Elsevier

Seminars in Immunology

Volume 25, Issue 4, 15 November 2013, Pages 263-272
Seminars in Immunology

Review
Identity crisis of Th17 cells: Many forms, many functions, many questions

https://doi.org/10.1016/j.smim.2013.10.021Get rights and content

Highlights

  • Th17 cells are potent regulators linked to induction and persistence of inflammation.

  • The human Th17 compartment is heterogeneous with pro- and anti-inflammatory constituents.

  • Th17 cells acquire additional effector functions in inflamed tissue.

  • Development and plasticity of Th17 cells are regulated by cytokines and metabolic checkpoints.

Abstract

Th17 cells are a subset of CD4+ effector T cells characterized by expression of the IL-17-family cytokines, IL-17A and IL-17F. Since their discovery nearly a decade ago, Th17 cells have been implicated in the regulation of dozens of immune-mediated inflammatory diseases and cancer. However, attempts to clarify the development and function of Th17 cells in human health and disease have generated as many questions as answers. On one hand, cytokine expression in Th17 cells appears to be remarkably dynamic and is subject to extensive regulation (both positive and negative) in tissue microenvironments. On the other hand, accumulating evidence suggests that the human Th17 subset is a heterogeneous population composed of several distinct pro- and anti-inflammatory subsets. Clearly, Th17 cells as originally conceived no longer neatly fit the long-standing paradigm of stable and irrepressible effector T cell function. Here we review current concepts surrounding human Th17 cells, with an emphasis on their plasticity, heterogeneity, and their many, tissue-specific functions. In spite of the challenges ahead, a comprehensive understanding of Th17 cells and their relationship to human disease is key to ongoing efforts to develop safer and more selective anti-inflammatory medicines.

Introduction

Inflammation is the fundamental process of immune activation. Despite its “killer” reputation, inflammation is required for immune memory and vaccine responses, protection from infectious microorganisms, and even tissue maintenance and repair in the absence of pathogenic infection [1], [2]. Of course, inflammation is also a common underlying pathophysiology observed in autoimmunity, cardiovascular disease, cancer, metabolic syndromes such as diabetes, and fibrosis [3], [4], [5], [6], [7]. A key determinant of whether or not inflammation turns pathologic is its ability to be resolved; in autoimmunity, for example, pathologic tissue damage is mediated by chronic, unresolved inflammation resulting from loss of immune tolerance to host tissues and T and B cell autoreactivity. On the other hand, it is also clear that not all inflammatory responses are the same. Qualitatively distinct inflammatory responses are coordinated at the level of antigen presenting cells (APCs) and carried out by CD4+ T helper (Th) cells, which differentiate from multipotent naïve precursor cells into a variety of phenotypically and functionally distinct effector subsets [8], [9]. Cytokine production (i.e., effector function) by Th cell subsets, in turn, orchestrates various aspects of innate and adaptive immune function.

Th cell differentiation is an instructive process that starts with the presentation of peptide antigens by APCs to T cells via MHC class II molecules. Cognate antigen recognition by naïve Th cell clones induces an elaborate network of activation signals through the T cell antigen receptor (TCR) complex, which combine with positive or negative co-stimulatory signals provided by major T cell co-stimulatory receptors (CD28, CTLA-4) and their ligands expressed on APCs (B7.1, B7.2) [10]. Whereas these signals initiate T cell activation and can influence effector T cell differentiation – depending on the strength and duration of TCR and co-stimulatory signals [11], [12], [13] – additional environmental cues, in the way of local cytokines, are required to specify T cell fate decisions. Cytokines bind to multimeric receptors expressed on Th cells, and regulate T cell differentiation via activation of JAK/STAT signaling pathways. STAT proteins reside in the cytoplasm of resting cells, and undergo rapid dimerization and nuclear translocation following their recruitment to activated (i.e., ligand-bound) cytokine receptors and phosphorylation by receptor-associated JAKs. Once in the nucleus, STAT proteins induce the expression of lineage-specifying transcription factors and coordinate downstream transcriptional programs that underlie Th cell differentiation. There are 7 mammalian STAT proteins (STAT1-4, STAT5a, STAT5b, STAT6), and all but STAT2 play essential roles in Th cell differentiation. A number of outstanding reviews are available elsewhere that detail JAK/STAT signaling pathways and their many functions during Th cell differentiation [14], [15].

