Skip to main content
Clinical and Experimental Immunology logoLink to Clinical and Experimental Immunology
. 2010 Feb;159(2):120–130. doi: 10.1111/j.1365-2249.2009.04038.x

Translational Mini-Review Series on Th17 Cells: Induction of interleukin-17 production by regulatory T cells

B Afzali *, P Mitchell *, R I Lechler *, S John , G Lombardi *
PMCID: PMC2810380  PMID: 19912251

Abstract

Uncommitted (naive) CD4+ T helper cells (Thp) can be induced to differentiate to specific lineages according to the local cytokine milieu, towards T helper type 1 (Th1), Th2, Th17 and regulatory T cell (Treg) phenotypes in a mutually exclusive manner. Each phenotype is characterized by unique signalling pathways and expression of specific transcription factors, notably T-bet for Th1, GATA-3 for Th2, forkhead box P3 (FoxP3) for Tregs and receptor-related orphan receptor (ROR)α and RORγt for Th17 cells. Tregs and Th17 cells have been demonstrated to arise from common precursors in a reciprocal manner based on exposure to transforming growth factor (TGF)-β or TGF-β plus interleukin (IL)-6 and carry out diametrically opposing functions, namely suppression or propagation of inflammation, respectively. However, while epigenetic modifications in Th1 and Th2 differentiated cells prevents their conversion to other phenotypes, Th17 cells generated in vitro using TGF-β and IL-6 are unstable and can convert to other phenotypes, especially Th1, both in vitro and in vivo. Tregs are generated from naive precursors both in the thymus (natural, nTregs) and in the periphery (induced, iTregs). The highly suppressive function of Tregs enables them to control many inflammatory diseases in animals and makes them particularly attractive candidates for immunotherapy in humans. The stability of the Treg phenotype is therefore of paramount importance in this context. Recent descriptions of Treg biology have suggested that components of pathogens or inflammatory mediators may subvert the suppressive function of Tregs in order to allow propagation of adequate immune responses. Unexpectedly, however, a number of groups have now described conversion of Tregs to the Th17 phenotype induced by appropriate inflammatory stimuli. These observations are particularly relevant in the context of cell therapy but may also explain some of the dysregulation seen in autoimmune diseases. In this paper, we review Treg to Th17 conversion and propose some potential mechanisms for this phenomenon.

Keywords: autoimmune disease, human, regulatory T cells, Th17, transcription factors

Introduction

Random rearrangement of T cell receptor (TCR) genes in the thymus during ontogeny unsurprisingly generates some T cells with cognate specificity for self-antigens, imparting an inherent potential in the immune system for self-reactivity and autoimmune disease. While this capacity is reduced by the negative selection of autoreactive thymocytes by the AIRE (autoimmune regulator protein)-directed [1] ectopic expression of tissue specific antigens (TSAs) on medullary thymic epithelial cells (mTECs) and dendritic cells (DCs) [2,3] (‘central tolerance’), this is an incomplete process, with thymic émigrés containing a proportion of autoreactive cells. As a result, the mature T cell repertoire retains the capacity for autoimmunity. In order to prevent the development of autoimmune diseases, peripheral mechanisms of retaining tolerance to self-antigens (‘peripheral tolerance’) exclude recirculating T cells from the sites of their cognate antigens, fail to provide adequate co-stimulation to T cells and actively ‘suppress’ the activation of autoreactive cells.

Although a number of immunoregulatory cells have been described in the literature, [415], it is thought that CD4+ T cells expressing high levels of the interleukin (IL)-2 receptor α chain, CD25 are the most important in the maintenance of peripheral tolerance. These CD4+CD25hi regulatory T cells (Tregs) are derived developmentally from the neonatal thymus [16], but can also be generated directly from naive precursors in the periphery through appropriate activation and cytokine receptor engagement (see below). The former, referred to as natural (n)Tregs, develop in response to self-antigens expressed in the thymus and maintain peripheral self-tolerance while the latter, referred to as induced (i)Tregs, are thought to develop in response to environmental antigens and maintain tolerance to non-self components such as gut flora and ingested material. These two populations have few characteristics that can distinguish them in the peripheral blood (differences between nTregs and iTregs are summarized in the review by Horwitz et al.[17]), therefore for the purposes of the present paper they will be considered together. The critical, non-redundant, importance of Tregs in mammalian biology is highlighted by the development of life-threatening autoimmune diseases in both humans and mice who are deficient in this population (as a result of mutations in the FOXP3 and foxp3 genes, respectively; see below) [15,1820].

