute to disease are largely unknown. In the past 5 years, the profiling of genome-wide gene expression, epigenomic features and gut microbiota composition in intestinal tissue and faecal samples has uncovered distinct molecular signatures that define IBD subtypes, including within Crohn’s disease and ulcerative colitis. In this Review, we summarize studies in both adult and paediatric patients that have identified different IBD subtypes, which in some cases have been associated with distinct clinical phenotypes. We posit that genome-scale molecular phenotyping in large cohorts holds great promise not only to further our understanding of the diverse molecular causes of IBD but also for improving clinical trial design to develop more personalized disease management and treatment.

Key points

  • The inflammatory bowel diseases, Crohn’s disease and ulcerative colitis, are highly heterogeneous in presentation, disease course and response to therapy.

  • Genome-wide association studies have identified >240 loci associated with IBD, but this knowledge has not yet contributed to improved patient care.

  • Sequencing-based assays enable the identification and quantification of molecular phenotypes, including gene and microRNA expression levels, locations of active regulatory elements and gut microbial composition, that are altered in disease.

  • Genome-wide characterization of tissues and cells from patients with Crohn’s disease has identified specific molecular subtypes that are associated with different clinical phenotypes.

  • Molecular profiling in specific intestinal cell populations is necessary to pinpoint mechanisms that contribute to disease phenotypes, including those caused by genetic variation.

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: Contributing factors to IBD phenotypes.
Fig. 2: Overview of connections between molecular and disease phenotypes.
Fig. 3: Clinical characteristics of Crohn’s disease.

Similar content being viewed by others

References

  1. Sartor, R. B. Mechanisms of disease: pathogenesis of Crohn’s disease and ulcerative colitis. Nat. Clin. Pract. Gastroenterol. Hepatol. 3, 390–407 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Singh, S., Fumery, M., Sandborn, W. J. & Murad, M. H. Systematic review with network meta-analysis: first- and second-line pharmacotherapy for moderate-severe ulcerative colitis. Aliment. Pharmacol. Ther. 47, 162–175 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Allen, P. B. et al. Review article: moving towards common therapeutic goals in Crohn’s disease and rheumatoid arthritis. Aliment. Pharmacol. Ther. 45, 1058–1072 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Abraham, C., Dulai, P. S., Vermeire, S. & Sandborn, W. J. Lessons learned from trials targeting cytokine pathways in patients with inflammatory bowel diseases. Gastroenterology 152, 374–388 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Gerich, M. E. & McGovern, D. P. Towards personalized care in IBD. Nat. Rev. Gastroenterol. Hepatol. 11, 287–299 (2014).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tomczak, K., Czerwinska, P. & Wiznerowicz, M. The Cancer Genome Atlas (TCGA): an immeasurable source of knowledge. Contemp. Oncol. (Pozn) 19, A68–A77 (2015).

    Google Scholar 

  8. Prat, A. & Perou, C. M. Deconstructing the molecular portraits of breast cancer. Mol. Oncol. 5, 5–23 (2011).

    Article  CAS  PubMed  Google Scholar 

  9. Lenz, G. et al. Molecular subtypes of diffuse large B cell lymphoma arise by distinct genetic pathways. Proc. Natl Acad. Sci. USA 105, 13520–13525 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Verhaak, R. G. et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17, 98–110 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cancer Genome Atlas Research Network. et al. Integrated genomic characterization of endometrial carcinoma. Nature 497, 67–73 (2013).

    Article  CAS  Google Scholar 

  12. Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543–550 (2014).

    Article  CAS  Google Scholar 

  13. Satsangi, J., Silverberg, M. S., Vermeire, S. & Colombel, J. F. The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut 55, 749–753 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Panes, J. & Rimola, J. Perianal fistulizing Crohn’s disease: pathogenesis, diagnosis and therapy. Nat. Rev. Gastroenterol. Hepatol. 14, 652–664 (2017).

