What is the relationship between Crohns disease and inflammatory bowel disease?

Crohn’s disease is an inflammatory bowel disease that causes painful swelling and redness (inflammation) inside the digestive tract. This can lead to symptoms such as abdominal pain and diarrhoea.

Inflammation is a normal way in which the immune system defends the body when fighting off invaders, such as bacteria or viruses. Usually, the inflammation is switched off once the invaders are destroyed. In Crohn’s disease, a problem with the immune system causes the inflammation to continue, damaging the walls of the digestive tract.

What are the symptoms of Crohn’s disease?

Common symptoms include diarrhoea, abdominal pain and cramping, fatigue, reduced appetite and weight loss. Symptoms tend to vary between individuals and come and go over time, as the inflammation flares up then eases again.

Other symptoms may include:

Who gets Crohn’s disease and what causes it?

Crohn’s disease can develop at any age, but usually first appears in people aged 15 to 30.

Experts are not sure why Crohn’s disease occurs in some people. But they don't believe it is caused by diet or stress alone.

A combination of genetic, environmental and infectious factors may cause a fault in the immune system, leading to inflammation of the bowel.

Genetics

Studies have shown that you are slightly more likely to develop Crohn's disease if you have a close relative (mother, father, sister or brother) with the disease.

Researchers have identified several genes that are more common in people with Crohn's disease or colitis, but they have not been able to show that any of these genes actually cause these conditions.

Environment

Crohn's disease is more common in people living in Australia, Western Europe and America than in developing countries in Asia and Africa. Researchers have suggested that a modern, Western lifestyle may play some role in causing Crohn's disease in susceptible people.

Infection

Some scientists think that a previous bacterial infection can trigger an abnormal immune response in some people, leading to Crohn's disease developing. This is still under investigation.

How is Crohn’s disease diagnosed?

Crohn’s disease shares many symptoms with other common conditions, such as ulcerative colitis, irritable bowel syndrome, gastroenteritis and coeliac disease. Your doctor will probably examine you and take a detailed history of your symptoms to help rule these out.

There is no single test that can be used to diagnose Crohn's disease, so a combination of tests is usually required:

  • Blood tests can rule out other medical conditions, and certain 'markers' in the blood can indicate that inflammation is present.
  • A stool sample may find other possible causes of diarrhoea and inflammation, such as an infection.
  • A colonoscopy (or sigmoidoscopy) may be performed, where a thin, flexible tube containing a tiny camera looks inside the bowel for ulcers, inflammation and bleeding.
  • A biopsy (small sample of tissue) may be taken from inside the bowel so a pathologist can examine it under a microscope to look for signs of disease.

Other types of imaging (such as an x-ray) are sometimes used to help in the diagnosis and to help rule out other diseases.

How is Crohn’s disease treated?

Although there is no cure for Crohn’s disease, treatment can help to minimise the impact of the condition on your life. Your doctor or specialist may also recommend a specific diet that suits your situation.

Medications

Crohn’s disease involves periods of ‘relapse’ when the inflammation in the bowel flares up, and periods of ‘remission’ when the inflammation calms down. The aim of treatment is to treat relapses when they occur and give the bowel a chance to heal. Medications are also used to help maintain remission, improve general wellbeing and prevent complications from developing.

Medications commonly used to control inflammation in Crohn’s disease include:

You may also be advised to take medicines that control diarrhoea, relieve pain and supplement your diet (for example, to boost your iron levels, vitamin D and calcium).

Surgery

Surgery for Crohn's disease can remove or widen sections of the bowel that are badly affected by disease. The healthy ends of the bowel are usually re-joined to each other.

Sometimes, a stoma (an artificial opening in the stomach that diverts faeces or urine into a bag) is required if the disease is very severe. It’s normal to feel uneasy about the idea of living with a stoma, but it often greatly improves a person’s quality of life.

Living with Crohn’s disease

With careful management, most people with Crohn’s are able to enjoy life, including work, travel, recreation, sex and having children.

To keep healthy, consider:

  • keeping a food diary to check if there are any foods that make your symptoms worse during a flare-up
  • asking your doctor about supplements if you think you may be malnourished
  • quitting cigarettes, if you have Crohn's disease and smoke. There is evidence that smoking can make Crohn's disease more severe and harder to control
  • exercising regularly to lift your mood and help relieve stress
  • learning relaxation techniques to help manage stress

What are the complications of Crohn’s disease?

