eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3/2012
vol. 37
 
Share:
Share:

Review paper
Dual role of Th17 cells in Crohn’s disease

Jacek Karczewski
,
Maciej Mazur
,
Marek Karczewski

(Centr Eur J Immunol 2012; 37 (3): 286-289)
Online publish date: 2012/10/27
Article file
- Dual role of Th17.pdf  [0.09 MB]
Get citation
 
PlumX metrics:
 

Introduction

Crohn’s disease (CD) is a chronic relapsing, destructive inflammatory disease of the gastrointestinal (GI) tract, the cause of which remains still unknown. In Europe, CD incidence rates vary between 0.7 (Croatia) and 9.8 (Scotland) cases per 100 000 persons-years, while prevalence ranges from 8.3 (Croatia) to 214 (UK) [1]. Undoubtedly, CD is of multifactorial nature, involving genetic predispositions, environmental factors, intestinal microbal flora and the immune system. Accumulating evidence suggests that CD results from the imbalance between pro- and anti-inflammatory agents and the overactivity of T helper (Th)1 and possibly Th17 cells [2]. Some data also suggest that the defect within the regulatory T cell compartment might be at least partially responsible for the imbalance [3]. This mini-review is aimed to summarize the current knowledge on the potential role of Th17 cells in the pathogenesis of CD.

Th1/Th2 paradigm in CD

The current view on CD is that inflammation is caused by an IL-12-driven Th1 response, which results in the generation of interferon  (IFN-), the key inflammatory mediator [2]. The predominance of Th1 response is consistent with the presence of granulomas in CD patients, which is a well-described outcome of Th1-related immune response [4].

This view has also been supported by multiple IBD mouse models of CD [1]. A large number of CD4+ T cells that highly express T-bet and STAT-4, the Th1-master regulators, have been found in the intestinal mucosa of CD patients [5, 6]. CD4+ T cells in the intestinal lamina propria of CD patients produce large amount of IFN- and lower amount of IL-4 than that of healthy control [6]. Macrophages in the intestinal lamina propria of CD patients produce large amount of IL-12 [7]. Lymphocytes in the intestinal lamina propria of CD patients express elevated levels of IL-12R, and produce large amount of IFN- in response to IL-12 [8]. Similar picture can be found in the peripheral blood of CD patients [9]. The most convincing evidence that CD is a Th1-cytokine mediated disease, however, is the efficacy of biologic therapies directed against tumor necrosis factor (TNF)-, IL-12 and IFN- [10].

Th17 cells

Recently, much attention has also been given to the possible involvement of Th17 cells in the pathogenesis of CD [11, 12]. This newly identified subset of T-helper cells distinct from Th1/Th2 produce various proinflammatory cytokines, including IL-17A, IL-17F, IL-21, IL-22 and TNF- and play an important role in the protective immunity against the extracellular pathogens such as bacteria and fungi [13]. Th17 cells differentiate in the presence of IL-6 and transforming growth factor (TGF)-, through the expression of retinoic-acid-receptor-related orphan receptor (ROR)-t [13]. While TGF- drives Smad signaling, IL-6 activates STAT3 pathway, which promotes the expression of RORt, a master regulator of Th17 cells. IL-21, in turn, seems to be an important cytokine for the differentiation of Th17 cells. IL-6 promotes the production of IL-21 from Th17 cells independent of RORt, and then IL-21 upregulates the expression of RORt through the activation of STAT3 pathway. This process provides a positive feedback in the differentiation of Th17 cells, and is called an amplification. Although IL-21 is not essential for the polarization of Th17 cells, it is necessary for their amplification. The key cytokine responsible for stabilization and expansion of Th17 cells, in turn, is the member of IL-12 cytokine, IL-23. This cytokine is produced by professional antigen-presenting cells (APCs) during the inflammatory response to both pathogenic and nonpathogenic stimuli [14]. Also intestinal microbiota is essential for the development of Th17 cells, since mice in germ-free condition are characterized by lower percentage of this subset [15]. Th17 cells play a crucial role for the protective immunity against intestinal pathogens. A high expression of IL-23 has been found in mucosa of human ileum, as well as an upregulation of Th17 cells in the human GALT [16].