    Article  PubMed  Google Scholar 

  15. Gecse, K. B. et al. A global consensus on the classification, diagnosis and multidisciplinary treatment of perianal fistulising Crohn’s disease. Gut 63, 1381–1392 (2014).

    Article  PubMed  Google Scholar 

  16. Mohammed Vashist, N. et al. Endoscopic scoring indices for evaluation of disease activity in ulcerative colitis. Cochrane Database Syst. Rev. 1, CD011450 (2018).

    PubMed  Google Scholar 

  17. Fedorak, R. N. Is it time to re-classify Crohn’s disease? Best Pract. Res. Clin. Gastroenterol. 18 (Suppl), 99–106 (2004).

    Article  PubMed  Google Scholar 

  18. Guizzetti, L. et al. Development of clinical prediction models for surgery and complications in Crohn’s disease. J. Crohns Colitis 12, 167–177 (2018).

    Article  PubMed  Google Scholar 

  19. Fumery, M. et al. Systematic review with meta-analysis: recurrence of Crohn’s disease after total colectomy with permanent ileostomy. Aliment. Pharmacol. Ther. 45, 381–390 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Benchimol, E. I. et al. Trends in epidemiology of pediatric inflammatory bowel disease in Canada: distributed network analysis of multiple population-based provincial health administrative databases. Am. J. Gastroenterol. 112, 1120–1134 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Moran, C. J. Very early onset inflammatory bowel disease. Semin. Pediatr. Surg. 26, 356–359 (2017).

    Article  PubMed  Google Scholar 

  22. de Bie, C. I. et al. Disease phenotype at diagnosis in pediatric Crohn’s disease: 5-year analyses of the EUROKIDS Registry. Inflamm. Bowel Dis. 19, 378–385 (2013).

    Article  PubMed  Google Scholar 

  23. Rigoli, L. & Caruso, R. A. Inflammatory bowel disease in pediatric and adolescent patients: a biomolecular and histopathological review. World J. Gastroenterol. 20, 10262–10278 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  24. PROKIIDS Data Centre. RISK Stratification Project. prokiids https://prokiids.com/RISK_Public.html (2019).

  25. Peloquin, J. M., Goel, G., Villablanca, E. J. & Xavier, R. J. Mechanisms of pediatric inflammatory bowel disease. Annu. Rev. Immunol. 34, 31–64 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Dulai, P. S., Singh, S., Ohno-Machado, L. & Sandborn, W. J. Population health management for inflammatory bowel disease. Gastroenterology 154, 37–45 (2018).

    Article  PubMed  Google Scholar 

  27. Levine, A. et al. Pediatric modification of the Montreal classification for inflammatory bowel disease: the Paris classification. Inflamm. Bowel Dis. 17, 1314–1321 (2011).

    Article  PubMed  Google Scholar 

  28. Lovasz, B. D. et al. Evolution of disease phenotype in adult and pediatric onset Crohn’s disease in a population-based cohort. World J. Gastroenterol. 19, 2217–2226 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Gordon, H., Trier Moller, F., Andersen, V. & Harbord, M. Heritability in inflammatory bowel disease: from the first twin study to genome-wide association studies. Inflamm. Bowel Dis. 21, 1428–1434 (2015).

    PubMed  Google Scholar 

  30. Satsangi, J., Grootscholten, C., Holt, H. & Jewell, D. P. Clinical patterns of familial inflammatory bowel disease. Gut 38, 738–741 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mirkov, M. U., Verstockt, B. & Cleynen, I. Genetics of inflammatory bowel disease: beyond NOD2. Lancet Gastroenterol. Hepatol. 2, 224–234 (2017).

