Elsevier

Clinical Immunology

Volume 148, Issue 2, August 2013, Pages 246-253
Clinical Immunology

Review
The role of IL-17-producing Foxp3+ CD4+ T cells in inflammatory bowel disease and colon cancer

https://doi.org/10.1016/j.clim.2013.05.003Get rights and content

Highlights

  • Foxp3+ TReg cells are able to express RORγt and produce IL-17.

  • IL-17+ TReg cells are accumulated in the inflamed intestinal mucosa.

  • IL-17+ TReg cells have potent T-cell suppressive activity.

  • RORγt+ TReg cells are unable to suppress inflammation.

  • IL-17+/RORγt+ TReg cells are pathogenic TReg cells with proinflammatory properties.

Abstract

The intestinal epithelium and underlying lamina propria contain T cells that play important roles in maintaining colonic homeostasis. These T cells mediate substantial and specific regulation to ensure that pathogenic microorganisms are eliminated while commensal bacteria are tolerated. There is considerable evidence supporting the notion that the altered ratio between Foxp3+CD4+ T regulatory cells and T effector cells in the colonic microenvironment might contribute to the initiation and progression of inflammation and eventually development of colon cancer. Recent findings on the heterogeneity and plasticity of T regulatory cells, such as the identification of IL-17+Foxp3+CD4+ and the RORγt+Foxp3+CD4+ subsets, in patients with colorectal inflammation and cancer have provided a new twist in our understanding of the pathogenesis of colonic diseases. Phenotypic and functional properties of IL-17-producing Foxp3+CD4+ T cells as well as the significant implications of these cells in the initiation and progression of colorectal diseases are discussed in this review.

Introduction

Regulatory T (TReg) cells expressing the transcription factor forkhead box P3 (Foxp3) are naturally present in the immune system. TReg cells suppress the activation, proliferation and effector functions of a wide range of immune cells, including CD4+ and CD8+ T cells, natural killer (NK) and NKT cells, B cells and antigen presenting cells in vitro and in vivo [1]. TReg cells have key roles in the prevention of autoimmune responses and the development of immunopathology as well as in the maintenance of homeostasis. As a double-edged sword, TReg cells can also suppress antitumor immune responses and can favor tumor progression. The unique functional properties of TReg cells make these cells a primary target in the search for new cell-based immunotherapeutic approaches. However, recent studies showed that TReg cells are not a homogeneous and terminally differentiated cell population. Instead, these cells are heterogeneous in gene expression, phenotype and function. In addition, TReg cells are strongly affected by other immune components including effector cells and innate immune cells during initiation and progression of disease. Thus, to achieve the translational goal of applying TReg cells in the treatment of diseases, understanding of the TReg cell immunobiology in the context of various tissues and pathogenesis of various diseases is required. Lymphocytes of mucosal tissues form a relatively autonomous immune subsystem, with specialized adaptations appropriate for this particular microenvironment [2], [3], [4]. The intestinal mucosa is normally maintained in a state of controlled inflammation in which equilibrium exists between protective immunity and tolerance to self-antigen and commensal bacteria [5]. Protective immunity against different classes of pathogens depends on the generation of distinct types of immune responses mediated and coordinated by effector cells specifically TH1, TH2 and TH17. On the other hand, TReg cells are involved in the maintenance of tolerance. A series of observations suggests that TReg cells correlate with poor prognosis in many cancer types, including breast, lung, melanoma, and ovarian carcinoma [6] due to their suppressive effects on anti-tumor immunity. However, several studies have shown that TReg cells are protective in cancer by virtue of their ability to control inflammation in an IL-10 dependent manner [7]. In fact, Foxp3 expression has been indicative of better prognosis in gastric cancer, head and neck, and breast cancer [8], [9], [10]. Recent identification of IL-17-producing Foxp3+CD4+ T cells in IBD, colon cancer and polyposis might provide a potential explanation for the conflicting observations regarding the role of TReg cells in cancer. Furthermore, the advancements in understanding TReg immunobiology locally and systemically and the influence of inflammatory microenvironments on the differentiation of effector cells and on the stability of TReg cells would improve the use of TReg as cell-based immunotherapy.