A small number of people with Crohn’s can develop inflammation in other parts of the body, such as the liver, skin, joints and eyes.

Regular monitoring by a gastroenterologist, as well as colonoscopies, may help prevent complications from developing. Medications, including steroids and drugs designed to prevent inflammation — and occasionally surgery — may be needed.

Osteoporosis (thinning of the bones) can develop as a side effect of long-term corticosteroid use.

Crohn's disease can cause bowel obstruction. When this happens, symptoms include increasing abdominal pain and cramping, vomiting and bloating. This is a medical emergency that requires a trip to hospital.

If you suspect that you are having a life-threating medical emergency, call triple zero (000) immediately and ask for an ambulance.

Crohn's disease can also cause complications anywhere in the gut, from the mouth to the anus. Complications around the anus can include abscesses (boils), flaps of thickened skin, and fissures.

Fistulas occur in up to 1 in 3 people with Crohn’s disease. These are tunnels, or ‘tracks’, that connect the intestines to other parts of the gastrointestinal tract, other organs or to the outer skin surface. They usually develop in areas of severe scarring and ulceration. A large fistula may require surgery to flush out any contents and promote healing.

What is the difference between Crohn’s disease and ulcerative colitis?

Ulcerative colitis is also an inflammatory bowel disease (IBD). However, the 2 diseases affect the digestive tract in different ways:

  • Crohn’s disease can affect any part of the digestive tract, from the mouth to the anus (back passage) but usually affects the last section of the small bowel (the ileum) and/or the colon. Inflammation can extend into the entire thickness of the bowel wall.
  • Ulcerative colitis only affects the large bowel (colon and/or rectum), and inflammation is only in the surface layers of the bowel lining. It also causes ulcers (tiny sores) to form in the lining of the bowel.

Resources and support

Speak to your doctor or gastroenterologist. To find a health professional near you, use the healthdirect service finder.

For information and support, visit Crohn's & Colitis Australia or call the IBD helpline on 1800 138 029.

For more information about living with a stoma, visit the Australian Council of Stoma Associations.

To find an accredited practising dietitian, who may be able to help manage your Crohn’s disease with diet, visit the Dietitians Association of Australia.

Last reviewed: October 2020

Inflammatory bowel disease (IBD) is a term for two conditions (Crohn’s disease and ulcerative colitis) that are characterized by chronic inflammation of the gastrointestinal (GI) tract. Prolonged inflammation results in damage to the GI tract.

1. Aniwan S, Park SH, Loftus EV Jr.. Epidemiology, Natural History, and Risk Stratification of Crohn's Disease. Gastroenterol Clin North Am. 2017;46(3):463–80. [PubMed] [Google Scholar]

2. Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. 2018;390(10114):2769–78. [PubMed] [Google Scholar]

3. Shouval DS, Rufo PA. The Role of Environmental Factors in the Pathogenesis of Inflammatory Bowel Diseases: A Review. JAMA Pediatr. 2017;171(10):999–1005. [PubMed] [Google Scholar]

4. Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018;11(1):1–10. [PubMed] [Google Scholar]

5. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448(7152):427–34. [PubMed] [Google Scholar]

6. Cader MZ, Kaser A. Recent advances in inflammatory bowel disease: mucosal immune cells in intestinal inflammation. Gut. 2013;62(11):1653–64. [PubMed] [Google Scholar]

7. Spehlmann ME, Begun AZ, Burghardt J, Lepage P, Raedler A, Schreiber S. Epidemiology of inflammatory bowel disease in a German twin cohort: results of a nationwide study. Inflamm Bowel Dis. 2008;14(7):968–76. [PubMed] [Google Scholar]

8. Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of inflammatory bowel disease. Nature. 2011;474(7351):307–17. [PMC free article] [PubMed] [Google Scholar]

9. Orholm M, Munkholm P, Langholz E, Nielsen OH, Sorensen TI, Binder V. Familial occurrence of inflammatory bowel disease. N Engl J Med. 1991;324(2):84–8. [PubMed] [Google Scholar]

10. Tysk C, Lindberg E, Jarnerot G, Floderus-Myrhed B. Ulcerative colitis and Crohn's disease in an unselected population of monozygotic and dizygotic twins. A study of heritability and the influence of smoking. Gut. 1988;29(7):990–6. [PMC free article] [PubMed] [Google Scholar]

