#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

New knowledge of the pathogenesis of Crohn’s disease


Authors: B. Ambrůzová 1;  M. Rédováihash2 1,2,3 1,2,3
Authors‘ workplace: Advanced Cell Immunotherapy Unit, Lékařská fakulta MU Brno, vedoucí doc. MUDr. Dalibor Valík, Ph. D. 1;  Klinika komplexní onkologické péče, MOÚ Brno, přednosta prof. MUDr. Rostislav Vyzula, CSc. 2;  Výzkumná skupina Molekulární onkologie II – solidní nádory Středoevropského technologického institutu Brno, vedoucí RNDr. Ondřej Slabý, Ph. D. 3;  Gastroenterologické oddělení, MOÚ Brno, vedoucí MUDr. Milana Šachlová, CSc. et Ph. D. 4
Published in: Vnitř Lék 2012; 58(4): 291-298
Category: Reviews

Overview

Crohn’s disease is a complex chronic inflammatory disease of the gastrointestinal tract with multifactorial pathogenesis. Over the recent years, there has been rather a sharp increase in the incidence of Crohn’s disease and, even though this disease had been known for some time, the cause remains unknown. Studies exploring genetic basis of Crohn’s disease have provided new knowledge of the pathogenesis of this disease, suggesting that this may be associated with a failure of mechanisms behind symbiosis of gut microflora and intestinal mucosal immune system. Crohn’s disease seems to be caused by inadequate immune response to intestinal flora in genetically predisposed individuals. Crohn’s disease has been linked to a number of genes. Many of them are related to the modulation of non-specific immune response, defects of which are considered to be key in Crohn’s disease pathogenesis. The aim of this review paper is to summarize the new knowledge on the pathogenesis of Crohn’s disease at the level of polymorphisms of the NOD2, ATG16L1 genes and the IL23-Th17-lymfocytes signalling pathway genes and to consider further research directions in this disease.

Key words:
Crohn’s disease – pathogenesis – NOD2 ATG16L1 – IL23-Th17-lymphocytes


Sources

1. Kozuch PL, Hanauer SB. Treatment of inflammatory bowel disease: a review of medical therapy. World J Gastroenterol 2008; 14: 354–377.

2. Abraham C, Cho JH. Inflammatory Bowel Disease. N Engl J Med 2009; 361: 2066–2078.

3. Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 2007; 447: 661–678.

4. Hampe J, Franke A, Rosenstiel P et al. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet 2007; 39: 207–211.

5. Massey DC, Parkes M. Common pathways in Crohn’s disease and other inflammatory diseases revealed by genomics. Gut 2007; 56: 1489–1492.

6. Rioux JD, Xavier RJ, Taylor KD et al. Genome-wide association study identifies new susceptibility loci for Crohn disease and implicates autophagy in disease pathogenesis. Nat Genet 2007; 39: 596–604.

7. Van Limbergen J, Wilson DC, Satsanagi J. The genetics of Crohn’s disease. Annu Rev Genomics Hum Genet 2009; 10: 89–116.

8. Molodecky NA, Kaplan GG. Environmental Risk Factors for Inflammatory Bowel Disease. Gastroenterol Hepatol 2010; 6: 339–346.

9. Baumgart DC, Carding SR. Inflammatory bowel disease: cause and immunobiology. Lancet 2007; 369: 1627–1640.

10. Rutgeerts P, Goboes K, Peeters M et al. Effect of faecal stream diversion on recurrence of Crohn’s disease in the neoterminal ileum. Lancet 1991; 338: 771–774.

11. Sartor RB. Therapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology 2004; 126: 1620–1633.

12. Chassaing B, Rolhion N, de Vallé A et al. Crohn disease – associated adherent-invasive E. coli bacteria target mouse and human Peyer’s patches via long polar fimbriae. J Clin Invest 2011; 121: 966–997.

13. Henderson P, Van Limbergen JE, Schwarze J et al. Function of intestinal epithelium and its dysregulation in inflammatory bowel disease. Inflamm Bowel Dis 2011; 17: 382–395.

14. Gaudier E, Hoebler C. Physiological role of mucins in the colonic barrier integrity. Gastroenterol Clin Biol 2006; 30: 965–974.

15. Ramasundara M, Leach ST, Lemberg DA et al. Defensins and inflammation: the role of defensins in inflammatory bowel disease. J Gastroenterol Hepatol 2009; 24: 202–208.

16. Wehkamp J, Stange EF, Fellermann K. Defensin-immunology in inflammatory bowel disease. Gastroenterologie Clin Biol 2009; 33 (Suppl 3): 137–144.

17. Kyd JM, Cripps AW. Functional differences between M cells and enterocytes in sampling luminal antigens. Vaccine 2008; 26: 6221–6224.