Th cell differentiation was long considered a binary process, leading to either IFNγ-producing Th1 cells or Th2 cells that secrete IL-4, IL-5 and IL-13. Whereas Th1 cells develop in an IL-12/STAT4- and IFNγ/STAT1-dependent manner that requires activation of the “master” Th1 transcription factor T-bet (TBX21), Th2 differentiation requires IL-4/STAT6 and the Th2-specifying transcription factor, GATA-3 [16], [17], [18], [19]. Th1 cells, by virtue of IFNγ production, activate CD8+ cytotoxic T lymphocytes, NK cells, and phagocytes to regulate immunity against intracellular bacterial pathogens and viruses, whereas Th2 cells and their effector cytokines provide help to B cells, regulate class-switch antibody recombination, and recruit eosinophils for immune responses against many extracellular bacteria and large parasitic worms (i.e., helminthes) (reviewed in [20]). For nearly 20 years, the Th1/Th2 paradigm was thought to explain all aspects of immunity and inflammation. However, starting with the discovery of Th17 cells in the mid-2000s, the repertoire of effector T cell lineages has expanded dramatically, and now includes Th17 cells, induced T regulatory (iTreg) cells, T follicular helper (Tfh) cells, as well as Th22 and Th9 cells that express IL-22 and IL-9, respectively [9], [21], [22]. With the exception of Th17 cells, the regulation and in vivo functions of these new T cell subsets are only beginning to be unraveled.

The concept of a distinct, pro-inflammatory, IL-17-producing effector T cell subset began with work from Sedgwick and colleagues [23], who showed that IL-23 and not IL-12 is the critical driver of autoimmunity in mice. IL-23 is an IL-12-related heterodimeric pro-inflammatory cytokine comprised of IL-12 p40 and the unique IL-23 p19 subunit [24], [25]. Whereas mice lacking only IL-23 (p19−/−) were completely protected from experimental autoimmune encephalomyelitis (EAE), animals specifically lacking IL-12 (p35−/−) remained susceptible to disease, despite a defect in Th1 cell development [23]. IL-23 was found to induce IL-17A-expressing effector T cells, and these cells were responsible for IL-23-mediated EAE [26], [27]. However, because IL-23 did not induce IL-17A expression in naïve T cells in vitro, the notion of Th17 cells remained enigmatic. Shortly thereafter, pioneering work from the Littman, Stockinger, and Kuchroo, labs described the in vitro differentiation of IL-17-expressing Th17 cells, using combinations of IL-6, IL-21, and TGFβ (i.e., TGFβ1) [28], [29], [30], [31], [32]. Because TGFβ had previously only been considered an anti-inflammatory cytokine that regulated, among other things, development of FOXP3-expressing iTreg cells [33], Weaver et al., thus coined Th17 cells, “an effector lineage with regulatory ties” [34]. Whereas TGFβ alone induces FOXP3, the combination of TGFβ and IL-6 induces Th17 cell development via STAT3-dependent induction of the Th17-specific orphan nuclear receptor, RORγt (RORC in humans) [31], [35], [36]. In turn, STAT3 and RORγt synergize with other, general transcription factors, including the AP-1 family member BATF, IRF4, NFκB, and NFAT to regulate transcription and chromatin remodeling at the IL17A/IL17F locus [37]. In addition to IL17A/F, Th17 cells express a cast of pro-inflammatory cytokines, such as IL-22, TNFα and GM-CSF and, importantly, they selectively express the IL-23 receptor (IL-23R) [38]. In this way, IL-23 regulates the growth and inflammatory function of Th17 cells following their initial differentiation (discussed below). Indeed, many more molecules and pathways have since been defined that both enhance and inhibit the development of Th17 cells, and those are reviewed elsewhere [38], [39]. Here, we focus on the regulation of Th17 cell effector function and plasticity post-initial differentiation, particularly as it relates to inflammation in human autoimmunity and cancer.

Unlike inbred mice housed in barrier facilities, humans have a large, heterogeneous, and highly variable (person-to-person) endogenous effector/memory T cell compartment. Thus, the immune system as seen in human peripheral blood affords a unique view into the natural diversity of T cell effector lineages and provides important and complimentary information to that derived from artificial in vitro T cell culture systems and experimental animal models of autoimmunity. Among the most important insights coming from the study of endogenous human effector/memory T cells is that effector T cell lineages co-express unique lymphocyte homing receptors together with lineage-defining cytokines and lineage-specifying transcription factors [40], [41], [42], [43], [44]. For example, IFNγ-producing Th1 cells are highly enriched within human memory cells that express the chemokine receptor CXCR3 [40], [43]. Th2 cells not only express IL-4, IL-5, and IL-13; they also express the chemokine receptor CCR4 and the chemotactic receptor of prostaglandin D2, CRTH2 [40], [43], [45]. Th17 cells uniformly express the inflammatory chemokine receptor CCR6 [46], [47], though these cells are further heterogenous and can express additional chemokine receptors in combination (see below). Co-regulation of effector cytokines and lymphoid homing receptors confers tissue-specific T cell effector function. For example, tissues produce a variety of chemokines, both at steady-state and upon infection, stress, or damage. Chemokine receptors thus serve to direct T cell traffic into organs and tissues where their effector cytokines are needed for immune responses against specific microbes or for tissue maintenance and repair (reviewed in [48]). Like most aspects of the immune system, chemokines are also a double-edged sword that can contribute to chronic and autoimmune inflammation. The molecular link between effector cytokines and lymphoid-homing receptors comes by virtue of lineage-specific transcription factors (e.g., T-bet, GATA-3, RORγt), which activate chemokine receptor gene expression in a subset-specific manner; forced expression of T-bet, GATA-3, RORγt in cultured human naïve T cells is sufficient to induce CXCR3, CCR4/CRTH2, and CCR6 expression in Th1, Th2, and Th17 cells, respectively [49], [50], [51].