11. Ruemmele FM, El Khoury MG, Talbotec C, Maurage C, Mougenot JF, Schmitz J, et al. Characteristics of inflammatory bowel disease with onset during the first year of life. J Pediatr Gastroenterol Nutr. 2006;43(5):603–9. [PubMed] [Google Scholar]

12. Bianco AM, Zanin V, Girardelli M, Magnolato A, Martelossi S, Tommasini A, et al. A common genetic background could explain early-onset Crohn's disease. Med Hypotheses. 2012;78(4):520–2. [PubMed] [Google Scholar]

13. Worthey EA, Mayer AN, Syverson GD, Helbling D, Bonacci BB, Decker B, et al. Making a definitive diagnosis: successful clinical application of whole exome sequencing in a child with intractable inflammatory bowel disease. Genet Med. 2011;13(3):255–62. [PubMed] [Google Scholar]

14. Krieg A, Correa RG, Garrison JB, Le Negrate G, Welsh K, Huang Z, et al. XIAP mediates NOD signaling via interaction with RIP2. Proc Natl Acad Sci U S A. 2009;106(34):14524–9. [PMC free article] [PubMed] [Google Scholar]

15. Glocker EO, Kotlarz D, Boztug K, Gertz EM, Schaffer AA, Noyan F, et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 2009;361(21):2033–45. [PMC free article] [PubMed] [Google Scholar]

16. Uhlig HH. Monogenic diseases associated with intestinal inflammation: implications for the understanding of inflammatory bowel disease. Gut. 2013;62(12):1795–805. [PubMed] [Google Scholar]

17. McGovern DP, Kugathasan S, Cho JH. Genetics of Inflammatory Bowel Diseases. Gastroenterology. 2015;149(5):1163–76 e2. [PMC free article] [PubMed] [Google Scholar]

18. Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47(9):979–86. [PMC free article] [PubMed] [Google Scholar]

19. Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119–24. [PMC free article] [PubMed] [Google Scholar]

20. Hindorff LA, Sethupathy P, Junkins HA, Ramos EM, Mehta JP, Collins FS, et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A. 2009;106(23):9362–7. [PMC free article] [PubMed] [Google Scholar]

21. International Union of Immunological Societies Expert Committee on Primary I, Notarangelo LD, Fischer A, Geha RS, Casanova JL, Chapel H, et al. Primary immunodeficiencies: 2009 update. J Allergy Clin Immunol. 2009;124(6):1161–78. [PMC free article] [PubMed] [Google Scholar]

22. Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol. 2003;3(7):521–33. [PubMed] [Google Scholar]

23. Shaw MH, Kamada N, Warner N, Kim YG, Nunez G. The ever-expanding function of NOD2: autophagy, viral recognition, and T cell activation. Trends Immunol. 2011;32(2):73–9. [PMC free article] [PubMed] [Google Scholar]

24. Kuballa P, Huett A, Rioux JD, Daly MJ, Xavier RJ. Impaired autophagy of an intracellular pathogen induced by a Crohn's disease associated ATG16L1 variant. PLoS One. 2008;3(10):e3391. [PMC free article] [PubMed] [Google Scholar]

25. Travassos LH, Carneiro LA, Ramjeet M, Hussey S, Kim YG, Magalhaes JG, et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat Immunol. 2010;11(1):55–62. [PubMed] [Google Scholar]

26. Cooney R, Baker J, Brain O, Danis B, Pichulik T, Allan P, et al. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med. 2010;16(1):90–7. [PubMed] [Google Scholar]

27. Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, Daly MJ, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006;314(5804):1461–3. [PMC free article] [PubMed] [Google Scholar]

28. Sandborn WJ, Feagan BG, Fedorak RN, Scherl E, Fleisher MR, Katz S, et al. A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn's disease. Gastroenterology. 2008;135(4): 1130–41. [PubMed] [Google Scholar]

29. Allais L, Kerckhof FM, Verschuere S, Bracke KR, De Smet R, Laukens D, et al. Chronic cigarette smoke exposure induces microbial and inflammatory shifts and mucin changes in the murine gut. Environ Microbiol. 2016;18(5):1352–63. [PubMed] [Google Scholar]

