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:

Lessons learned — resolving the enigma of genetic factors in IBS

Key Points

  • Genetic studies in IBS range from family and twin studies to candidate gene approaches and genome-wide association studies

  • Despite enlarged sample sizes, increased statistical power and meta-analyses, positive associations between gene variations and IBS subtypes are still scarce and many have not been reproduced

  • Epigenetic and pharmacogenetic approaches are in their infancy

  • A major pitfall in IBS research is the lack of large homogenized case–control cohorts recruited according to standardized and harmonized criteria

Abstract

IBS is the most prevalent functional gastrointestinal disorder and phenotypically characterized by chronic abdominal discomfort, pain and altered defecation patterns. The pathophysiology of IBS is multifactorial, albeit with a substantial genetic component. To date, studies using various methodologies, ranging from family and twin studies to candidate gene approaches and genome-wide association studies, have identified several genetic variants in the context of IBS. Yet, despite enlarged sample sizes, increased statistical power and meta-analyses in the past 7 years, positive associations are still scarce and/or have not been reproduced. In addition, epigenetic and pharmacogenetic approaches remain in their infancy. A major hurdle is the lack of large homogenized case–control cohorts recruited according to standardized and harmonized criteria. The COST Action BM1106 GENIEUR (GENes in Irritable Bowel Syndrome Research Network EURope) has been established to address these obstacles. In this Review, the (epi)genetic working group of GENIEUR reports on the current state-of-the-art in the field, highlights fundamental flaws and pitfalls in current IBS (epi)genetic research and provides a vision on how to address and improve (epi)genetic approaches in this complex disorder in the future.

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

Figure 1: Multiple layers of complexity on environmental and genetic or epigenetic levels contribute to the pathogenesis of IBS and comorbid conditions.
Figure 2: IBS-related pathways, based on genetic and epigenetic findings including potential pharmacogenetic targets.
Figure 3: Future approach in IBS genetics or epigenetics research.

Similar content being viewed by others

References

  1. Drossman, D. A., Camilleri, M., Mayer, E. A. & Whitehead, W. E. AGA technical review on irritable bowel syndrome. Gastroenterology 123, 2108–2131 (2002).

    PubMed  Google Scholar 

  2. Azpiroz, F. et al. Mechanisms of hypersensitivity in IBS and functional disorders. Neurogastroenterol. Motil. 19, 62–88 (2007).

    CAS  PubMed  Google Scholar 

  3. Longstreth, G. F. et al. Functional bowel disorders. Gastroenterology 130, 1480–1491 (2006).

    PubMed  Google Scholar 

  4. Spiller, R. C. et al. The Patient Health Questionnaire 12 Somatic Symptom scale as a predictor of symptom severity and consulting behaviour in patients with irritable bowel syndrome and symptomatic diverticular disease. Aliment. Pharmacol. Ther. 32, 811–820 (2010).

    CAS  PubMed  Google Scholar 

  5. North, C. S. et al. The presentation of irritable bowel syndrome in the context of somatization disorder. Clin. Gastroenterol. Hepatol. 2, 787–795 (2004).

    PubMed  Google Scholar 

  6. Frissora, C. L. & Koch, K. L. Symptom overlap and comorbidity of irritable bowel syndrome with other conditions. Curr. Gastroenterol. Rep. 7, 264–271 (2005).

    PubMed  Google Scholar 

  7. Cole, J. A., Rothman, K. J., Cabral, H. J., Zhang, Y. & Farraye, F. A. Migraine, fibromyalgia, and depression among people with IBS: a prevalence study. BMC Gastroenterol. 6, 26 (2006).

    PubMed  PubMed Central  Google Scholar 

  8. Hillilä, M. T., Färkkilä, N. J. & Färkkilä, M. A. Societal costs for irritable bowel syndrome — a population based study. Scand. J. Gastroenterol. 45, 582–591 (2010).

    PubMed  Google Scholar 

  9. Larauche, M., Mulak, A. & Tache, Y. Stress and visceral pain: from animal models to clinical therapies. Exp. Neurol. 233, 49–67 (2012).

    PubMed  Google Scholar 

  10. Spiller, R. C. Role of infection in irritable bowel syndrome. J. Gastroenterol. 42 (Suppl. 17), 41–47 (2007).

    PubMed  Google Scholar 

  11. Mayer, E. A. Gut feelings: the emerging biology of gut−brain communication. Nat. Rev. Neurosci. 12, 453–66 (2011).

    CAS  PubMed  Google Scholar 

  12. Öhman, L., Törnblom, H. & Simren, M. Crosstalk at the mucosal border: importance of the gut microenvironment in IBS. Nat. Rev. Gastroenterol. Hepatol. 12, 36–49 (2015).