Section snippets

Phenotypes of human Th17 cells

All human peripheral blood memory (CD45RO+) T cells that express IL-17A following ex vivo stimulation are CCR6+ [46], [47]. However, as noted above, CCR6+ human memory T cells are also highly heterogeneous in their expression of other chemokine receptors and cytokines. Two major subsets of endogenous human Th17 cells have been described; CCR6+CCR4+ cells, which produce IL-17A but not IFNγ; and CCR6+CXCR3+ cells that can express IL-17A, either alone or together with IFNγ [46], [47], [52], [53].

Functions of human Th17 cells

The functions of Th17 cells are as poorly understood as they are important. However, after nearly a decade of research, several general principals have begun to emerge. In this section we discuss the consequences of Th17 cell plasticity in autoimmunity and cancer.

Regulation of human Th17 cells

Much has been learned about how Th17 cells develop from naïve precursor cells. In contrast, the molecules and signaling pathways that regulate Th17 cell effector function and plasticity are only beginning to be understood. As noted throughout, cytokines play a major role in the regulation of Th17 cells, both in quiescent and inflamed tissues. However, Th17 cells also appear to be uniquely sensitive (versus other effector T cell lineages) to fluctuations in metabolic parameters. In some

Concluding remarks

Th17 cells play major roles in the development and persistence of inflammatory pathologies. Yet while substantial gains have been made in our understanding of Th17 cell function, many more questions are now evident. As discussed here, Th17 cells come in many forms, display highly dynamic functions, and appear uniquely adept at taking on additional effector functions as dictated by inflammatory tissue microenvironments. In order to gain a truly holistic appreciation for all the various disease-

Acknowledgments

Due to space constraints, we apologize for being unable to cite other appropriate studies. Funding provided by The Scripps Research Institute.

References (162)

  • I.I. Ivanov et al.

    The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells

    Cell

    (2006)
  • M. Veldhoen et al.

    TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells

    Immunity

    (2006)
  • C.T. Weaver et al.

    Th17: an effector CD4 T cell lineage with regulatory T cell ties

    Immunity

    (2006)
  • L. Durant et al.

    Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis

    Immunity

    (2010)
  • X.O. Yang et al.

    STAT3 regulates cytokine-mediated generation of inflammatory helper T cells

    Journal of Biological Chemistry

    (2007)
  • M. Ciofani et al.

    A validated regulatory network for Th17 cell specification

    Cell

    (2012)
  • P. Muranski et al.

    Essentials of Th17 cell commitment and plasticity

    Blood

    (2013)
  • W.W. Lee et al.

    Regulating human Th17 cells via differential expression of IL-1 receptor

    Blood

    (2010)
  • H.M. McGee et al.

    IL-22 promotes fibroblast-mediated wound repair in the skin

    Journal of Investigative Dermatology

    (2013)
  • R. Mukasa et al.

    Epigenetic instability of cytokine and transcription factor gene loci underlies plasticity of the T helper 17 cell lineage

    Immunity

    (2010)
  • G. Wei et al.

    Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells

    Immunity

    (2009)
  • F. Annunziato et al.

    Defining the human T helper 17 cell phenotype

    Trends in Immunology

    (2012)
  • P. Muranski et al.

    Th17 cells are long lived and retain a stem cell-like molecular signature

    Immunity

    (2011)
  • E.G. Harper et al.

    Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: implications for psoriasis pathogenesis

    Journal of Investigative Dermatology

    (2009)
  • A. Peters et al.

    Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation

    Immunity

    (2011)
  • B. Pulendran et al.

    Immunological mechanisms of vaccination

    Nature Immunology

    (2011)
  • A. Mantovani et al.