30. Monick MM, Powers LS, Walters K, Lovan N, Zhang M, Gerke A, et al. Identification of an autophagy defect in smokers' alveolar macrophages. J Immunol. 2010;185(9):5425–35. [PMC free article] [PubMed] [Google Scholar]

31. Sher ME, Bank S, Greenberg R, Sardinha TC, Weissman S, Bailey B, et al. The influence of cigarette smoking on cytokine levels in patients with inflammatory bowel disease. Inflamm Bowel Dis. 1999;5(2):73–8. [PubMed] [Google Scholar]

32. Hou JK, Abraham B, El-Serag H. Dietary intake and risk of developing inflammatory bowel disease: a systematic review of the literature. Am J Gastroenterol. 2011;106(4):563–73. [PubMed] [Google Scholar]

33. Jowett SL, Seal CJ, Pearce MS, Phillips E, Gregory W, Barton JR, et al. Influence of dietary factors on the clinical course of ulcerative colitis: a prospective cohort study. Gut. 2004;53(10):1479–84. [PMC free article] [PubMed] [Google Scholar]

34. Ananthakrishnan AN, Khalili H, Konijeti GG, Higuchi LM, de Silva P, Fuchs CS, et al. Long-term intake of dietary fat and risk of ulcerative colitis and Crohn's disease. Gut. 2014;63(5):776–84. [PMC free article] [PubMed] [Google Scholar]

35. Ananthakrishnan AN, Khalili H, Konijeti GG, Higuchi LM, de Silva P, Korzenik JR, et al. A prospective study of long-term intake of dietary fiber and risk of Crohn's disease and ulcerative colitis. Gastroenterology. 2013;145(5):970–7. [PMC free article] [PubMed] [Google Scholar]

36. Vinolo MA, Rodrigues HG, Nachbar RT, Curi R. Regulation of inflammation by short chain fatty acids. Nutrients. 2011;3(10):858–76. [PMC free article] [PubMed] [Google Scholar]

37. Shaw SY, Blanchard JF, Bernstein CN. Association between the use of antibiotics in the first year of life and pediatric inflammatory bowel disease. Am J Gastroenterol. 2010;105(12):2687–92. [PubMed] [Google Scholar]

38. Selby W, Pavli P, Crotty B, Florin T, Radford-Smith G, Gibson P, et al. Two-year combination antibiotic therapy with clarithromycin, rifabutin, and clofazimine for Crohn's disease. Gastroenterology. 2007;132(7):2313–9. [PubMed] [Google Scholar]

39. Olszak T, An D, Zeissig S, Vera MP, Richter J, Franke A, et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science. 2012;336(6080):489–93. [PMC free article] [PubMed] [Google Scholar]

40. Khalili H, Higuchi LM, Ananthakrishnan AN, Richter JM, Feskanich D, Fuchs CS, et al. Oral contraceptives, reproductive factors and risk of inflammatory bowel disease. Gut. 2013;62(8):1153–9. [PMC free article] [PubMed] [Google Scholar]

41. Ananthakrishnan AN, Higuchi LM, Huang ES, Khalili H, Richter JM, Fuchs CS, et al. Aspirin, nonsteroidal anti-inflammatory drug use, and risk for Crohn disease and ulcerative colitis: a cohort study. Ann Intern Med. 2012;156(5):350–9. [PMC free article] [PubMed] [Google Scholar]

42. Kvasnovsky CL, Aujla U, Bjarnason I. Nonsteroidal anti-inflammatory drugs and exacerbations of inflammatory bowel disease. Scand J Gastroenterol. 2015;50(3):255–63. [PubMed] [Google Scholar]

43. Bager P, Simonsen J, Nielsen NM, Frisch M. Cesarean section and offspring's risk of inflammatory bowel disease: a national cohort study. Inflamm Bowel Dis. 2012;18(5):857–62. [PubMed] [Google Scholar]

44. Ng SC, Tang W, Leong RW, Chen M, Ko Y, Studd C, et al. Environmental risk factors in inflammatory bowel disease: a population-based case-control study in Asia-Pacific. Gut. 2015;64(7):1063–71. [PubMed] [Google Scholar]

45. Chu KM, Watermeyer G, Shelly L, Janssen J, May TD, Brink K, et al. Childhood helminth exposure is protective against inflammatory bowel disease: a case control study in South Africa. Inflamm Bowel Dis. 2013;19(3):614–20. [PubMed] [Google Scholar]