Section snippets

Heterogeneity of TReg cells

Foxp3+CD4+ TReg cells are not homogeneous in gene expression, phenotype and suppressive function. The transcription factor Foxp3, a master control gene for the development and function of both mouse and human TReg cells [11], [12], [13], [14], is currently the definitive marker of TReg cells. However, Foxp3 can be transiently expressed in activated human T cells. This transient Foxp3 expression in T cells does not enable suppression but, instead, makes separation of TReg cells from activated T

Characterization of in vitro induced IL-17-producing Foxp3+CD4+ T cells

TReg cells are well known to produce IL-10 and TGF-β during activation and these cytokines are considered to be part of the mechanisms via which TReg cells mediate their suppressive function [21]. Recent findings reveal that TReg cells are capable of producing IL-17. There is an intimate link between TH17 cells and TReg cells. Exposure of antigen-activated naïve T cells to TGF-β in vitro results in transcriptional up-regulation of both Foxp3 and RORγt [22]. Furthermore, T cells co-expressing

Roles of TReg cells and TH17 cells in inflammation and colon cancer

The intestinal tract contains large numbers of immune cells involved in the encounter with microbial antigens. Under normal conditions, the lumen of the intestine is covered by a layer of intestinal epithelial cells (IECs), which are joined together by tight junctions forming a protective layer impeding bacterial invasion [34]. Immune responses in the colon take place in three distinct compartments: the lamina propria, the isolated lymphoid follicles (ILFs) scattered throughout the colon, and

Paradoxical roles of IL-17-producing Foxp3+CD4+ cells in anti-inflammatory response and anti-tumor immunity

Inflammatory and tumor tissues often have lower frequency of CD69+ T effector cells but a higher frequency of TH17 cells and TReg cells compared to healthy or untransformed tissues [53]. The mechanisms by which TH17 cells and TReg cells accumulate in such inflammatory and tumor tissues and their roles in the pathogenesis of these diseases remain unsolved. The identification of IL-17+Foxp3+ T cells in inflamed intestinal mucosa of patients with IBD and cancer but not in healthy control might

Conclusions

The immunobiology of TReg cells has been intensively investigated since the discovery of TReg cells and their master transcription factor Foxp3, more than a decade ago. Such efforts have led to significant advances in our understanding of the development of TReg cells and the molecular mechanisms underlying their suppression function. Recent studies have focused on understanding the reciprocal relationship between TReg and other immune components surrounding TReg cells. Through these studies it

Conflict of interest statement

The authors declare that there are no conflicts of interest.

References (61)

  • S. Huber et al.

    Th17 cells express interleukin-10 receptor and are controlled by Foxp3(−) and Foxp3 + regulatory CD4 + T cells in an interleukin-10-dependent manner

    Immunity

    (2011)
  • D.R. Littman et al.

    Th17 and regulatory T cells in mediating and restraining inflammation

    Cell

    (2010)
  • J. Bienenstock et al.

    A common mucosal immunologic system involving the bronchus, breast and bowel

    Adv. Exp. Med. Biol.

    (1978)
  • R. Jorde et al.

    Diurnal profiles of gastrointestinal regulatory peptides

    Scand. J. Gastroenterol.

    (1985)
  • C.J. Smart et al.

    T lymphocytes of the human colonic mucosa: functional and phenotypic analysis

    Clin. Exp. Immunol.

    (1988)
  • R. Duchmann et al.

    Tolerance exists towards resident intestinal flora but is broken in active inflammatory bowel disease (IBD)

    Clin. Exp. Immunol.

    (1995)
  • P.D. Bos et al.