Treg function

While the precise means of Treg function are not entirely understood it is likely that they possess a functional repertoire of suppressive mechanisms, which would be consistent with diverse descriptions of suppression through direct cell-to-cell contact, production of soluble mediators [2123] and activity through intermediary cells [24,25]. As a result, Tregs have the in vitro ability to inhibit proliferation and production of cytokines [notably IL-2 and interferon (IFN)-γ] by non-regulatory, traditional T cells (CD4+CD25-) [2629] as well as responses of CD8+ T cells, monocytes and natural killer (NK) cells [26,30,31]. These predicates translate in vivo to a greater number of functions other than the maintenance of tolerance to self-components (i.e. prevention of autoimmune disease) [32] and include control of allergic diseases [33], maintenance of gastrointestinal (GI) tolerance [34] and maternal acceptance of semi-allogeneic fetal antigens [35]. A detailed review on Treg functions is provided by O'Connor et al. in this series [36].

Tregs in infection and Treg subversion

Tregs can clearly regulate responses to infectious agents [37,38]; however, appropriate responses to pathogens should balance the need for antigen clearance against excessive tissue injury engendered by an over-exuberant immune system (Fig. 1). Excessive Treg activity is observed in persistent infections such as murine models of Leishmaniasis, malaria and tuberculosis [3941] and in human diseases such as upper GI persistence of Helicobacter pylori, human immunodeficiency virus (HIV) and hepatitis C virus (HCV) infections [4245], suggesting the possibility of a link between pathogen persistence and Treg-mediated suppression. Subversion of Treg function for the generation of appropriate immune responses to effect efficient pathogen clearance may therefore be an advantage or, indeed, a necessity.

Fig. 1.

Fig. 1

Regulatory T cells during infection. In the context of infection, excessive regulatory T cell (Treg) activity may leave the host susceptible to overwhelming infection (left side of the swing), while too little Treg activity may result in excessive tissue injury from unbridled inflammation (right side of the swing).

Indeed, accumulating evidence supports the assertion that interactions between Tregs and an infective/inflammatory environment leads to the subversion of their suppressive function. The salient experiments demonstrate a direct effect of Toll-like receptor (TLR) ligation on Tregs to block their suppression [46,47] and modulation of dendritic cell (DC) activity by lipopolysaccharide (LPS) to induce restricted Treg activity [48] in a manner that is independent of direct ligation of the TLR on Tregs[49,50]. Indeed, appropriately activated DC can break the ‘anergic’ state of Tregs and promote proliferation in this usually hypoproliferative population [51]. Our own (unpublished) observations and those of others suggest that proinflammatory cytokines, in particular IL-1β, IL-6 and tumour necrosis factor (TNF)-α, released by DC following interaction with pathogens, can subvert the suppressive effects of Tregs. Both IL-1β and IL-6 can block Treg-mediated suppression of effector cell proliferation [48,52], although IL-6 may require the presence of IL-1 to overcome regulation [49]. There are some data from humans to suggest that TNF-α can inhibit Treg function [53] with some supporting, but circumstantial, evidence showing a numerical increase in forkhead box P3 (FoxP3)+ T cells and restoration of defective regulatory function in patients with rheumatoid arthritis treated with anti-TNF-α therapy [54]. The inevitable question is whether subverted Tregs remain ‘dormant’ Tregs or undergo a stable change of phenotype to an alternative lineage.

T helper type 17 (Th17) cells, development, function and stability

IL-17 is a proinflammatory cytokine with non-redundant functions in the clearance of extracellular pathogens (see also [55] for further detail). This is seen readily in both IL-17R-deficient mice, which demonstrate great susceptibility to lethal bacterial infections [56,57], and in IL-17-deficient humans as part of the hyper-immunoglobulin E (IgE) syndrome (HIES), where recurrent infections are a feature [58,59]. The significant proinflammatory features of IL-17 have been reviewed previously, as has the compelling evidence for the role of IL-17 in inflammatory/autoimmune conditions of mice and the considerable body of evidence suggesting an important role for IL-17 in the aetiopathogenesis of inflammatory and autoimmune diseases in humans [60,61]. The majority of IL-17 is derived from a population of CD4+ Th cells discrete from Th1 and Th2, known as Th17.