46. Cholapranee A, Ananthakrishnan AN. Environmental Hygiene and Risk of Inflammatory Bowel Diseases: A Systematic Review and Meta-analysis. Inflamm Bowel Dis. 2016;22(9):2191–9. [PMC free article] [PubMed] [Google Scholar]

47. Sartor RB, Wu GD. Roles for Intestinal Bacteria, Viruses, and Fungi in Pathogenesis of Inflammatory Bowel Diseases and Therapeutic Approaches. Gastroenterology. 2017;152(2):327–39 e4. [PMC free article] [PubMed] [Google Scholar]

48. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2007;104(34):13780–5. [PMC free article] [PubMed] [Google Scholar]

49. Peterson DA, Frank DN, Pace NR, Gordon JI. Metagenomic approaches for defining the pathogenesis of inflammatory bowel diseases. Cell Host Microbe. 2008;3(6):417–27. [PMC free article] [PubMed] [Google Scholar]

50. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500(7461):232–6. [PubMed] [Google Scholar]

51. Varela E, Manichanh C, Gallart M, Torrejon A, Borruel N, Casellas F, et al. Colonisation by Faecalibacterium prausnitzii and maintenance of clinical remission in patients with ulcerative colitis. Aliment Pharmacol Ther. 2013;38(2):151–61. [PubMed] [Google Scholar]

52. Fujimoto T, Imaeda H, Takahashi K, Kasumi E, Bamba S, Fujiyama Y, et al. Decreased abundance of Faecalibacterium prausnitzii in the gut microbiota of Crohn's disease. J Gastroenterol Hepatol. 2013;28(4):613–9. [PubMed] [Google Scholar]

53. Takahashi K, Nishida A, Fujimoto T, Fujii M, Shioya M, Imaeda H, et al. Reduced Abundance of Butyrate-Producing Bacteria Species in the Fecal Microbial Community in Crohn's Disease. Digestion. 2016;93(1):59–65. [PubMed] [Google Scholar]

54. Ahmed I, Roy BC, Khan SA, Septer S, Umar S. Microbiome, Metabolome and Inflammatory Bowel Disease. Microorganisms. 2016;4(2). [PMC free article] [PubMed] [Google Scholar]

55. van der Waaij LA, Harmsen HJ, Madjipour M, Kroese FG, Zwiers M, van Dullemen HM, et al. Bacterial population analysis of human colon and terminal ileum biopsies with 16S rRNA-based fluorescent probes: commensal bacteria live in suspension and have no direct contact with epithelial cells. Inflamm Bowel Dis. 2005;11(10):865–71. [PubMed] [Google Scholar]

56. Schultsz C, Van Den Berg FM, Ten Kate FW, Tytgat GN, Dankert J. The intestinal mucus layer from patients with inflammatory bowel disease harbors high numbers of bacteria compared with controls. Gastroenterology. 1999;117(5):1089–97. [PubMed] [Google Scholar]

57. Png CW, Linden SK, Gilshenan KS, Zoetendal EG, McSweeney CS, Sly LI, et al. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol. 2010;105(11):2420–8. [PubMed] [Google Scholar]

58. Soderholm JD, Olaison G, Peterson KH, Franzen LE, Lindmark T, Wiren M, et al. Augmented increase in tight junction permeability by luminal stimuli in the non-inflamed ileum of Crohn's disease. Gut. 2002;50(3):307–13. [PMC free article] [PubMed] [Google Scholar]

59. Buhner S, Buning C, Genschel J, Kling K, Herrmann D, Dignass A, et al. Genetic basis for increased intestinal permeability in families with Crohn's disease: role of CARD15 3020insC mutation? Gut. 2006;55(3):342–7. [PMC free article] [PubMed] [Google Scholar]

60. Irvine EJ, Marshall JK. Increased intestinal permeability precedes the onset of Crohn's disease in a subject with familial risk. Gastroenterology. 2000;119(6):1740–4. [PubMed] [Google Scholar]

61. May GR, Sutherland LR, Meddings JB. Is small intestinal permeability really increased in relatives of patients with Crohn's disease? Gastroenterology. 1993; 104(6) :1627–32. [PubMed] [Google Scholar]

62. Muise AM, Walters TD, Glowacka WK, Griffiths AM, Ngan BY, Lan H, et al. Polymorphisms in E-cadherin (CDH1) result in a mis-localised cytoplasmic protein that is associated with Crohn's disease. Gut. 2009;58(8):1121–7. [PubMed] [Google Scholar]