    PubMed  Google Scholar 

  13. Fukudo, S. & Kanazawa, M. Gene, environment, and brain−gut interactions in irritable bowel syndrome. J. Gastroenterol. Hepatol. 26 (Suppl. 3), 110–115 (2011).

    CAS  PubMed  Google Scholar 

  14. Buonavolonta, R. et al. Familial aggregation in children affected by functional gastrointestinal disorders. J. Pediatr. Gastroenterol. Nutr. 50, 500–505 (2010).

    CAS  PubMed  Google Scholar 

  15. Saito, Y. A. et al. Irritable bowel syndrome aggregates strongly in families: a family-based case-control study. Neurogastroenterol. Motil. 20, 790–797 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Saito, Y. A. et al. Familial aggregation of irritable bowel syndrome: a family case-control study. Am. J. Gastroenterol. 105, 833–841 (2010).

    PubMed  PubMed Central  Google Scholar 

  17. Bellentani, S. et al. A simple score for the identification of patients at high risk of organic diseases of the colon in the family doctor consulting room. Fam. Pract. 7, 307–312 (1990).

    CAS  PubMed  Google Scholar 

  18. Kalantar, J. S., Locke, G. R. 3rd, Zinsmeister, A. R., Beighley, C. M. & Talley, N. J. Familial aggregation of irritable bowel syndrome: a prospective study. Gut 52, 1703–1707 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Waehrens, R., Ohlsson, H., Sundquist, J., Sundquist, K. & Zoller, B. Risk of irritable bowel syndrome in first-degree, second-degree and third-degree relatives of affected individuals: a nationwide family study in Sweden. Gut 64, 215–221 (2015).

    PubMed  Google Scholar 

  20. Whorwell, P. J., McCallum, M., Creed, F. H. & Roberts, C. T. Non-colonic features of irritable bowel syndrome. Gut 27, 37–40 (1986).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Saito, Y. A. The role of genetics in IBS. Gastroenterol. Clin. North Am. 40, 45–67 (2011).

    PubMed  PubMed Central  Google Scholar 

  22. Bengtson, M. B., Ronning, T., Vatn, M. H. & Harris, J. R. Irritable bowel syndrome in twins: genes and environment. Gut 55, 1754–1759 (2006).

    PubMed  PubMed Central  Google Scholar 

  23. Lembo, A., Zaman, M., Jones, M. & Talley, N. J. Influence of genetics on irritable bowel syndrome, gastro-oesophageal reflux and dyspepsia: a twin study. Aliment. Pharmacol. Ther. 25, 1343–1350 (2007).

    CAS  PubMed  Google Scholar 

  24. Morris-Yates, A., Talley, N. J., Boyce, P. M., Nandurkar, S. & Andrews, G. Evidence of a genetic contribution to functional bowel disorder. Am. J. Gastroenterol. 93, 1311–1317 (1998).

    CAS  PubMed  Google Scholar 

  25. Bengtson, M. B., Aamodt, G., Vatn, M. H. & Harris, J. R. Co-occurrence of IBS and symptoms of anxiety or depression, among Norwegian twins, is influenced by both heredity and intrauterine growth. BMC Gastroenterol. 15, 9 (2015).

    PubMed  PubMed Central  Google Scholar 

  26. McGowan, P. O. et al. Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat. Neurosci. 12, 342–348 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Ke, X. et al. Intrauterine growth retardation affects expression and epigenetic characteristics of the rat hippocampal glucocorticoid receptor gene. Physiol. Genomics 42, 177–189 (2010).

    CAS  PubMed  Google Scholar 

  28. Levy, R. L. et al. Irritable bowel syndrome in twins: heredity and social learning both contribute to etiology. Gastroenterology 121, 799–804 (2001).

    CAS  PubMed  Google Scholar 

  29. Levy, R. L. et al. Increased somatic complaints and health-care utilization in children: effects of parent IBS status and parent response to gastrointestinal symptoms. Am. J. Gastroenterol. 99, 2442–2451 (2004).

    PubMed  Google Scholar 

  30. Videlock, E. J. et al. Childhood trauma is associated with hypothalamic−pituitary−adrenal axis responsiveness in irritable bowel syndrome. Gastroenterology 137, 1954–1962 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Niesler, B. et al. 5-HTTLPR and STin2 polymorphisms in the serotonin transporter gene and irritable bowel syndrome: effect of bowel habit and sex. Eur. J. Gastroenterol. Hepatol. 22, 856–861 (2010).