There has been much interest in the differentiation of Th17 cells from naive precursors and it is now understood that Th17 commitment is linked reciprocally to that of Tregs. While transforming growth factor (TGF)-β differentiates murine naive CD4+ T cells to Tregs, the presence of IL-6, in addition to TGF-β, skews the commitment towards Th17 [6264]. There is greater debate regarding human Th17 differentiation. These pathways of differentiation are discussed in more depth in the review by de Jong and Lord in this series [65]. However, it is important to note that the evidence indicates that Th17 cells are unstable or that the phenotype may be an intermediately differentiated state. In particular, bulk CD4+ T cells primed to produce IL-17 by polyclonal activation (anti-CD3 and anti-CD28) in the presence of IL-23 can be redirected away from IL-17 production towards a Th1 phenotype by subsequent TCR activation in the absence of IL-23 or by induction of the Th1 specifying transcription factor, T-bet, suggesting that the Th17 state may be either unstable or a non-terminally differentiated one [66]. This is corroborated by in vivo murine data demonstrating that the adoptive transfer of highly purified islet-specific Th17 cells, devoid of IFN-γ producing populations, causes type 1 diabetes mellitus in recipient mice through the conversion of the Th17 population to a Th1 phenotype (as characterized by cytokine and transcription factor profiles) [67]. This is also observed in experimental autoimmune encephalomyelitis (EAE) models, where fate-mapping of adoptively transferred Th17-skewed cells reveals a significant conversion in vivo to the Th1 lineage [68]. All these findings suggest that there is considerably more plasticity among ‘skewed, lineage-committed’ Th17 cells than thought previously, and contrasts with Th1 and Th2 lineages which are resistant to further differentiation as a result of epigenetic modifications of gene loci associated with the reciprocal lineage [69], ensuring that Th1 and Th2 phenotypes remain stably expressed.

Treg to Th17 conversion

A number of groups, including our own, have investigated the subversion of Tregs by inflammatory cytokines in both mouse and man and found that, in addition to reduced suppressive activity on target cells, inflammatory cytokines direct Tregs to differentiate into the Th17 lineage and produce IL-17. That this conversion is not the result of outgrowth of a contaminating Th17 precommitted population is indicated by the demonstration of double-positive cells for the Treg transcription factor FoxP3 and IL-17 (our unpublished observations), which is suggestive of an intermediate, transitional, stage. The conversion of Tregs to Th17 cells has now been reported by a number of groups, in both mouse and human, as shown in Table 1[7079], albeit with a very interesting difference. While murine Tregs are converted predominantly to Th17 under the influence of IL-6 [70,74,75,78], the same cytokine which skews naive Th differentiation away from Treg and towards Th17 [6264], human Tregs are largely resistant to IL-6 and differentiate to the Th17 lineage in an IL-1-dependent manner [73,76,77]. Of note here, one recent murine study has shown that IL-1 signalling is also essential for Th17 lineage differentiation in mice, and that IL-6 induces IL-1R expression on T cells. In this report, IL-1r1−/− animals had higher percentages of FoxP3+ T cells compared to wild-type counterparts, and in an EAE model wild-type, but not IL-1r1−/−, FoxP3+ T cells produced IL-17 in the central nervous system (CNS), suggesting a greater similarity in Th17 differentiation and Treg to Th17 conversion between humans and mice than thought previously [79]. Murine Tregs can be directed towards the Th17 lineage through receptor–ligand interactions on DC that activate them to produce the appropriate cytokine environment, including (Curdlan-induced) Dectin-1 activation [72] and B7 cross-linking on DC [78]. Conversely, murine Tregs can be protected from IL-6-driven Th17 conversion following exposure to TGF-β and IL-2, as these cytokines in concert reduce surface expression of the IL-6 receptor [75]. As a result, it has been proposed that TGF-β iTregs are more resistant to Th17 conversion in mice than nTregs[75]. This is the only publication that demonstrates a potential difference between nTregs and iTregs in the propensity to convert to the Th17 lineage and should be accepted only with the caveats that the observed effect cannot be said categorically to be due to inherent differences between nTregs and iTregs and not the result of TGF-β and IL-2 signalling per se, and that the concentrations of TGF-β and IL-2 used in iTreg generation in vitro are orders of magnitude higher than those seen in vivo.