18. Ismail AS, Behrendt CL, Hooper LV. Reciprocal interactions between commensal bacteria and gamma delta intraepithelial lymphocytes during mucosal injury. J Immunol 2009; 182: 3047–3054.

19. Yoshida M, Claypool SM, Wagner JS et al. Human neonatal Fc receptor mediates transport of IgG into luminal secretions for delivery of antigens to mucosal dendritic cells. Immunity 2004; 20: 769–778.

20. Marchesi J, Shanahan F. The Normal intestinal microbiota. Curr Opin Infect Dis 2007; 20: 508–513.

21. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inlammatory bowel disease. Nature 2007; 448: 427–434.

22. Hugot JP, Chamaillard M, Zouali H et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature 2001; 411: 599–603.

23. Ogura Y, Bonen DK, Inohara N et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature 2001; 411: 603–606.

24. Inohara N, Ogura Y, Fontalba A et al. Host recognition of bacterial muramyl dipeptide mediated through NOD2. Implications for Crohn’s disease. J Biol Chem 2003; 278: 5509–5512.

25. Abraham C, Cho JH. Functional consequence of NOD2 (CARD15) mutations. Inflamm Bowel Dis 2006; 12: 641–650.

26. Tanabe T, Chamaillard M, Oqura Y et al. Regulatory regions and critical residues of NOD2 involved in muramyl dipeptide recognition. EMBO J 2004; 23: 1587–1597.

27. Lees CW, Sastsanagi J. Genetics of inflammatory bowel disease: implications for disease pathogenesis and natural history. Expert Rev Gastroenterol Hepatol 2009; 3: 513–534.

28. van Heel DA, Ghosh S, Butler M et al. Muramyl dipeptide and toll-like receptor sensitivity in NOD2-associated Crohn’s disease. Lancet 2005; 365: 1794–1796.

29. Wehkamp J, Salzman NH, Porter E et al. Reduced Paneth cell α-defensins in ileal Crohn’s disease. Proc Natl Acad Sci USA 2005; 102: 18129–18134.

30. Wehkamp J, Wang G, Kübler I et al. The Paneth cell alpha-defensin deficiency of ileal Crohn’s disease is linked to Wnt/Tcf-4. J Immunol 2007; 179: 3109–3118.

31. Wehkamp J, Stange EF, Fellermann K. Defensin-immunology in inflammatory bowel disease. Gastroenterologie Clin Biol 2009; 33 (Suppl 3): 137–144.

32. Sartor RB. Does Mycobacterium avium subspecies paratuberculosis cause Crohn’s disease? Gut 2005; 54: 896–898.

33. Coulombe F, Divangahi M, Veyrier F et al. Increased NOD2-mediated recognition of N-glycolyl muramyl dipeptide. J Exp Med 2009; 206: 1709–1716.

34. Rahman MK, Midtling EH, Svingen PA et al. The Pathogen Recognition Receptor NOD2 Regulates Human FOXP3+ T Cell Survival. J Immunol 2010; 184: 7247–7256.

35. Jess T, Riis L, Jespersgaard C et al. Disease concordance, zygosity, and NOD2/CARD15 status: follow-up of a population-based cohort of Danish twins with inflammatory bowel disease. Am J Gastroenterol 2005; 100: 2486–2492.

36. Cho JH, Weaver CT. The genetics of inflammatory bowel disease. Gastroenterology 2007; 133: 1327–1339.

37. Hradsky O, Lenicek M, Dusatkova P et al. Variants of CARD15, TNFA and PTPN22 and susceptibility to Crohn’s disease in the Czech population: high frequency of the CARD15 1007fs. Tissue Antigens 2008; 71: 538–547.

38. Lesage S, Zouali H, Cézard JP et al. EPWG-IBD Group; EPIMAD Group; GETAID Group. CARD15/NOD2 mutational analysis and genotype-phenotype correlation in 612 patients with inflammatory bowel disease. Am J Hum Genet 2002; 70: 845–857.

39. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F et al. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004; 118: 229–241.

40. Yamamoto-Furusho JK, Podolsky DK. Innate immunity in inflammatory bowel disease. World J Gastroenterol 2007; 13: 5577.

41. Cario E, Podolsky DK. Differential alteration in intestinal epithelial cell expression of toll-like receptor 3 (TLR3) and TLR4 in inflammatory bowel disease. Infect Immun 2000; 68: 7010–7017.

42. González-Navajas JM, Fine S, Law J et al. TLR4 signaling in effector CD4+ T cells regulates TCR activation and experimental colitis in mice. J Clin Invest 2010; 120: 570–581.