Potential role of Th17 cells in CD

IL-17A, often referred to as IL-17, is a main proinflammatory cytokine produced by Th17 cells. It has been reported that Th17 subset is deeply involved in the pathogenesis of various diseases that were thought to be either Th1 or Th2 dominant previously. IL-17A is upregulated in and has been associated with several autoimmune diseases including inflammatory bowel disease (comprising CD and UC), rheumatoid arthritis, psoriasis, psoriatic arthritis, ankylosing spondylitis and multiple sclerosis [11]. Its involvement in other pathological inflammatory conditions like uveitis, cystic fibrosis, chronic obstructive pulmonary disease and other inflammatory airway diseases, allograft rejection, cancer, and intraperitoneal abscesses and adhesions has been also implied [11]. Mounting evidence seems to support the view that Th17 cells are somehow involved in CD pathogenesis. A high expression of IL-17A mRNA was found in intestinal mucosa of CD patients [17]. The elevated fecal IL-17A levels were found in active CD, accompanied by high percentage of IL-17A and IL-23 producing cells in lamina priopria of CD patients [12]. Several mouse studies have identified IL-23 as a major driver of intestinal inflammation via inflammatory mediators including IL-17A and IL-6 [17-19]. A genome-wide association study (GWAS) has indicated that IL-23R and five additional genes involved in Th17 differentiation are associated with susceptibility to CD [17]. It is not clear how IL-23R polymorphism might predispose to CD, however the identification of both disease-protective and risk-associated variants of the gene suggests that the IL-23 signaling may play a crucial role in maintaining immune homeostasis in the intestine. Also, recently conducted phase II clinical trials have reported that blockage of p40, the shared subunit of IL-12 and IL-23, is effective in CD treatment, and has a therapeutic effect similar to that of anti-TNF- therapy [20, 21]. The anti-p40 antibodies were assumed to abrogate IFN- mediated intestinal inflammation through blockage of

IL-12, however the other possibility implies that inflammation attributed to IL-12 is mediated by IL-23 and its downstream cytokines IL-17A and IL-22. All these data have provided support for the idea that IL-17A plays a major role in CD pathogenesis and its inhibition might represent a potential approach for treating active course of the disease.

Although these observations suggest the importance of the IL-23/IL-17 axis in CD, the exact role of IL-17 in the pathogenesis of disease remains unclear, since also its protective role in the intestine inflammation has been proposed based on T cell dependent and T cell independent models of colitis [22, 23]. This view is supported by the poor therapeutic effect accompanied by a higher rate of adverse events compared to placebo group obtained in recent phase II clinical trial of anti-IL-17 therapy in active CD patients [24]. It should be remembered that Th17 cells constitute a heterogonous subpopulation, differing in properties and functions [2]. There is evidence that differentiation of Th17 cells in the absence of IL-23 leads to Th17 cells producing a strong anti-inflammatory cytokine, IL-10 [25]. Th17 cells induced in the presence of IL-1, on the contrary, have a unique mRNA profile and increased capacity to induce inflammation [26]. In milieu lacking TGF-, IL-12 and

IL-23 Th17 cells acquire the capacity to produce IFN- [2].

Of great importance for immune homeostasis is also a somewhat reciprocal relationship of Th17 cells with regulatory T cells through their shared use of TGF- as a differentiation factor [27-29]. Current data demonstrate that active CD is, like many other autoimmune diseases, characterized by a numerical and/or functional defect of a regulatory T cell subpopulation [9, 30, 31]. In spite of the presence in the inflamed GI tract of CD patients, their number and/or suppression function is probably insufficient to control an ongoing inflammation [30]. It is suspected that generation of these cells is inhibited by active blocking by Th17 cells and chronic overproduction of Th17 cytokines [14]. It is also worth noting that in some circumstances Th17 cells can acquire strong immunosuppressive capabilities [32]. These regulatory Th17 cells are IL-17A+ FoxP3- and suppressive capacity is dependent on IL-10, TGF- and cytotoxic T lymphocyte antigen 4 (CTLA4) [32]. Another study has reported the isolation of regulatory Th17 cells expressing Foxp3 and exhibiting strong suppressive activity from the lamina propria of CD patients [33]. There is also evidence that regulatory T cells and Th17 cells may play complementary roles in the intestinal homeostasis. In vitro studies have indicated that regulatory T cells can promote the development of Th17 cells in a TGF--dependent manner [34, 35]. Regulatory T cells themselves can be induced to secrete IL-17 in vitro [35].