Table 1.

Factors inducing regulatory T cells (Treg) to T helper type 17 (Th17) conversion in mouse and human.

Species Reference Condition mediating Treg to Th17 differentiation Additional information
Mouse [71] IL-6 Naturally occurring Tregs were more susceptible to Th17 conversion than in vitro IL-2/TGF-β generated Tregs
[70] IL-6 Enhanced by IL-1 and IL-23
[66] IL-6 LPS activated DC produced sufficient IL-6 to also have this effect
[75] IL-1 Il-1r1−/− animals had higher percentages of FoxP3+ T cells compared to wild-type counterparts. In an EAE model, wild-type, but not Il-1r1−/−, FoxP3+ T cells produced IL-17 in the CNS
[74] B7-DC cross-linking antibody in an IL-6-dependent manner Adoptively transferred Th17-converted Tregs induced antigen-specific diabetes mellitus in recipient animals
[68] Dectin-1 activated DC IL-23 produced by the DC was critical in this model
Human [72] IL-1β plus IL-2 Both naive and memory Tregs converted to Th17, although memory Tregs were more efficiently skewed. IL-6 did not convert Tregs to Th17
[69] Mainly IL-1β plus either IL-2 or IL-15 Histone deacetylase enzyme inhibition protected Tregs from Th17 conversion
[73] Mainly IL-1β plus IL-2 IL-17 concentrations were enhanced by IL-6, IL-21 and IL-23 in addition
[67] IL1β plus IL-6 and IL-2 HLA-DR- Tregs were more potently converted to Th17 than HLA-DR+ Tregs. IL-6 was more potent than IL-1β in DR- Treg skewing to Th17 but the two synergized together

In addition to cell activation through the T cell receptor (TCR).

Interleukin (IL)-2/IL-15 do not convert Tregs to Th17 but enhance the efficiency of conversion by other cytokines. CNS: central nervous system; DC: dendritic cell; EAE: experimental autoimmune encephalomyelitis; HLA-DR: human leucocyte antigen D-related; FoxP3: forkhead box P3; LPS: lipopolysaccharide; TGF: transforming growth factor.

Some of these reports have demonstrated that Th17 cells derived from Tregs share common features with Th17 cells generated from naive precursors, including expression of the chemokine receptor CCR6 [73,76,80]. CCR6 is a chemokine receptor expressed on the surface of Th17 cells, under the control of the Th17 transcription factor receptor-related orphan receptors (ROR)α and RORγt, which directs their migration into sites of inflammation [81]. Interestingly, although ‘converted’ Tregs also express CCR6 (as well as other chemokine receptors in common with Th17 cells [82]), in contrast to Th17 cells they do not express CCL20 [macrophage inflammatory protein (MIP)-3α][81], which is the only known ligand for CCR6 [83]. Th17 cells therefore recruit other Th17 cells and Tregs into sites of inflammation through secretion of CCL20 [81]. Indeed, chronically inflamed tissues in human diseases are characterized by the presence of infiltrating Th17 cells expressing CCR6 [84], and mice are protected from developing EAE if the CCR6–CCL20 interaction is neutralized [81]. Expression of CCR6 by Th17 cells derived from conversion of Tregs[73,76] is therefore a significant finding, and suggests that these Th17 cells have the capacity to migrate to sites of inflammation containing CCL20. However, these observations should be tempered by murine data showing that IL-17 is produced from both CCR6- and CCR6+ Tregs at sites of disease (in this case, the CNS) [81].

In humans, the biological relevance of Treg to Th17 conversion seen in vitro is unknown; however, human memory phenotype (CD45RO+) FoxP3+ Tregs isolated ex vivo have been shown very recently to secrete IL-17 and to express the Th17 transcription factor RORγt constitutively [85], suggesting that IL-17 production from Tregs also occurs in vivo. The reversal of the regulatory function of Tregs, and skewing of phenotype towards production of IL-17, a cytokine known to be important in human autoimmune diseases [60], may provide a link between the loss of regulation and high levels of IL-17 seen in some of these disorders. In addition, mice in which the IL-1 receptor antagonist gene has been silenced develop spontaneous autoimmune T cell-mediated arthritis, an IL-17-mediated condition [86,87], due to excessive IL-1 signalling [88]. These mice do not exhibit arthritis when kept germ-free, but rapidly develop pathological features when exposed to a single species of indigenous gut flora (Lactobacillus bifidus) or to signalling through TLRs [89]. The epidemiological association between infections and the development of human autoimmune diseases could indicate a similar mechanism through altered Treg function and the promotion of IL-17, potentially also mediated through IL-1 or associated TLR signalling pathways.