    Google Scholar 

  32. Cleynen, I. et al. Inherited determinants of Crohn’s disease and ulcerative colitis phenotypes: a genetic association study. Lancet 387, 156–167 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  33. McGovern, D. P., Kugathasan, S. & Cho, J. H. Genetics of inflammatory bowel diseases. Gastroenterology 149, 1163–1176 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Uhlig, H. H. et al. The diagnostic approach to monogenic very early onset inflammatory bowel disease. Gastroenterology 147, 990–1007 (2014).

    Article  PubMed  Google Scholar 

  36. Kammermeier, J. et al. Targeted gene panel sequencing in children with very early onset inflammatory bowel disease—evaluation and prospective analysis. J. Med. Genet. 51, 748–755 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Uhlig, H. H. Monogenic diseases associated with intestinal inflammation: implications for the understanding of inflammatory bowel disease. Gut 62, 1795–1805 (2013).

    Article  CAS  PubMed  Google Scholar 

  38. Benchimol, E. I. et al. Epidemiology of pediatric inflammatory bowel disease: a systematic review of international trends. Inflamm. Bowel Dis. 17, 423–439 (2011).

    Article  PubMed  Google Scholar 

  39. Huang, H. et al. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature 547, 173–178 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  41. Hugot, J. P. et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature 411, 599–603 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Consortium, U. I. G. et al. Genome-wide association study of ulcerative colitis identifies three new susceptibility loci, including the HNF4A region. Nat. Genet. 41, 1330–1334 (2009).

    Article  CAS  Google Scholar 

  43. McGovern, D. P. et al. Genome-wide association identifies multiple ulcerative colitis susceptibility loci. Nat. Genet. 42, 332–337 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Martinez-Sanchez, A., Rutter, G. A. & Latreille, M. MiRNAs in β-cell development, identity, and disease. Front. Genet. 7, 226 (2016).

    PubMed  Google Scholar 

  45. Gray, J. M. et al. Genomic views of transcriptional enhancers: essential determinants of cellular identity and activity-dependent responses in the CNS. J. Neurosci. 35, 13819–13826 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Levine, M., Cattoglio, C. & Tjian, R. Looping back to leap forward: transcription enters a new era. Cell 157, 13–25 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Fisher, A. G. Cellular identity and lineage choice. Nat. Rev. Immunol. 2, 977–982 (2002).

    Article  CAS  PubMed  Google Scholar 

  48. Furey, T. S. ChIP-seq and beyond: new and improved methodologies to detect and characterize protein-DNA interactions. Nat. Rev. Genet. 13, 840–852 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Misra, R. & Anderson, D. C. Treatment of recurrent premenstrual orogenital aphthae with implants of low doses of testosterone. BMJ 299, 834 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang, Z., Gerstein, M. & Snyder, M. RNA-seq: a revolutionary tool for transcriptomics. Nat. Rev. Genet. 10, 57–63 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ilott, N. E. & Ponting, C. P. Predicting long non-coding RNAs using RNA sequencing. Methods 63, 50–59 (2013).

    Article  CAS  PubMed  Google Scholar 

  52. Cookson, W., Liang, L., Abecasis, G., Moffatt, M. & Lathrop, M. Mapping complex disease traits with global gene expression. Nat. Rev. Genet. 10, 184–194 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gaffney, D. J. et al. Dissecting the regulatory architecture of gene expression QTLs. Genome Biol. 13, R7 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mangravite, L. M. et al. A statin-dependent QTL for GATM expression is associated with statin-induced myopathy. Nature 502, 377–380 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. de Jong, S. et al. Expression QTL analysis of top loci from GWAS meta-analysis highlights additional schizophrenia candidate genes. Eur. J. Hum. Genet. 20, 1004–1008 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Lawrenson, K. et al. Cis-eQTL analysis and functional validation of candidate susceptibility genes for high-grade serous ovarian cancer. Nat. Commun. 6, 8234 (2015).