    Macrophage plasticity and polarization in tissue repair and remodelling

    Journal of Pathology

    (2013)
  • A.K. Ghosh et al.

    Molecular basis of organ fibrosis: potential therapeutic approaches

    Experimental Biology and Medicine

    (2013)
  • J.J. O'Shea et al.

    JAKs and STATs in immunity, immunodeficiency, and cancer

    New England Journal of Medicine

    (2013)
  • G. Wick et al.

    The immunology of fibrosis

    Annual Review of Immunology

    (2013)
  • C.T. Weaver et al.

    IL-17 family cytokines and the expanding diversity of effector T cell lineages

    Annual Review of Immunology

    (2007)
  • A.H. Sharpe

    Mechanisms of costimulation

    Immunological Reviews

    (2009)
  • A. Langenkamp et al.

    Kinetics of dendritic cell activation: impact on priming of TH1, TH2 and nonpolarized T cells

    Nature Immunology

    (2000)
  • L. Gabrysova et al.

    Antigenic strength controls the generation of antigen-specific IL-10-secreting T regulatory cells

    European Journal of Immunology

    (2010)
  • K. Inagaki-Ohara et al.

    SOCS, inflammation, and cancer

    JAK-STAT

    (2013)
  • M. Afkarian et al.

    T-bet is a STAT1-induced regulator of IL-12R expression in naive CD4+ T cells

    Nature Immunology

    (2002)
  • J. Zhu et al.

    Differentiation of effector CD4 T cell populations (*)

    Annual Review of Immunology

    (2010)
  • M.H. Kaplan

    Th9 cells: differentiation and disease

    Immunological Reviews

    (2013)
  • S.G. Tangye et al.

    The good, the bad and the ugly – TFH cells in human health and disease

    Nature Reviews Immunology

    (2013)
  • D.J. Cua et al.

    Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain

    Nature

    (2003)
  • R.A. Kastelein et al.

    Discovery and biology of IL-23 and IL-27: related but functionally distinct regulators of inflammation

    Annual Review of Immunology

    (2007)
  • C.L. Langrish et al.

    IL-23 drives a pathogenic T cell population that induces autoimmune inflammation

    Journal of Experimental Medicine

    (2005)
  • E. Bettelli et al.

    Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells

    Nature

    (2006)
  • L. Zhou et al.

    IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways

    Nature Immunology

    (2007)
  • T. Korn et al.

    IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells

    Nature

    (2007)
  • M.O. Li et al.

    Transforming growth factor-beta regulation of immune responses

    Annual Review of Immunology

    (2006)
  • L.A. Zuniga et al.

    Th17 cell development: from the cradle to the grave

    Immunological Reviews

    (2013)
  • L. Rivino et al.

    Chemokine receptor expression identifies Pre-T helper (Th)1, Pre-Th2, and nonpolarized cells among human CD4+ central memory T cells

    Journal of Experimental Medicine

    (2004)
  • F. Sallusto et al.

    Heterogeneity of CD4+ memory T cells: functional modules for tailored immunity

    European Journal of Immunology

    (2009)
  • F. Sallusto et al.

    Two subsets of memory T lymphocytes with distinct homing potentials and effector functions

    Nature

    (1999)
  • Cited by (58)

    • IL-17A treatment influences murine susceptibility to experimental Riemerella anatipestifer infection

      2020, Developmental and Comparative Immunology
      Citation Excerpt :

      Together, these results indicate that unlike ducks, infection of mice with R. anatipestifer did not induce proinflammatory cytokines, including IL-17A and IL-23, at early time points, although mice succumbed to intraperitoneal inoculation with this pathogen. The biological activity of IL-17A includes a critical role in protective immunity to various pathogens, including bacteria, whereas unregulated expression of IL-17A is associated with pathogenic immune responses and autoimmune disorders (Min et al., 2013; Sundrud and Trivigno., 2013; Whibley and Gaffen, 2015). IL-23, which is involved in the IL-17 pathway in inflammation, drives and maintains the differentiation of Th17 cell (Sundrud and Trivigno., 2013; Whibley and Gaffen, 2015).

    • The paradox of Th17 cell functions in tumor immunity

      2017, Cellular Immunology
      Citation Excerpt :

      Hepatocellular carcinoma patients who then demonstrated greater death rate and decreased survival indicated microvascular density (MVD) and lymphocyte infiltration with IL-17 [71]. IL-17 is associated with Jak-Stat family signaling, specially STAT3, in numerous diseases [170]. According to reports, STAT3 is connected with IL-17-induced VEGF production in malignant tumors [124,171,172].

    View all citing articles on Scopus
    View full text