    CAS  PubMed  Google Scholar 

  32. Choi, Y. J. et al. Association between SLC6A4 serotonin transporter gene lainked polymorphic region and ADRA2A−1291C>G and irritable bowel syndrome in Korea. J. Neurogastroenterol. Motil. 20, 388–399 (2014).

    PubMed  PubMed Central  Google Scholar 

  33. Kim, H. J. et al. Association of distinct α2 adrenoceptor and serotonin transporter polymorphisms with constipation and somatic symptoms in functional gastrointestinal disorders. Gut 53, 829–837 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Van Kerkhoven, L. A., Laheij, R. J. & Jansen, J. B. Meta-analysis: a functional polymorphism in the gene encoding for activity of the serotonin transporter protein is not associated with the irritable bowel syndrome. Aliment. Pharmacol. Ther. 26, 979–986 (2007).

    CAS  PubMed  Google Scholar 

  35. Yeo, A. et al. Association between a functional polymorphism in the serotonin transporter gene and diarrhoea predominant irritable bowel syndrome in women. Gut 53, 1452–1458 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Park, J. M. et al. Serotonin transporter gene polymorphism and irritable bowel syndrome. Neurogastroenterol. Motil. 18, 995–1000 (2006).

    CAS  PubMed  Google Scholar 

  37. Kumar, S., Ranjan, P., Mittal, B. & Ghoshal, U. C. Serotonin transporter gene (SLC6A4) polymorphism in patients with irritable bowel syndrome and healthy controls. J. Gastrointestin. Liver Dis. 21, 31–38 (2012).

    PubMed  Google Scholar 

  38. Pata, C. et al. Serotonin transporter gene polymorphism in irritable bowel syndrome. Am. J. Gastroenterol. 97, 1780–1784 (2002).

    CAS  PubMed  Google Scholar 

  39. Saito, Y. A. et al. A genetic association study of 5-HTT LPR and GNβ3 C825T polymorphisms with irritable bowel syndrome. Neurogastroenterol. Motil. 19, 465–470 (2007).

    CAS  PubMed  Google Scholar 

  40. Sikander, A. et al. Serotonin transporter promoter variant: analysis in Indian IBS patients and control population. J. Clin. Gastroenterol. 43, 957–961 (2009).

    CAS  PubMed  Google Scholar 

  41. Lee, D. Y. et al. Serotonin transporter gene polymorphism in healthy adults and patients with irritable bowel syndrome. Kor. J. Gastroenterol. 43, 18–22 (in Korean) (2004).

    Google Scholar 

  42. Wang, B. M. et al. Serotonin transporter gene polymorphism in irritable bowel syndrome. Zhonghua Nei Ke Za Zhi 43, 439–441 (in Chinese) (2004).

    CAS  PubMed  Google Scholar 

  43. Li, Y. et al. The association of serotonin transporter genetic polymorphisms and irritable bowel syndrome and its influence on tegaserod treatment in Chinese patients. Dig. Dis. Sci. 52, 2942–2949 (2007).

    CAS  PubMed  Google Scholar 

  44. Yuan, J. et al. Association study of serotonin transporter SLC6A4 gene with Chinese Han irritable bowel syndrome. PLoS ONE 9, e84414 (2014).

    PubMed  PubMed Central  Google Scholar 

  45. Kohen, R. et al. The serotonin transporter polymorphism rs25531 is associated with irritable bowel syndrome. Dig. Dis. Sci. 54, 2663–2670 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Lesch, K. P. et al. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region. Science 274, 1527–1531 (1996).

    CAS  PubMed  Google Scholar 

  47. Jarrett, M. E. et al. Relationship of SERT polymorphisms to depressive and anxiety symptoms in irritable bowel syndrome. Biol. Res. Nurs. 9, 161–169 (2007).

    CAS  PubMed  Google Scholar 

  48. Blom, R. M. et al. Association between a serotonin transporter promoter polymorphism (5HTTLPR) and personality disorder traits in a community sample. J. Psychiatr. Res. 45, 1153–1159 (2011).

    PubMed  PubMed Central  Google Scholar 

  49. Camilleri, M. et al. Candidate genes and sensory functions in health and irritable bowel syndrome. Am. J. Physiol. Gastrointest. Liver Physiol. 295, G219–G225 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Farmer, A. D. et al. Psychophysiological responses to pain identify reproducible human clusters. Pain 154, 2266–2276 (2013).

    CAS  PubMed  Google Scholar 

  51. Pezawas, L. et al. 5-HTTLPR polymorphism impacts human cingulate−amygdala interactions: a genetic susceptibility mechanism for depression. Nat. Neurosci. 8, 828–834 (2005).