Demonstrations of the capacity of Tregs to convert to the Th17 lineage also suggests that infiltrating CD4+ cells bearing the phenotype of Tregs (CD4+CD25+FoxP3+) at sites of infection [42] where IL-1β or IL-6 are highly expressed may not necessarily effect a suppressive function, but might instead participate in clearance of the inciting pathogen through conversion to the Th17 lineage. The stability of the Th17 phenotype in this model is an important consideration: given that Th17 cells generated from naive precursors are not stable either in vitro or in vivo[6668], prolonged Treg-derived Th17 persistence at sites of inflammation may engender excessive tissue injury. Although this has not been addressed sufficiently in the literature, some available data suggest that restoration of suppressive function may be possible upon exposure to IL-2 [71].

The Treg–Th17 axis and mechanisms of conversion

In the context of concerted efforts to use expanded populations of Tregs for adoptive therapy in human inflammatory diseases, descriptions of Treg to Th17 conversion are important observations, as transition of adoptively transferred cells from an anti- to a proinflammatory lineage may exacerbate, rather than ameliorate, disease. Therefore, an understanding of the mechanisms underlying this conversion and methods to stabilize the Treg phenotype have become important aspects of Treg biology. Although the mechanisms have not been elucidated fully, there are some suggestions in the available literature, which will be reviewed here. These include upstream signalling and transcription factor interactions.

Transcription factor interactions – FoxP3 and RORγt

Several members of the retinoic acid receptor (RAR) orphan receptor (ROR) family have been described as transcription factors expressed specifically in Th17 cells. These include RORα and RORγt [9092], which are encoded by the genes RORA and RORC. RORγt is induced by TGF-β and IL-6 in naive Thp and leads to transcription of IL-17 [90]. As expected, overexpression of RORγt promotes Th17 differentiation. However, while RORγt-deficient mice have reduced numbers of Th17 cells, the population is not depleted [90]. This is because RORα is also expressed highly in TGF-β/IL-6-induced Th17 cells [91]. This related transcription factor synergizes with RORγt to induce Th17 differentiation, and elimination of both RORα and RORγt (double-deficient animals) at the same time is required to deplete Th17 differentiation effectively and protect against Th17-driven autoimmune diseases [91].

The Scurfy mouse (sf), an X-linked mutant strain, described in 1949 (loc. cit. [93], exhibits a series of autoimmune features including skin scaliness, diarrhoea and death (between 2 and 4 weeks after birth) in association with CD4+ T cell hyperproliferation, multi-organ CD4+ cell infiltration [94] and over-production of several inflammatory cytokines [95]. This fatal autoimmune lymphoproliferative syndrome maps to a gene locus on the X chromosome called foxp3, which has been described as a member of the forkhead/winged-helix family of transcription factors [96]. The foxp3 gene is highly conserved between species and a mutation in the human gene, FOXP3, has been identified as the causative factor responsible for the human equivalent of Scurfy, the immunodysregulation, polyendocrinopathy and enteropathy, X-linked syndrome (IPEX), also known as X-linked autoimmunity and allergic dysregulation syndrome (XLAAD) [19,97,98]. Both the mouse and human disease lack discrete circulating Tregs, which suggests that foxp3 and FOXP3 are essential for normal Treg development in the two species, respectively. This position is strengthened by the failure of foxp3 knock-out mice to develop circulating Tregs; these animals develop a Scurfy-like syndrome from which they can be rescued by the adoptive transfer of Tregs from a foxp3 replete animal [99]. Furthermore, ectopic or over-expression of foxp3 in CD4+CD25- mouse cells results in development of a Treg phenotype [97,99,100]. In mice, FoxP3 expression is a good phenotypic marker of Tregs[101,102]; in humans, however, FoxP3 does not allow the unambiguous identification of Tregs[103], as FoxP3 is induced during TCR stimulation in conventional CD4+ T cells [104106] (in much the same manner as CD25) and there is some debate as to whether the induced CD4+CD25+FoxP3+ population is suppressive or anergic [104,105]. The biology of FoxP3 is still understood incompletely, but there are suggestions that FoxP3 may function as a transcriptional inhibitor through associations with nuclear factor of activated T cells (NFAT) and nuclear factor-kappa B (NF-κB) [107,108].