63. Gassler N, Rohr C, Schneider A, Kartenbeck J, Bach A, Obermuller N, et al. Inflammatory bowel disease is associated with changes of enterocytic junctions. Am J Physiol Gastrointest Liver Physiol. 2001;281(1):G216–28. [PubMed] [Google Scholar]

64. Darsigny M, Babeu JP, Dupuis AA, Furth EE, Seidman EG, Levy E, et al. Loss of hepatocyte-nuclear-factor-4alpha affects colonic ion transport and causes chronic inflammation resembling inflammatory bowel disease in mice. PLoS One. 2009;4(10):e7609. [PMC free article] [PubMed] [Google Scholar]

65. Pabst O, Zweigerdt R, Arnold HH. Targeted disruption of the homeobox transcription factor Nkx2-3 in mice results in postnatal lethality and abnormal development of small intestine and spleen. Development. 1999;126(10):2215–25. [PubMed] [Google Scholar]

66. van der Flier LG, Clevers H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu Rev Physiol. 2009;71:241–60. [PubMed] [Google Scholar]

67. Johansson ME, Ambort D, Pelaseyed T, Schutte A, Gustafsson JK, Ermund A, et al. Composition and functional role of the mucus layers in the intestine. Cell Mol Life Sci. 2011;68(22):3635–41. [PubMed] [Google Scholar]

68. Van der Sluis M, De Koning BA, De Bruijn AC, Velcich A, Meijerink JP, Van Goudoever JB, et al. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology. 2006;131(1):117–29. [PubMed] [Google Scholar]

69. Herbert DR, Yang JQ, Hogan SP, Groschwitz K, Khodoun M, Munitz A, et al. Intestinal epithelial cell secretion of RELM-beta protects against gastrointestinal worm infection. J Exp Med. 2009;206(13):2947–57. [PMC free article] [PubMed] [Google Scholar]

70. McDole JR, Wheeler LW, McDonald KG, Wang B, Konjufca V, Knoop KA, et al. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature. 2012;483(7389):345–9. [PMC free article] [PubMed] [Google Scholar]

71. McVay LD, Keilbaugh SA, Wong TM, Kierstein S, Shin ME, Lehrke M, et al. Absence of bacterially induced RELMbeta reduces injury in the dextran sodium sulfate model of colitis. J Clin Invest. 2006;116(11):2914–23. [PMC free article] [PubMed] [Google Scholar]

72. Itoh H, Beck PL, Inoue N, Xavier R, Podolsky DK. A paradoxical reduction in susceptibility to colonic injury upon targeted transgenic ablation of goblet cells. J Clin Invest. 1999;104(11):1539–47. [PMC free article] [PubMed] [Google Scholar]

73. Salzman NH, Underwood MA, Bevins CL. Paneth cells, defensins, and the commensal microbiota: a hypothesis on intimate interplay at the intestinal mucosa. Semin Immunol. 2007;19(2):70–83. [PubMed] [Google Scholar]

74. Fritz T, Niederreiter L, Adolph T, Blumberg RS, Kaser A. Crohn's disease: NOD2, autophagy and ER stress converge. Gut. 2011;60(11):1580–8. [PMC free article] [PubMed] [Google Scholar]

75. Hampe J, Franke A, Rosenstiel P, Till A, Teuber M, Huse K, et al. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nat Genet. 2007;39(2):207–11. [PubMed] [Google Scholar]

76. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132(1):27–42. [PMC free article] [PubMed] [Google Scholar]

77. Cadwell K, Liu JY, Brown SL, Miyoshi H, Loh J, Lennerz JK, et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature. 2008;456(7219):259–63. [PMC free article] [PubMed] [Google Scholar]

78. Smythies LE, Sellers M, Clements RH, Mosteller-Barnum M, Meng G, Benjamin WH, et al. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Invest. 2005;115(1):66–75. [PMC free article] [PubMed] [Google Scholar]

79. Smith PD, Ochsenbauer-Jambor C, Smythies LE. Intestinal macrophages: unique effector cells of the innate immune system. Immunol Rev. 2005;206:149–59. [PubMed] [Google Scholar]

80. Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol. 2007;8(10):1086–94. [PubMed] [Google Scholar]