    CAS  PubMed  Google Scholar 

  52. Fukudo, S. et al. Impact of serotonin transporter gene polymorphism on brain activation by colorectal distention. NeuroImage 47, 946–951 (2009).

    CAS  PubMed  Google Scholar 

  53. Spiller, R. C. Targeting the 5-HT3 receptor in the treatment of irritable bowel syndrome. Curr. Opin. Pharmacol. 11, 68–74 (2011).

    CAS  PubMed  Google Scholar 

  54. Garsed, K. et al. A randomised trial of ondansetron for the treatment of irritable bowel syndrome with diarrhoea. Gut 63, 1617–1625 (2014).

    CAS  PubMed  Google Scholar 

  55. Walstab, J., Rappold, G. & Niesler, B. 5-HT3 receptors: role in disease and target of drugs. Pharmacol. Ther. 128, 146–169 (2010).

    CAS  PubMed  Google Scholar 

  56. Camilleri, M. et al. Efficacy and safety of alosetron in women with irritable bowel syndrome: a randomised, placebo-controlled trial. Lancet 355, 1035–1040 (2000).

    CAS  PubMed  Google Scholar 

  57. Fukudo, S., Ida, M., Akiho, H., Nakashima, Y. & Matsueda, K. Effect of ramosetron on stool consistency in male patients with irritable bowel syndrome with diarrhea. Clin. Gastroenterol. Hepatol. 12, 953–959.e4 (2014).

    CAS  PubMed  Google Scholar 

  58. Kapeller, J. et al. First evidence for an association of a functional variant in the microRNA-510 target site of the serotonin receptor-type 3E gene with diarrhea predominant irritable bowel syndrome. Hum. Mol. Genet. 17, 2967–2977 (2008).

    CAS  PubMed  Google Scholar 

  59. Kapeller, J. et al. A coding variant in the serotonin receptor 3c subunit is associated with diarrhea predominant irritable bowel syndrome. Gastroenterology 136, A-155–A-156 (2009).

    Google Scholar 

  60. Zhang, Y., Huang, Y. & Bo, P. Association between diarrhea-predominant irritable bowel syndrome and HTR3A, HTR3E gene polymorphism in Yangzhou, Jiangsu province, China. Zhonghua Liu Xing Bing Xue Za Zhi 34, 721–724 (in Chinese) (2013).

    CAS  PubMed  Google Scholar 

  61. Gu, Q. Y., Zhang, J., Feng, Y. C., Dai, G. R. & Du, W. P. Association of genetic polymorphisms in HTR3A and HTR3E with diarrhea predominant irritable bowel syndrome. Int. J. Clin. Exp. Med. 8, 4581–4585 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. de Vries, D., ter Linde, J., van Herwaarden, M., Smout, A. & Samsom, M. Serotonin receptor 3a polymorphism C178t is associated with visceral hypersensitivity in GERD. Gastroenterology Abstr. 132, A276 (2007).

    Google Scholar 

  63. Mujakovic, S. et al. Serotonin receptor 3A polymorphism c.-42C > T is associated with severe dyspepsia. BMC Med. Genet. 12, 140 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Melke, J. et al. A polymorphism in the serotonin receptor 3A (HTR3A) gene and its association with harm avoidance in women. Arch. Gen. Psychiatry 60, 1017–1023 (2003).

    CAS  PubMed  Google Scholar 

  65. Niesler, B. et al. Serotonin receptor gene HTR3A variants in schizophrenic and bipolar affective patients. Pharmacogenetics 11, 21–27 (2001).

    CAS  PubMed  Google Scholar 

  66. Cloninger, C. R. A systematic method for clinical description and classification of personality variants. A Proposal. Arch. Gen. Psychiatry 44, 573–588 (1987).

    CAS  PubMed  Google Scholar 

  67. Kilpatrick, L. A. et al. The HTR3A polymorphism c. -42C>T is associated with amygdala responsiveness in patients with irritable bowel syndrome. Gastroenterology 140, 1943–1951 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Yamada, K. et al. Distinguishable haplotype blocks in the HTR3A and HTR3B region in the Japanese reveal evidence of association of HTR3B with female major depression. Biol. Psychiatry 60, 192–201 (2006).

    CAS  PubMed  Google Scholar 

  69. Hammer, C. et al. Functional variants of the serotonin receptor type 3A and B gene are associated with eating disorders. Pharmacogenet. Genomics 19, 790–799 (2009).