Recent evidence suggests that similar mechanisms may regulate the commitment of Thp between Treg and Th17. In human cells, FoxP3 exists in two separate but equally expressed isoforms: one (FoxP3), which is encoded by a full length mRNA and the other a truncated form lacking exon 2 (FoxP3Δ2), which is coded by a splice variant mRNA [104,109]. Tregs, perhaps unexpectedly, also express Th17-specifying transcription factors, notably RORα[110] and RORγt [111]. However, co-immunoprecipitation experiments have shown that FoxP3 binds to RORα and RORγt and inhibits their biological activity in a dose-dependent fashion [110,111]. This interaction is mediated through a (LxxLL) motif in the FoxP3 second exon; as expected, the FoxP3Δ2 isoform is unable to bind RORα or RORγt [110,111].

A similar interaction has subsequently been described, by the same group and others, in murine cells. Specifically, both FoxP3 and RORγt are co-expressed in naive CD4+ T cells exposed to TGF-β, where FoxP3 inhibits RORγt directly through a physical interaction, repressing the Th17 programme [111]. In these experiments exposure of Thp to TGF-β leads to rapid induction of RORγt [92], but the binding of RORγt to the IL-17 promoter is suppressed by interaction with FoxP3 [112]. Upon addition of exogenous IL-6 or IL-21, the inhibitory effect of FoxP3 on IL-17 induction is circumvented [111] and FoxP3 levels are reduced [112]. The interaction between FoxP3 and RORγt in murine cells is also dependent upon the second exon of FoxP3 [111,112]. These observations have also been confirmed by another, independent group [74].

These interactions can, in part, explain the conversion of Tregs to Th17, at least in mice. While TGF-β induces both FoxP3 and RORγt expression, IL-6 does not alter expression of RORγt but inhibits FoxP3. As a result, exposure of Tregs to IL-6 down-modulates FoxP3 preferentially and reduces the physical inhibition of RORγt, permitting binding to the IL-17 gene promoter. In addition, very recent murine data suggest that IL-1 regulates expression of RORγt [79].

Signal transducer and activator of transcription (STAT) 3 and 5

The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway is a receptor-coupled signal transduction mechanism linking cytokine–receptor interactions to gene expression. There are seven STAT (STAT1-4, 5A, 5B and 6) and four JAK [JAK1-3 and TYK2 (tyrosine kinase 2)] proteins in humans (reviewed in [113]). Specific JAKs are associated with the cytoplasmic tails of multimeric cytokine receptors, and are activated upon ligand-induced receptor oligomerization [113,114]. Activated JAKs phosphorylate specific tyrosine residues on cytoplasmic tails of their associated cytokine receptors, creating docking sites for the SH2 (Src-homology-2) domain of STAT proteins, and then activate the docked STATs through tyrosine phosphorylation. Activated STATs dimerize and translocate to the nucleus to regulate gene transcription by binding to specific promoter regions. Suppressor of cytokine signalling (SOCS) proteins induced by STAT signal transduction feed back to regulate STAT signalling negatively. The JAK/STAT/SOCS pathway is depicted in Fig. 2.

Fig. 2.

Fig. 2

The Janus kinase/signal transducer and activator of transcription/suppressor of cytokine signalling (JAK/STAT/SOCS) axis. Cytokine engagement of their cognate receptors (a) leads to receptor dimerization, which activates JAK kinases. Activated JAKs phosphorylate the cytoplasmic tails of the receptor (b), creating docking sites for Src-homology-2 (SH2)-domain-containing proteins such as STATs (c). Docked STATs are activated by JAKs, dissociate from the receptor and dimerize (d). Dimerized STATs migrate to the nucleus and associate with specific STAT binding sites (e) and (positively or negatively) regulate expression of STAT-responsive genes, including suppressor of cytokine signalling (SOCS) proteins. SOCS proteins also contain an SH2-domain and feed back to inhibit this pathway through competition with STAT proteins for SH2-domain-docking sites (f), SH2-recruitment to the receptor cytoplasmic domain followed by JAK inhibition (g) or direct inhibition of JAK activity (h). In addition, SOCS proteins contain a SOCS box motif which contains an E3 ubiquitin–ligase complex, through which the SOCS protein can ubiquitinate JAK proteins and target them for proteosomal degradation (i).