    Article  CAS  PubMed  Google Scholar 

  57. Li, Q. et al. Integrative eQTL-based analyses reveal the biology of breast cancer risk loci. Cell 152, 633–641 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Chun, S. et al. Limited statistical evidence for shared genetic effects of eQTLs and autoimmune-disease-associated loci in three major immune-cell types. Nat. Genet. 49, 600–605 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kodzius, R. et al. CAGE: cap analysis of gene expression. Nat. Methods 3, (211–222 (2006).

    Google Scholar 

  60. Andersson, R. et al. An atlas of active enhancers across human cell types and tissues. Nature 507, 455–461 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Chu, T. et al. Chromatin run-on and sequencing maps the transcriptional regulatory landscape of glioblastoma multiforme. Nat. Genet. 50, 1553–1564 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mahony, S. & Pugh, B. F. Protein-DNA binding in high-resolution. Crit. Rev. Biochem. Mol. Biol. 50, 269–283 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Park, P. J. ChIP-seq: advantages and challenges of a maturing technology. Nat. Rev. Genet. 10, 669–680 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Sun, Z., Cunningham, J., Slager, S. & Kocher, J. P. Base resolution methylome profiling: considerations in platform selection, data preprocessing and analysis. Epigenomics 7, 813–828 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Belmont, A. S. Large-scale chromatin organization: the good, the surprising, and the still perplexing. Curr. Opin. Cell Biol. 26, 69–78 (2014).

    Article  CAS  PubMed  Google Scholar 

  66. Fullwood, M. J., Han, Y., Wei, C. L., Ruan, X. & Ruan, Y. Chromatin interaction analysis using paired-end tag sequencing. Curr. Protoc. Mol. Biol. 89, 21.15.1–21.15.25 (2010).

    Google Scholar 

  67. Vinken, M. Regulation of connexin signaling by the epigenetic machinery. Biochim. Biophys. Acta 1859, 262–268 (2016).

    Article  CAS  PubMed  Google Scholar 

  68. Kanno, Y., Vahedi, G., Hirahara, K., Singleton, K. & O’Shea, J. J. Transcriptional and epigenetic control of T helper cell specification: molecular mechanisms underlying commitment and plasticity. Annu. Rev. Immunol. 30, 707–731 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Bartel, D. P. Metazoan microRNAs. Cell 173, 20–51 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Pritchard, C. C., Cheng, H. H. & Tewari, M. MicroRNA profiling: approaches and considerations. Nat. Rev. Genet. 13, 358–369 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Thompson, D. M. & Parker, R. Stressing out over tRNA cleavage. Cell 138, 215–219 (2009).

    Article  CAS  PubMed  Google Scholar 

  72. Lee, Y. S., Shibata, Y., Malhotra, A. & Dutta, A. A novel class of small RNAs: tRNA-derived RNA fragments (tRFs). Genes Dev. 23, 2639–2649 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. De Guire, V. et al. Circulating miRNAs as sensitive and specific biomarkers for the diagnosis and monitoring of human diseases: promises and challenges. Clin. Biochem. 46, 846–860 (2013).

    Article  PubMed  CAS  Google Scholar 

  74. Stork, C. & Zheng, S. Genome-wide profiling of RNA-protein interactions using CLIP-seq. Methods Mol. Biol. 1421, 137–151 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Tanoue, T., Atarashi, K. & Honda, K. Development and maintenance of intestinal regulatory T cells. Nat. Rev. Immunol. 16, 295–309 (2016).

    Article  CAS  PubMed  Google Scholar 

  76. Hyland, N. P. & Cryan, J. F. Microbe-host interactions: influence of the gut microbiota on the enteric nervous system. Dev. Biol. 417, 182–187 (2016).

    Article  CAS  PubMed  Google Scholar 

  77. Lynch, S. V. & Pedersen, O. The human intestinal microbiome in health and disease. N. Engl. J. Med. 375, 2369–2379 (2016).