    CAS  PubMed  Google Scholar 

  70. Hammer, C. et al. Replication of functional serotonin receptor type 3A and B variants in bipolar affective disorder: a European multicenter study. Transl. Psychiatry 2, e103 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Aibiki, L. et al. Impact of serotonin receptor-3 gene polymorphism on irritable bowel syndrome. Gastroenterology 132, A134–A135 (2007).

    Google Scholar 

  72. Ek, W. E. et al. Exploring the genetics of irritable bowel syndrome: a GWA study in the general population and replication in multinational case-control cohorts. Gut 64, 1774–1782 (2015).

    CAS  PubMed  Google Scholar 

  73. Iidaka, T. et al. A variant C178T in the regulatory region of the serotonin receptor gene HTR3A modulates neural activation in the human amygdala. J. Neurosci. 25, 6460–6466 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Fukudo, S. et al. Impact of serotonin-3 receptor gene polymorphism on brain activation by rectal distention in human. Gastroenterology 136, A170 (2009).

    Google Scholar 

  75. Horjales-Araujo, E. et al. Polymorphism in serotonin receptor 3B is associated with pain catastrophizing. PLoS ONE 8, e78889 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Mulak, A. et al. Association of polymorphisms in 5-HT2A and 5-HT2C receptors genes with depressive and anxiety disorders in patients with irritable bowel syndrome. Gastroenterology 144, S725 (2013).

    Google Scholar 

  77. Jun, S., Kohen, R., Cain, K. C., Jarrett, M. E. & Heitkemper, M. M. Associations of tryptophan hydroxylase gene polymorphisms with irritable bowel syndrome. Neurogastroenterol. Motil. 23, 233–e116 (2011).

    CAS  PubMed  Google Scholar 

  78. Jun, S. E., Kohen, R., Cain, K. C., Jarrett, M. E. & Heitkemper, M. M. TPH gene polymorphisms are associated with disease perception and quality of life in women with irritable bowel syndrome. Biol. Res. Nurs. 16, 95–104 (2014).

    CAS  PubMed  Google Scholar 

  79. Pata, C. et al. Association of the -1438 G/A and 102 T/C polymorphism of the 5-Ht2A receptor gene with irritable bowel syndrome 5-Ht2A gene polymorphism in irritable bowel syndrome. J. Clin. Gastroenterol. 38, 561–566 (2004).

    CAS  PubMed  Google Scholar 

  80. Beyder, A. et al. Loss-of-function of the voltage-gated sodium channel NaV1.5 (channelopathies) in patients with irritable bowel syndrome. Gastroenterology 146, 1659–1668 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Saito, Y. A. et al. Sodium channel mutation in irritable bowel syndrome: evidence for an ion channelopathy. Am. J. Physiol. Gastrointest. Liver Physiol. 296, G211–G218 (2009).

    CAS  PubMed  Google Scholar 

  82. Wouters, M. M. et al. Genetic variants in CDC42 and NXPH1 as susceptibility factors for constipation and diarrhoea predominant irritable bowel syndrome. Gut 63, 1103–1111 (2014).

    CAS  PubMed  Google Scholar 

  83. van den Oord, E. J. et al. Genomewide association analysis followed by a replication study implicates a novel candidate gene for neuroticism. Arch. Gen. Psychiatry 65, 1062–1071 (2008).

    PubMed  Google Scholar 

  84. Sikander, A. et al. Association of alpha 2A adrenergic receptor gene (ADRA2A) polymorphism with irritable bowel syndrome, microscopic and ulcerative colitis. Clin. Chim. Acta 411, 59–63 (2010).

    CAS  PubMed  Google Scholar 

  85. Camilleri, M. et al. Cannabinoid receptor 1 gene and irritable bowel syndrome: phenotype and quantitative traits. Am. J. Physiol. Gastrointest. Liver Physiol. 304, G553–G560 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Camilleri, M. et al. Genetic variation in endocannabinoid metabolism, gastrointestinal motility, and sensation. Am. J. Physiol. Gastrointest. Liver Physiol. 294, G13–G19 (2008).

    CAS  PubMed  Google Scholar 

  87. Karling, P. et al. The relationship between the val158met catechol-O-methyltransferase (COMT) polymorphism and irritable bowel syndrome. PLoS ONE 6, e18035 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Jiang, Y., Nie, Y., Li, Y. & Zhang, L. Association of cannabinoid type 1 receptor and fatty acid amide hydrolase genetic polymorphisms in Chinese patients with irritable bowel syndrome. J. Gastroenterol. Hepatol. 29, 1186–1191 (2014).