    Treg cells in cancer: a case of multiple personality disorder

    Sci. Transl. Med.

    (2012)
  • E. Gounaris et al.

    T-regulatory cells shift from a protective anti-inflammatory to a cancer-promoting proinflammatory phenotype in polyposis

    Cancer Res.

    (2009)
  • C. Badoual et al.

    Prognostic value of tumor-infiltrating CD4 + T-cell subpopulations in head and neck cancers

    Clin. Cancer Res.

    (2006)
  • M. Haas et al.

    Stromal regulatory T-cells are associated with a favourable prognosis in gastric cancer of the cardia

    BMC Gastroenterol.

    (2009)
  • S. Ladoire et al.

    Presence of Foxp3 expression in tumor cells predicts better survival in HER2-overexpressing breast cancer patients treated with neoadjuvant chemotherapy

    Breast Cancer Res. Treat.

    (2011)
  • J.D. Fontenot et al.

    Foxp3 programs the development and function of CD4 + CD25 + regulatory T cells

    Nat. Immunol.

    (2003)
  • S. Hori et al.

    Control of regulatory T cell development by the transcription factor Foxp3

    Science

    (2003)
  • R. Khattri et al.

    An essential role for Scurfin in CD4 + CD25 + T regulatory cells

    Nat. Immunol.

    (2003)
  • G. Roncador et al.

    Analysis of FOXP3 protein expression in human CD4 + CD25 + regulatory T cells at the single-cell level

    Eur. J. Immunol.

    (2005)
  • S. Sakaguchi et al.

    FOXP3 + regulatory T cells in the human immune system

    Nat. Rev. Immunol.

    (2010)
  • D. Valmori et al.

    A peripheral circulating compartment of natural naive CD4 Tregs

    J. Clin. Invest.

    (2005)
  • J.L. Coombes et al.

    A functionally specialized population of mucosal CD103 + DCs induces Foxp3 + regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism

    J. Exp. Med.

    (2007)
  • L. Zhou et al.

    TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function

    Nature

    (2008)
  • K.S. Voo et al.

    Identification of IL-17-producing FOXP3 + regulatory T cells in humans

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • Cited by (70)

    • Dynamic stromal cellular reaction throughout human colorectal adenoma-carcinoma sequence: A role of TH17/IL-17A

      2021, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      In the adenoma stage, TH17 cells could be the main cellular source for IL-17A, however in the CRC stage other cells might join the production of IL-17A. For instance, studies have shown that IL-17 could be produced from stromal cells such as fibroblast [56] and FoxP3-positive regulatory T cells [57], these cells are significantly increased and activated in the CRC microenvironment [4,13,48,58,59]. Stromal response in reaction to tumorigenesis includes a series cellular and functional changes i.e. stromal cell proliferation, population expansion and phenotypical differentiation, growth factor production, enhanced angiogenesis and extracellular matrix modification [60].

    • Myeloid-Derived IL-33 Limits the Severity of Dextran Sulfate Sodium–Induced Colitis

      2021, American Journal of Pathology
      Citation Excerpt :

      Because Tregs suppress excessive cytokine responses in both mouse and human colitis13,14 and IL-33 both expands Treg numbers and promotes their suppressive functions,15 the impact of loss of myeloid IL-33 on intestinal Treg subsets was studied. Among Treg subsets that suppress inflammation, Foxp3+RORγt+ Tregs selectively suppress type 17 helper T-cell (Th17) responses, whereas Gata3+Foxp3+ Tregs may preferentially suppress type 2 helper T-cell responses.16,17 Using a flow cytometry approach, the expression of Foxp3 and RORγt by T cells was evaluated in colon lamina propria (Figure 3A).

    View all citing articles on Scopus

    This study is supported by NIH grants HL107997-01, R56AI43552 and by the Leukemia and Lymphoma Society Translational Research Program TRP 6222–11.

    View full text