Conclusions

Standard treatment of CD includes immunosuppression with corticosteroids, thiopurines, or methotrexate, and anti-TNF therapy for patients with persistent disease activity [36]. Although most CD patients respond to anti-TNF therapy, secondary failures due to intolerance or loss of initial response are common [37, 38]. Thus, novel therapies based on different mechanisms are needed. In spite of the fact that various clinical trials with antibodies to IL-17A have reported therapeutic benefit in multiple patients populations, especially with rheumatoid arthritis and psoriasis, the poor outcome obtained in recent clinical trial in active CD patients indicate that the role of Th17 cells and IL-17A might be much more complex and unique in the intestine. It appears that the local cytokine milieu at site of inflammation is the key factor in the development of Th17 cells and their conversion into distinct subtypes characterized by different levels of proinflammatory activity and playing potentially opposing roles. It seems to be particularly relevant to the immune homeostasis of GI tract. Therefore, further research is needed to elucidate the role of Th17 cells and its downstream cytokines in the pathogenesis of CD.

References

 1. Tersigni R, Prantera C (2010). Crohn's disease. A multidisciplinary approach. 1st ed. Springer-Verlag, Milan.

 2. Strober W, Fuss IJ (2011). Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroentero-logy; 140: 1756-1767.

 3. Allez M, Mayer L (2004): Regulatory T cells. Peace keeprs in the gut. Inflamm Bowel Dis 2004; 10: 666-676.

 4. Targan SR, Shanahan F, Karp LC (2010): Inflammatory Bowel Disease: Translating Basic Science into Clinical Practice.

5 ed: Wiley-Blackwell.

 5. Neurath MF, Weigmann B, Finotto S, et al. (2002): The transcription factor T-bet regulates mucosal T cell activation in experimental colitis and Crohn's disease. J Exp Med 195: 1129-1143.

 6. Fuss IJ, Neurath M, Boirivant M, et al. (1996): 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 157: 1261-1270.

 7. Monteleone G, Biancone L, Marasco R, et al. (1997): Interleukin 12 is expressed and actively released by Crohn's disease intestinal lamina propria mononuclear cells. Gastroenterology 112: 1169-1178.

 8. Okazawa A, Kanai T, Watanabe M, et al. (2002): Th1-mediated intestinal inflammation in Crohn's disease may be induced by activation of lamina propria lymphocytes through synergistic stimulation of interleukin-12 and interleukin-18 without T cell receptor engagement. Am J Gastroenterol 2002; 97: 3108-3117.

 9. Karczewski J, Karczewski M (2011): Immunoregulatory defect in patients with active Crohn's disease. Inflamm Bowel Dis, doi: 10.1002/ibd.21934.

10. Rutgeerts P, Vermeire S, Van Assche G (2009): Biological therapies for inflammatory bowel diseases. Gastroenterology 136: 1182-1197.

11. Witkowski J, Ksiazek K, Jorres A (2004): Interleukin-17: a mediator of inflammatory responses. Cell Mol Life Sci 61: 567-579.

12. Holtta V, Klemetti P, Sipponen T, et al. (2008): IL-23/IL-17 immunity as a hallmark of Crohn's disease. Inflammatory Bowel Disease 14: 1175-1184.

13. Weaver CT, Hatton RD, Mangan PR, Harrington LE (2007): IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol 25: 821-852.

14. Abraham C, Cho J (2009): Interleukin-23/Th17 pathways and inflammatory bowel disease. Inflammatory Bowel Disease 15: 1090-1100.