    CAS  PubMed  Google Scholar 

  89. Park, J. M. et al. Cannabinoid receptor 1 gene polymorphism and irritable bowel syndrome in the Korean population: a hypothesis-generating study. J. Clin. Gastroenterol. 45, 45–49 (2011).

    CAS  PubMed  Google Scholar 

  90. Zhou, Q., Souba, W. W., Croce, C. M. & Verne, G. N. MicroRNA-29a regulates intestinal membrane permeability in patients with irritable bowel syndrome. Gut 59, 775–784 (2010).

    CAS  PubMed  Google Scholar 

  91. Dunlop, S. P. et al. Abnormal intestinal permeability in subgroups of diarrhea-predominant irritable bowel syndromes. Am. J. Gastroenterol. 101, 1288–1294 (2006).

    PubMed  Google Scholar 

  92. Martinez, C. et al. The jejunum of diarrhea-predominant irritable bowel syndrome shows molecular alterations in the tight junction signaling pathway that are associated with mucosal pathobiology and clinical manifestations. Am. J. Gastroenterol. 107, 736–746 (2012).

    CAS  PubMed  Google Scholar 

  93. Martinez, C. et al. Diarrhoea-predominant irritable bowel syndrome: an organic disorder with structural abnormalities in the jejunal epithelial barrier. Gut 62, 1160–1168 (2013).

    CAS  PubMed  Google Scholar 

  94. Villani, A. C. et al. Genetic risk factors for post-infectious irritable bowel syndrome following a waterborne outbreak of gastroenteritis. Gastroenterology 138, 1502–1513 (2010).

    CAS  PubMed  Google Scholar 

  95. Bashashati, M. et al. Cytokine gene polymorphisms are associated with irritable bowel syndrome: a systematic review and meta-analysis. Neurogastroenterol. Motil. 24, e1102–e1566 (2012).

    Google Scholar 

  96. Czogalla, B. et al. A meta-analysis of immunogenetic Case−Control Association Studies in irritable bowel syndrome. Neurogastroenterol. Motil. 27, 717–727 (2015).

    CAS  PubMed  Google Scholar 

  97. Lee, Y. J. & Park, K. S. Irritable bowel syndrome: emerging paradigm in pathophysiology. World J. Gastroenterol. 20, 2456–2469 (2014).

    PubMed  PubMed Central  Google Scholar 

  98. Zucchelli, M. et al. Association of TNFSF15 polymorphism with irritable bowel syndrome. Gut 60, 1671–1677 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Swan, C. et al. Identifying and testing candidate genetic polymorphisms in the irritable bowel syndrome (IBS): association with TNFSF15 and TNFα. Gut 62, 985–994 (2013).

    CAS  PubMed  Google Scholar 

  100. Bamias, G. et al. Expression, localization, and functional activity of TL1A, a novel Th1-polarizing cytokine in inflammatory bowel disease. J. Immunol. 171, 4868–4874 (2003).

    CAS  PubMed  Google Scholar 

  101. Kugathasan, S. & Cohen, S. Searching for new clues in inflammatory bowel disease: tell tales from pediatric IBD natural history studies. Gastroenterology 135, 1038–1041 (2008).

    PubMed  Google Scholar 

  102. Bamias, G. et al. Upregulation and nuclear localization of TNF-like cytokine 1A (TL1A) and its receptors DR3 and DcR3 in psoriatic skin lesions. Exp. Dermatol. 20, 725–731 (2011).

    CAS  PubMed  Google Scholar 

  103. Pappu, B. P. et al. TL1A−DR3 interaction regulates Th17 cell function and Th17-mediated autoimmune disease. J. Exp. Med. 205, 1049–1062 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Kamada, N. et al. TL1A produced by lamina propria macrophages induces Th1 and Th17 immune responses in cooperation with IL-23 in patients with Crohn's disease. Inflamm. Bowel Dis. 16, 568–575 (2010).

    Google Scholar 

  105. Meylan, F. et al. The TNF-family cytokine TL1A promotes allergic immunopathology through group 2 innate lymphoid cells. Mucosal Immunol. 7, 958–968 (2014).

    CAS  PubMed  Google Scholar 

  106. van der Veek, P. P., van den Berg, M., de Kroon, Y. E., Verspaget, H. W. & Masclee, A. A. Role of tumor necrosis factor-α and interleukin-10 gene polymorphisms in irritable bowel syndrome. Am. J. Gastroenterol. 100, 2510–2516 (2005).