43. Browning BL, Huebner C, Petermann I et al. Has Toll-like receptor 4 been prematurely dismissed as an inflammatory bowel disease gene? Association study combined with meta-analysis shows strong evidence for association. Am J Gastroenterol 2007; 102: 2504–2512.

44. Arbour NC, Lorenz E, Schutte BC et al. TLR4 mutations are associated with endotoxin hyporesponsiveness in humans. Nat Genet 2000; 25: 187–191.

45. Ferwerda B, McCall MB, Alonso S et al. TLR4 polymorphisms, infectious diseases, and evolutionary pressure during migration of modern humans. Proc Natl Acad Sci USA 2007; 104: 16645–16650.

46. Watanabe T, Kitani A, Murray PJ et al. NOD2 is a negative regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat Immunol 2004; 5: 800–808.

47. Kullberg BJ, Ferwerda G, De Jong DJ et al. Crohn’s disease patients homozygous for the 3020insC NOD2 mutation have a defective NOD2/TLR4 cross-tolerance to intestinal stimuli. Immunology 2008; 123: 600–605.

48. Schmitd D, Dengjel J, Schoor O et al. Autophagy in innate and adaptive immunity against intracellular pathogens. J Mol Med 2006; 84: 194–202.

49. Homer CR, Richmond AL, Rebert NA et al. ATG16L1 and NOD2 Interact in an Autophagy-Dependent Antibacterial Pathway Implicated in Crohn’s Disease Pathogenesis. Gastroenterology 2010; 139: 1630–1641.

50. Kuballa P, Huett A, Rioux JD et al. Impaired autophagy of an intracellular pathogen induced by a Crohn’s disease associated ATG16L1 va­riant. PLoS One 2008; 3: e3391.

51. Saitoh T, Fujita N, Jang MH et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature 2008; 456: 264–268.

52. Cadwell K, Liu JY, Brown SL et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature 2008; 456: 259–263.

53. Hussey S, Travassos LH, Jones NL. Autophagy as an emerging dimension of adaptive and innate immunity. Semin Immunol 2009; 21: 233–241.

54. Parronchi P, Romagnani P, Annunziato F et al. Type 1 T-helper cell predominance and interleukin-12 expression in the gut of patients with Crohn’s disease. Am J Pathol 1997; 150: 823–832.

55. Duerr RH, Taylor KD, Brant SR et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 2006; 314: 1461–1463.

56. Wilson NJ, Boniface K, Chan JR et al. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat Immunol 2007; 8: 950–957.

57. Brand S. Crohn’s disease: Th1, Th17 or both? The change of a paradigm: new immunological and genetic insights implicate Th17 cells in the pathogenesis of Crohn’s disease. Gut 2009; 58: 1152–1167.

58. Oppmann B, Lesley R, Blom B et al. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity 2000; 13: 715–725.

59. Sandborn WJ, Feagan BG, Fedorak RN et al. Ustekinumab Crohn’s Disease Study Group. A randomized trial of ustekinumab, a human interleukin-12/23 monoclonal antibody, in pa­tients with moderate-to-severe Crohn’s disease. Gastroenterology 2008; 135: 1130–1141.

60. Monteleone I, Pallone F, Monteleone G. Interleukin-23 and Th17 Cells in the Control of Gut Inflammation. Mediators Inflamm 2009; 2009: 297645.

61. Andoh A, Zhang Z, Inatomi O et al. Interleukin-22, a member of the IL-10 subfamily, induces inflammatory responses in colonic subepithelial myofibroblasts. Gastroenterology 2005; 129: 969–984.

62. Monteleone G, Caruso R, Fina D et al. Control of matrix metalloproteinase production in human intestinal fibroblasts by interleukin 21. Gut 2006; 55: 1774–1780.

63. Liang SC, Tan XY, Luxenberg DP et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med 2006; 203: 2271–2279.

64. Dambacher J, Beigel F, Zitzmann K et al. The role of the novel Th17 cytokine IL-26 in intestinal inflammation. Gut 2009; 58: 1207–1217.

65. Manel N, Unutmaz D, Littman DR. The differentiation of human T(H)-17 cells requires transforming growth factor-β and induction of the nuclear receptor RORγt. Nat Immunol 2008; 9: 641–649.

66. Xu L, Kitani A, Fuss I et al. Cutting edge: regulatory T cells induce CD4+CD25−Foxp3− T cells or are self-induced to become Th17 cells in the absence of exogenous TGF-β. J Immunol 2007; 178: 6725–6729.

67. Laurence A, Tato CM, Davidson TS et al. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity 2007; 26: 371–381.

Labels
Diabetology Endocrinology Internal medicine
Login
Forgotten password

Enter the email address that you registered with. We will send you instructions on how to set a new password.

Login

Don‘t have an account?  Create new account

#ADS_BOTTOM_SCRIPTS#