81. Smith AM, Rahman FZ, Hayee B, Graham SJ, Marks DJ, Sewell GW, et al. Disordered macrophage cytokine secretion underlies impaired acute inflammation and bacterial clearance in Crohn's disease. J Exp Med. 2009;206(9):1883–97. [PMC free article] [PubMed] [Google Scholar]

82. Kamada N, Hisamatsu T, Okamoto S, Chinen H, Kobayashi T, Sato T, et al. Unique CD14 intestinal macrophages contribute to the pathogenesis of Crohn disease via IL-23/IFN-gamma axis. J Clin Invest. 2008;118(6):2269–80. [PMC free article] [PubMed] [Google Scholar]

83. Rescigno M Intestinal dendritic cells. Adv Immunol. 2010;107:109–38. [PubMed] [Google Scholar]

84. Niess JH, Brand S, Gu X, Landsman L, Jung S, McCormick BA, et al. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science. 2005;307(5707):254–8. [PubMed] [Google Scholar]

85. Medina-Contreras O, Geem D, Laur O, Williams IR, Lira SA, Nusrat A, et al. CX3CR1 regulates intestinal macrophage homeostasis, bacterial translocation, and colitogenic Th17 responses in mice. J Clin Invest. 2011;121(12):4787–95. [PMC free article] [PubMed] [Google Scholar]

86. Hart AL, Al-Hassi HO, Rigby RJ, Bell SJ, Emmanuel AV, Knight SC, et al. Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology. 2005;129(1):50–65. [PubMed] [Google Scholar]

87. Uhlig HH, McKenzie BS, Hue S, Thompson C, Joyce-Shaikh B, Stepankova R, et al. Differential activity of IL-12 and IL-23 in mucosal and systemic innate immune pathology. Immunity. 2006;25(2):309–18. [PubMed] [Google Scholar]

88. Arseneau KO, Cominelli F. Targeting leukocyte trafficking for the treatment of inflammatory bowel disease. Clin Pharmacol Ther. 2015;97(1):22–8. [PMC free article] [PubMed] [Google Scholar]

89. Danese S, Panes J. Development of drugs to target interactions between leukocytes and endothelial cells and treatment algorithms for inflammatory bowel diseases. Gastroenterology. 2014;147(5):981–9. [PubMed] [Google Scholar]

90. Jovani M, Danese S. Vedolizumab for the treatment of IBD: a selective therapeutic approach targeting pathogenic a4b7 cells. Curr Drug Targets. 2013;14(12):1433–43. [PubMed] [Google Scholar]

91. Vermeire S, O'Byrne S, Keir M, Williams M, Lu TT, Mansfield JC, et al. Etrolizumab as induction therapy for ulcerative colitis: a randomised, controlled, phase 2 trial. Lancet. 2014;384(9940):309–18. [PubMed] [Google Scholar]

92. Vermeire S, Ghosh S, Panes J, Dahlerup JF, Luegering A, Sirotiakova J, et al. The mucosal addressin cell adhesion molecule antibody PF-00547,659 in ulcerative colitis: a randomised study. Gut. 2011;60(8):1068–75. [PubMed] [Google Scholar]

93. Sandborn WJ, Feagan BG, Wolf DC, D'Haens G, Vermeire S, Hanauer SB, et al. Ozanimod Induction and Maintenance Treatment for Ulcerative Colitis. N Engl J Med. 2016;374(18):1754–62. [PubMed] [Google Scholar]

94. Fuss IJ, Neurath M, Boirivant M, Klein JS, de la Motte C, Strong SA, et al. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn's disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J Immunol. 1996;157(3):1261–70. [PubMed] [Google Scholar]

95. Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunol. 2014;14(5):329–42. [PubMed] [Google Scholar]

96. Danese S, Rudzinski J, Brandt W, Dupas JL, Peyrin-Biroulet L, Bouhnik Y, et al. Tralokinumab for moderate-to-severe UC: a randomised, double-blind, placebo- controlled, phase IIa study. Gut. 2015;64(2):243–9. [PubMed] [Google Scholar]

97. Reinisch W, Panes J, Khurana S, Toth G, Hua F, Comer GM, et al. Anrukinzumab, an anti-interleukin 13 monoclonal antibody, in active UC: efficacy and safety from a phase IIa randomised multicentre study. Gut. 2015;64(6): 894–900. [PubMed] [Google Scholar]