    CAS  PubMed  Google Scholar 

  107. Gonsalkorale, W. M., Perrey, C., Pravica, V., Whorwell, P. J. & Hutchinson, I. V. Interleukin 10 genotypes in irritable bowel syndrome: evidence for an inflammatory component? Gut 52, 91–93 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Schmulson, M. et al. IL-10 and TNF-α polymorphisms in subjects with irritable bowel syndrome in Mexico. Rev. Esp. Enferm. Dig. 105, 392–399 (2013).

    CAS  PubMed  Google Scholar 

  109. Barkhordari, E. et al. Proinflammatory cytokine gene polymorphisms in irritable bowel syndrome. J. Clin. Immunol. 30, 74–79 (2010).

    CAS  PubMed  Google Scholar 

  110. Romero-Valdovinos, M. et al. Interleukin-8 and -10 gene polymorphisms in irritable bowel syndrome. Mol. Biol. Rep. 39, 8837–8843 (2012).

    CAS  PubMed  Google Scholar 

  111. Olivo-Diaz, A. et al. Findings related to IL-8 and IL-10 gene polymorphisms in a Mexican patient population with irritable bowel syndrome infected with Blastocystis. Parasitol. Res. 111, 487–491 (2012).

    PubMed  Google Scholar 

  112. Qin, S. Y., Jiang, H. X., Lu, D. H. & Zhou, Y. Association of interleukin-10 polymorphisms with risk of irritable bowel syndrome: a meta-analysis. World J. Gastroenterol. 19, 9472–9480 (2013).

    PubMed  PubMed Central  Google Scholar 

  113. Santhosh, S. et al. Cytokine gene polymorphisms in irritable bowel syndrome in Indian population — a pilot case control study. Trop. Gastroenterol. 31, 30–33 (2010).

    CAS  PubMed  Google Scholar 

  114. Shiotani, A. et al. S100A expression and interleukin-10 polymorphisms are associated with ulcerative colitis and diarrhea predominant irritable bowel syndrome. Dig. Dis. Sci. 58, 2314–2323 (2013).

    CAS  PubMed  Google Scholar 

  115. Holliday, E. G. et al. Genome-wide association study identifies two novel genomic regions in irritable bowel syndrome. Am. J. Gastroenterol. 109, 770–772 (2014).

    CAS  PubMed  Google Scholar 

  116. Saito, Y. A. et al. A candidate gene association study of functional 'psychiatric' polymorphisms in irritable bowel syndrome (IBS). Gastroenterology 138, S90 (2010).

    Google Scholar 

  117. Dinan, T. G., Cryan, J., Shanahan, F., Keeling, P. W. & Quigley, E. M. IBS: an epigenetic perspective. Nat. Rev. Gastroenterol. Hepatol. 7, 465–471 (2010).

    PubMed  Google Scholar 

  118. Tran, L., Chaloner, A., Sawalha, A. H. & Greenwood Van-Meerveld, B. Importance of epigenetic mechanisms in visceral pain induced by chronic water avoidance stress. Psychoneuroendocrinology 38, 898–906 (2013).

    CAS  PubMed  Google Scholar 

  119. van den Wijngaard, R. M. et al. Susceptibility to stress induced visceral hypersensitivity in maternally separated rats is transferred across generations. Neurogastroenterol. Motil. 25, e780–e790 (2013).

    CAS  PubMed  Google Scholar 

  120. Zhou, Q. et al. MicroRNA 29 targets nuclear factor-kappaB-repressing factor and Claudin 1 to increase intestinal permeability. Gastroenterology 148, 158–169.e8 (2015).

    CAS  PubMed  Google Scholar 

  121. Zhou, Q. et al. Decreased miR-199 augments visceral pain in patients with IBS through translational upregulation of TRPV1. Gut http://dx.doi.org/10.1136/gutjnl-2013-306464 (2015).

  122. Fourie, N. H. et al. Elevated circulating miR-150 and miR-342-3p in patients with irritable bowel syndrome. Exp. Mol. Pathol. 96, 422–425 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Gheinani, A. H., Burkhard, F. C. & Monastyrskaya, K. Deciphering microRNA code in pain and inflammation: lessons from bladder pain syndrome. Cell. Mol. Life Sci. 70, 3773–3789 (2013).

    CAS  PubMed  Google Scholar 

  124. Ehrenreich, H. & Nave, K. A. Phenotype-Based Genetic Association Studies (PGAS) — towards understanding the contribution of common genetic variants to schizophrenia subphenotypes. Genes (Basel) 5, 97–105 (2014).

    Google Scholar 

  125. Clarke, G. M. et al. Basic statistical analysis in genetic case-control studies. Nat. Protoc. 6, 121–133 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Balding, D. J. A tutorial on statistical methods for population association studies. Nat. Rev. Genet. 7, 781–791 (2006).