    Article  CAS  PubMed  Google Scholar 

  78. Wagner, J. et al. Evaluation of PacBio sequencing for full-length bacterial 16S rRNA gene classification. BMC Microbiol. 16, 274 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Myer, P. R., Kim, M., Freetly, H. C. & Smith, T. P. L. Evaluation of 16S rRNA amplicon sequencing using two next-generation sequencing technologies for phylogenetic analysis of the rumen bacterial community in steers. J. Microbiol. Methods 127, 132–140 (2016).

    Article  CAS  PubMed  Google Scholar 

  80. Whelan, F. J. & Surette, M. G. A comprehensive evaluation of the sl1p pipeline for 16S rRNA gene sequencing analysis. Microbiome 5, 100 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Quince, C., Walker, A. W., Simpson, J. T., Loman, N. J. & Segata, N. Shotgun metagenomics, from sampling to analysis. Nat. Biotechnol. 35, 833–844 (2017).

    Article  CAS  PubMed  Google Scholar 

  82. Franzosa, E. A. et al. Relating the metatranscriptome and metagenome of the human gut. Proc. Natl Acad. Sci. USA 111, E2329–E2338 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Weiser, M. et al. Molecular classification of Crohn’s disease reveals two clinically relevant subtypes. Gut 67, 36–42 (2018).

    Article  CAS  PubMed  Google Scholar 

  84. Peck, B. C. et al. MicroRNAs classify different disease behavior phenotypes of Crohn’s disease and may have prognostic utility. Inflamm. Bowel Dis. 21, 2178–2187 (2015).

    Article  PubMed  Google Scholar 

  85. Haberman, Y. et al. Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature. J. Clin. Invest. 124, 3617–3633 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Lewis, J. D. et al. Inflammation, antibiotics, and diet as environmental stressors of the gut microbiome in pediatric Crohn’s disease. Cell Host Microbe 22, 247 (2017).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Shlyueva, D., Stampfel, G. & Stark, A. Transcriptional enhancers: from properties to genome-wide predictions. Nat. Rev. Genet. 15, 272–286 (2014).

    Article  CAS  PubMed  Google Scholar 

  90. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Ostuni, R. et al. Latent enhancers activated by stimulation in differentiated cells. Cell 152, 157–171 (2013).

    Article  CAS  PubMed  Google Scholar 

  92. Gosselin, D. et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159, 1327–1340 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Marigorta, U. M. et al. Transcriptional risk scores link GWAS to eQTLs and predict complications in Crohn’s disease. Nat. Genet. 49, 1517–1521 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wu, F. et al. Identification of microRNAs associated with ileal and colonic Crohn’s disease. Inflamm. Bowel Dis. 16, 1729–1738 (2010).

    Article  PubMed  Google Scholar 

  95. Hasler, R. et al. A functional methylome map of ulcerative colitis. Genome Res. 22, 2130–2137 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Mokry, M. et al. Many inflammatory bowel disease risk loci include regions that regulate gene expression in immune cells and the intestinal epithelium. Gastroenterology 146, 1040–1047 (2014).

    Article  CAS  PubMed  Google Scholar 

  97. Peters, L. A. et al. A functional genomics predictive network model identifies regulators of inflammatory bowel disease. Nat. Genet. 49, 1437–1449 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Lee, J. C. et al. Gene expression profiling of CD8+ T cells predicts prognosis in patients with Crohn disease and ulcerative colitis. J. Clin. Invest. 121, 4170–4179 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Huang, Y. et al. Early transcriptomic changes in the ileal pouch provide insight into the molecular pathogenesis of pouchitis and ulcerative colitis. Inflamm. Bowel Dis. 23, 366–378 (2017).

    PubMed  Google Scholar 

  100. 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).

    Article  CAS  PubMed  Google Scholar 

  101. Gaujoux, R. et al. Cell-centred meta-analysis reveals baseline predictors of anti-TNFalpha non-response in biopsy and blood of patients with IBD. Gut. https://doi.org/10.1136/gutjnl-2017-315494 (2018).