15. Atarashi K, Nishimura J, Shima T, et al. (2008): ATP drives lamina propria T(H)17 cell differentiation. Nature 455: 808-812.

16. Maloy KJ, Kullberg MC (2008): IL-23 and Th17 cytokines in intestinal homeostasis. Mucosal Immunology 1: 339-349.

17. Brand S (2009): 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 58: 1152-1167.

18. Yen D, Cheung J, Scheerens H, et al. (2006): IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6. J Clin Invest 2006; 116: 1310-1316.

19. Elson CO, Cong Y, Weaver CT, et al. (2007): Monoclonal anti-interleukin 23 reverses active colitis in a T cell-mediated model in mice. Gastroenterology 132: 2359-2370.

20. Mannon PJ, Fuss IJ, Mayer L, et al. (2004): Anti-interleukin-12 antibody for active Crohn's disease. N Engl J Med 351: 2069-2079.

21. Sandborn WJ, Feagan BG, Fedorak RN, et al. (2008): A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn's disease. Gastroenterology 135: 1130-1141.

22. O'Connor W, Kamanaka M, Booth CJ, et al. (2009): A protective function for interleukin 17A in T cell-mediated intestinal inflammation. Nature Immunology 10: 603-609.

23. Ogawa A (2004): Neutralization of interleukin-17 aggravates dextran sulfate sodium-induced colitis in mice. Clinical Immunology 110: 55-62.

24. Hueber W, Sands BE, Vandemeulebroecke M, et al. (2011): Inhibition of IL-17A by secukinumab is ineffective for Crohn's disease (CD). 6th European Crohn's and Colitis Organization (ECCO). Dublin.

25. McGeachy MJ, Bak-Jensen KS, Chen Y, et al. (2007): TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nature Immunology 8: 1390-1397.

26. Ghoreschi K, Laurence A, Yang XP, et al. (2010): Generation of pathogenic T(H)17 cells in the absence of TGF- signalling. Nature 467: 967-971.

27. Mangan PR, Harrington LE, O'Quinn DB, et al. (2006): Transforming growth factor-beta induces development of the T(H)17 lineage. Nature 441: 231-234.

28. McGeachy MJ, Cua DJ (2008): Th17 cell differentiation: the long and winding road. Immunity 28: 445-453.

29. Bettelli E, Carrier Y, Gao W, et al. (2006): Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441: 235-238.

30. Maul J, Loddenkemper C, Mundt P, et al. (2005): Peripheral and intestinal regulatory CD4+CD25high T cells in inflammatory bowel disease. Gastroenterology 128: 1868-1878.

31. Veltkamp C, Anstaett M, Wahl K, et al. (2011): Apoptosis of regulatory T lymphocytes is increased in chronic inflammatory bowel disease and reversed by anti-TNF{alpha} treatment. Gut 60: 1345-1353.

32. Esplugues E, Huber S, Gagliani N, et al. (2011): Control of TH17 cells occurs in the small intestine. Nature 475: 514-518.

33. Hovhannisyan Z, Treatman J, Littman DR, Mayer L (2011): Characterization of interleukin-17-producing regulatory T cells in inflamed intestinal mucosa from patients with inflammatory bowel diseases. Gastroenterology 140: 957-965.

34. Veldhoen M, Hocking RJ, Atkins CJ, et al. (2006): TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 24: 179-189.

35. Xu L, Kitani A, Fuss I, Strober W (2007): 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-beta. J Immunol 2007; 178: 6725-6729.

36. Burger D, Travis S (2011): Conventional medical management of inflammatory bowel disease. Gastroenterology 140: 1827-1837.

37. Hanauer SB, Feagan BG, Lichtenstein GR, et al. (2002): Maintenance infliximab for Crohn's disease: the ACCENT I randomised trial. Lancet 359: 1541-1549.

38. Colombel JF, Sandborn WJ, Rutgeerts P, et al. (2007): Adalimumab for maintenance of clinical response and remission in patients with Crohn's disease: the CHARM trial. Gastroenterology 132: 52-65.
Copyright: © 2012 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.

Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.