    CAS  PubMed  Google Scholar 

  127. Laird, N. M. & Lange, C. The Fundamentals of Modern Statistical Genetics: Statistics for Biology and Health (Springer, 2010).

    Google Scholar 

  128. Bearcroft, C. P., Perrett, D. & Farthing, M. J. Postprandial plasma 5-hydroxytryptamine in diarrhoea predominant irritable bowel syndrome: a pilot study. Gut 42, 42–46 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Camilleri, M. et al. Serotonin-transporter polymorphism pharmacogenetics in diarrhea-predominant irritable bowel syndrome. Gastroenterology 123, 425–432 (2002).

    CAS  PubMed  Google Scholar 

  130. Camilleri, M. et al. Pharmacogenetics of low dose clonidine in irritable bowel syndrome. Neurogastroenterol. Motil. 21, 399–410 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Wong, B. S. et al. Pharmacogenetic trial of a cannabinoid agonist shows reduced fasting colonic motility in patients with nonconstipated irritable bowel syndrome. Gastroenterology 141, 1638–1647.e7 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Wong, B. S. et al. Randomized pharmacodynamic and pharmacogenetic trial of dronabinol effects on colon transit in irritable bowel syndrome-diarrhea. Neurogastroenterol. Motil. 24, 358–e169 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Rao, A. S. et al. Chenodeoxycholate in females with irritable bowel syndrome-constipation: a pharmacodynamic and pharmacogenetic analysis. Gastroenterology 139, 1549–1558.e1 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Wong, B. S. et al. Pharmacogenetics of the effects of colesevelam on colonic transit in irritable bowel syndrome with diarrhea. Dig. Dis. Sci. 57, 1222–1226 (2012).

    CAS  PubMed  Google Scholar 

  135. Camilleri, M. et al. Effect of increased bile acid synthesis or fecal excretion in irritable bowel syndrome-diarrhea. Am. J. Gastroenterol. 109, 1621–1630 (2014).

    CAS  PubMed  Google Scholar 

  136. Knowles, C. H., Lindberg, G., Panza, E. & De Giorgio, R. New perspectives in the diagnosis and management of enteric neuropathies. Nat. Rev. Gastroenterol. Hepatol. 10, 206–218 (2013).

    CAS  PubMed  Google Scholar 

  137. Camilleri, M. Peripheral mechanisms in irritable bowel syndrome. N. Engl. J. Med. 367, 1626–1635 (2012).

    CAS  PubMed  Google Scholar 

  138. Ellinghaus, D. et al. Association between variants of PRDM1 and NDP52 and Crohn's disease, based on exome sequencing and functional studies. Gastroenterology 145, 339–347 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Xu, S. et al. Exome sequencing identifies DLG1 as a novel gene for potential susceptibility to Crohn's disease in a Chinese family study. PLoS ONE 9, e99807 (2014).

    PubMed  PubMed Central  Google Scholar 

  140. Fiskerstrand, T. et al. Familial diarrhea syndrome caused by an activating GUCY2C mutation. N. Engl. J. Med. 366, 1586–1595 (2012).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This manuscript has resulted from the collaboration and network activities of the genetics/epigenetics Working Group (WG3) under the frame of the international network GENIEUR (GENes in Irritable Bowel Syndrome Research Network EURope), which is currently funded by the COST (COoperation in Science and Technology) programme (BM1106, www.GENIEUR.eu). We thank A. Farmer, R. Spiller and C. Fischer for fruitful discussion and comments on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

B.N. and M.G. developed the concept, designed, wrote, assembled input data, and edited the manuscript; B.N. and M.M.W. created and revised the figures; A.M., B.N. and L.K.P. summed up the genetics and epigenetics findings in Tables 1 and 2; M.M.W., L.K.P., M.B.B., E.F., G.N., C.A.D., A.M. and J.S. reviewed the literature, selected the data and wrote the manuscript. All authors discussed the results and implications and commented on the manuscript at all stages.

Corresponding author

Correspondence to Beate Niesler.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

FURTHER INFORMATION

GENIEUR

Rome Foundation

PowerPoint slides

Supplementary information

Supplementary Table 1

Summary of genetic association data in IBS. (DOC 180 kb)

Supplementary Table 2

Summary of epigenetic data in IBS. (DOC 47 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gazouli, M., Wouters, M., Kapur-Pojskić, L. et al. Lessons learned — resolving the enigma of genetic factors in IBS. Nat Rev Gastroenterol Hepatol 13, 77–87 (2016). https://doi.org/10.1038/nrgastro.2015.206

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrgastro.2015.206

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