98. Nanau RM, Neuman MG. Metabolome and inflammasome in inflammatory bowel disease. Transl Res. 2012;160(1):1–28. [PubMed] [Google Scholar]

99. Oppmann B, Lesley R, Blom B, Timans JC, Xu Y, Hunte 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(5):715–25. [PubMed] [Google Scholar]

100. Teng MW, Bowman EP, McElwee JJ, Smyth MJ, Casanova JL, Cooper AM, et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med. 2015;21(7):719–29. [PubMed] [Google Scholar]

101. Feagan BG, Sandborn WJ, Gasink C, Jacobstein D, Lang Y, Friedman JR, et al. Ustekinumab as Induction and Maintenance Therapy for Crohn's Disease. N Engl J Med. 2016;375(20):1946–60. [PubMed] [Google Scholar]

102. Panaccione R, Sandborn WJ, Gordon GL, Lee SD, Safdi A, Sedghi S, et al. Briakinumab for treatment of Crohn's disease: results of a randomized trial. Inflamm Bowel Dis. 2015;21(6):1329–40. [PMC free article] [PubMed] [Google Scholar]

103. Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, Reinisch W, Higgins PD, et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut. 2012;61(12):1693–700. [PMC free article] [PubMed] [Google Scholar]

104. Weaver CT, Elson CO, Fouser LA, Kolls JK. The Th17 pathway and inflammatory diseases of the intestines, lungs, and skin. Annu Rev Pathol. 2013;8:477–512. [PMC free article] [PubMed] [Google Scholar]

105. Zhou L, Lopes JE, Chong MM, Ivanov II, Min R, Victora GD, et al. TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature. 2008;453(7192):236–40. [PMC free article] [PubMed] [Google Scholar]

106. Ghoreschi K, Laurence A, Yang XP, Tato CM, McGeachy MJ, Konkel JE, et al. Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling. Nature. 2010;467(7318):967–71. [PMC free article] [PubMed] [Google Scholar]

107. Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474(7351):298–306. [PubMed] [Google Scholar]

108. Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, et al. X- linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27(1): 18–20. [PubMed] [Google Scholar]

109. Yu QT, Saruta M, Avanesyan A, Fleshner PR, Banham AH, Papadakis KA. Expression and functional characterization of FOXP3+ CD4+ regulatory T cells in ulcerative colitis. Inflamm Bowel Dis. 2007;13(2):191–9. [PubMed] [Google Scholar]

110. Himmel ME, Yao Y, Orban PC, Steiner TS, Levings MK. Regulatory T-cell therapy for inflammatory bowel disease: more questions than answers. Immunology. 2012; 136(2):115–22. [PMC free article] [PubMed] [Google Scholar]

111. Galien R Janus kinases in inflammatory bowel disease: Four kinases for multiple purposes. Pharmacol Rep. 2016;68(4):789–96. [PubMed] [Google Scholar]

112. O'Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med. 2015;66:311–28. [PMC free article] [PubMed] [Google Scholar]

113. Sandborn WJ, Su C, Sands BE, D'Haens GR, Vermeire S, Schreiber S, et al. Tofacitinib as Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med. 2017;376(18):1723–36. [PubMed] [Google Scholar]

114. Paramsothy S, Paramsothy R, Rubin DT, Kamm MA, Kaakoush NO, Mitchell HM, et al. Faecal Microbiota Transplantation for Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. J Crohns Colitis. 2017;11(10):1180–99. [PubMed] [Google Scholar]

115. Dave M, Mehta K, Luther J, Baruah A, Dietz AB, Faubion WA Jr.. Mesenchymal Stem Cell Therapy for Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. Inflamm Bowel Dis. 2015;21(11):2696–707. [PMC free article] [PubMed] [Google Scholar]


Page 2

What is the relationship between Crohns disease and inflammatory bowel disease?

Mechanisms involved in the pathogenesis of Inflammatory Bowel Disease.

IBD pathogenesis is a result of the interplay between genetic, environmental, intestinal barrier and immune response factors. Abbreviations: GWAS, Genome-wide association studies; IBD, Inflammatory Bowel Diseases.

  • What is the relationship between Crohns disease and inflammatory bowel disease?
  • What is the relationship between Crohns disease and inflammatory bowel disease?

Click on the image to see a larger version.