    Article  PubMed  Google Scholar 

  102. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Li, Z. et al. Reciprocal changes of Foxp3 expression in blood and intestinal mucosa in IBD patients responding to infliximab. Inflamm. Bowel Dis. 16, 1299–1310 (2010).

    Article  CAS  PubMed  Google Scholar 

  104. Simon, J. M. et al. Alterations to chromatin in intestinal macrophages link IL-10 deficiency to inappropriate inflammatory responses. Eur. J. Immunol. 46, 1912–1925 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Keith, B. P. et al. Colonic epithelial miR-31 associates with the development of Crohn’s phenotypes. JCI Insight 3, 122788 (2018).

    Article  PubMed  Google Scholar 

  106. Zhang, Z. & Zhang, R. Epigenetics in autoimmune diseases: pathogenesis and prospects for therapy. Autoimmun. Rev. 14, 854–863 (2015).

    Article  CAS  PubMed  Google Scholar 

  107. Qazi, T. J., Quan, Z., Mir, A. & Qing, H. Epigenetics in Alzheimer’s disease: perspective of DNA methylation. Mol. Neurobiol. 55, 1026–1044 (2018).

    Article  CAS  PubMed  Google Scholar 

  108. Miranda-Morales, E. et al. Implications of DNA methylation in Parkinson’s disease. Front. Mol. Neurosci. 10, 225 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Koch, A. et al. Analysis of DNA methylation in cancer: location revisited. Nat. Rev. Clin. Oncol. 15, 459–466 (2018).

    Article  CAS  PubMed  Google Scholar 

  110. Widschwendter, M. et al. Epigenome-based cancer risk prediction: rationale, opportunities and challenges. Nat. Rev. Clin. Oncol. 15, 292–309 (2018).

    Article  PubMed  Google Scholar 

  111. Lin, Z. et al. Identification of disease-associated DNA methylation in intestinal tissues from patients with inflammatory bowel disease. Clin. Genet. 80, 59–67 (2011).

    Article  CAS  PubMed  Google Scholar 

  112. Sadler, T. et al. Genome-wide analysis of DNA methylation and gene expression defines molecular characteristics of Crohn’s disease-associated fibrosis. Clin. Epigenet. 8, 30 (2016).

    Article  CAS  Google Scholar 

  113. Petrey, A. C. & de la Motte, C. A. The extracellular matrix in IBD: a dynamic mediator of inflammation. Curr. Opin. Gastroenterol. 33, 234–238 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Consortium, E. P. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  CAS  Google Scholar 

  115. Meddens, C. A. et al. Systematic analysis of chromatin interactions at disease associated loci links novel candidate genes to inflammatory bowel disease. Genome Biol. 17, 247 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Ventham, N. T. et al. Integrative epigenome-wide analysis demonstrates that DNA methylation may mediate genetic risk in inflammatory bowel disease. Nat. Commun. 7, 13507 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Adams, A. T. et al. Two-stage genome-wide methylation profiling in childhood-onset Crohn’s Disease implicates epigenetic alterations at the VMP1/MIR21 and HLA loci. Inflamm. Bowel Dis. 20, 1784–1793 (2014).

    Article  PubMed  Google Scholar 

  118. Howell, K. J. et al. DNA methylation and transcription patterns in intestinal epithelial cells from pediatric patients with inflammatory bowel diseases differentiate disease subtypes and associate with outcome. Gastroenterology 154, 585–598 (2018).

    Article  CAS  PubMed  Google Scholar 

  119. Beres, N. J. et al. Altered mucosal expression of microRNAs in pediatric patients with inflammatory bowel disease. Dig. Liver Dis. 49, 378–387 (2017).

    Article  CAS  PubMed  Google Scholar 

  120. Sun, C. M., Wu, J., Zhang, H., Shi, G. & Chen, Z. T. Circulating miR-125a but not miR-125b is decreased in active disease status and negatively correlates with disease severity as well as inflammatory cytokines in patients with Crohn’s disease. World J. Gastroenterol. 23, 7888–7898 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Haberman, Y. et al. Long ncRNA landscape in the ileum of treatment-naive early-onset Crohn disease. Inflamm. Bowel Dis. 24, 346–360 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  122. Viladomiu, M. et al. IgA-coated E. coli enriched in Crohn’s disease spondyloarthritis promote TH17-dependent inflammation. Sci. Transl Med. 9, eaaf9655 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Palm, N. W. et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 158, 1000–1010 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Pascal, V. et al. A microbial signature for Crohn’s disease. Gut 66, 813–822 (2017).

    Article  CAS  PubMed  Google Scholar 

  125. Tyler, A. D. et al. Characterization of the gut-associated microbiome in inflammatory pouch complications following ileal pouch-anal anastomosis. PLOS ONE 8, e66934 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Kugathasan, S. et al. Prediction of complicated disease course for children newly diagnosed with Crohn’s disease: a multicentre inception cohort study. Lancet 389, 1710–1718 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  127. Mottawea, W. et al. Altered intestinal microbiota-host mitochondria crosstalk in new onset Crohn’s disease. Nat. Commun. 7, 13419 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. de Souza, H. S. P., Fiocchi, C. & Iliopoulos, D. The IBD interactome: an integrated view of aetiology, pathogenesis and therapy. Nat. Rev. Gastroenterol. Hepatol. 14, 739–749 (2017).

    Article  PubMed  Google Scholar 

  129. Strimbu, K. & Tavel, J. A. What are biomarkers? Curr. Opin. HIV AIDS 5, 463–466 (2010).

    PubMed  Google Scholar 

  130. Guler, E. N. Gene expression profiling in breast cancer and its effect on therapy selection in early-stage breast cancer. Eur. J. Breast Health 13, 168–174 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  131. Prat, A. et al. Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast 24 (Suppl. 2), S26–S35 (2015).

    Article  PubMed  Google Scholar 

  132. Lowry, P. W. et al. Measurement of thiopurine methyltransferase activity and azathioprine metabolites in patients with inflammatory bowel disease. Gut 49, 665–670 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The work of the authors is supported by 1R01DK104828-01A1 from National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (to T.S.F. and S.Z.S.), P01-DK094779-01A1 from NIDDK (to S.Z.S.), P30-DK034987 from NIDDK (to S.Z.S.), 1-16-ACE-47 American Diabetes Association Pathway Award (to P.S.), University of North Carolina (UNC) Nutrition Obesity Research Center Pilot & Feasibility Grant P30DK056350 (to P.S.), the Crohn’s and Colitis Foundation PRO-KIIDS NETWORK (to S.Z.S. and P.S.), the Sinai-Helmsley Alliance for Research Excellence (SHARE) from the Helmsley Trust (to T.S.F. and S.Z.S.) and the UNC University Cancer Research Fund (UCRF; to T.S.F. and P.S.).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed equally to this manuscript.

Corresponding authors

Correspondence to Terrence S. Furey or Shehzad Z. Sheikh.

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.

Related linkS

Genotype-Tissue Expression (GTEx): https://gtexportal.org/home/

Roadmap Epigenomics: http://www.roadmapepigenomics.org/

Sinai-Helmsley Alliance for Research Excellence (SHARE): https://sinai-helmsley.org/publicWebsite/index.php

The Cancer Genome Atlas: https://cancergenome.nih.gov

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Furey, T.S., Sethupathy, P. & Sheikh, S.Z. Redefining the IBDs using genome-scale molecular phenotyping. Nat Rev Gastroenterol Hepatol 16, 296–311 (2019). https://doi.org/10.1038/s41575-019-0118-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-019-0118-x

This article is cited by

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