The IL-2-induced JAK3/STAT5 pathway is an indispensible signal transduction mechanism for the direct induction of FOXP3 in Tregs[115], as animals deficient in IL-2, IL-2Rα (CD25), IL-2Rβ (CD122) or STAT5 have depleted numbers of Tregs, fail to express FoxP3 and develop autoimmune diseases [115118]. In these models, animals can be rescued from autoimmunity with restored Treg development if IL-2 signalling is re-established, for example, by reconstitution with bone marrow from an IL-2Rβ mutant that activates STAT5 exclusively or by ectopic activation of foxp3 in CD4+ T cells [116]. Similarly, STAT5 deficiency in humans results in loss of Tregs and immune dysregulation, while overexpression of STAT5 in CD4+CD25- cells leads to elevated levels of FoxP3 [119,120]. These observations can be explained by the direct binding of STAT5 to the foxp3 promoter [115,116], instigating an IL-2-directed STAT5-dependent positive regulation of foxp3. Indeed, Tregs show an obligate requirement, both in vivo and in vitro, for IL-2 and the structurally related IL-2 family members, IL-15 and IL-7, for maintenance of FoxP3 expression and suppressive function [119,121123].

Th17 development from naive precursors is dependent upon signal transduction through STAT3. In mice, RORC is a STAT3 target gene and Th17 differentiation is induced by STAT3 signalling cytokines, notably IL-6, IL-21 and IL-23, and can be abrogated effectively by a deficiency in STAT3 [124]. In humans, STAT3 deficiency from dominant negative mutations in the STAT3 gene occurs in the hyperIgE (HIES or Job) syndrome (OMIM 147060), which is characterized by morphological abnormalities, recurrent infections (particularly with Staphylococcus aureus and Candida sp.) and a deficiency of Th17 cells [59,125127]. Patients with HIES not only have reduced Th17 numbers, but their naive Th cells are resistant to Th17 differentiation under appropriate stimulatory conditions, with concomitant impairment of RORγt expression relative to healthy controls [59,126].

There are reasons to suspect that the STAT3/STAT5 signalling pathways are important in the conversion of Tregs to Th17. First, there is evidence to suggest that STAT5 and STAT3 cross-regulate the conversion of naive T cells to the Treg and Th17 lineages. Specifically, in mice, IL-2-induced STAT5 inhibits Th17 differentiation from IL-6 and TGF-β stimulated naive Th cells [128]. It should be noted that, although this inhibition is STAT5-dependent, it is unclear whether the mechanism is a direct STAT5 inhibition of IL-17 associated genes or an indirect effect of STAT5-induced FoxP3-directed inhibition of Th17 transcription factors (as described above). Similarly, STAT3-activating cytokines, such as IL-6 and IL-27, regulate FoxP3 negatively in a STAT3-dependent manner [62,115,129]. This enables IL-6-activated STAT3 to inhibit both FoxP3 expression and enable IL-17 production in naive T cells stimulated with TGF-β[74]. Not surprisingly, therefore, humans with HIES (who have mutations in STAT3) have a higher than normal percentage of cells bearing the phenotype of Tregs[59], while mice deficient in the IL-2 signalling cascade (notably IL-2 or STAT5) have a reduction in Tregs and an excess of Th17 cells in association with autoimmune disease.

Given that there appears to be functional antagonism between the STAT3 and STAT5 pathways during the polarization of naive T cells towards Treg or Th17, it can be hypothesized that the plasticity of differentiated Tregs may be regulated by the dominant STAT signal induced by local cytokines.

Interferon regulatory factor-4 (Irf-4)

There are reasons to suspect the involvement of other signalling pathways in the conversion of Tregs to Th17. These include the Irf-4 transcription factor. Irf-4 is a lymphocyte-restricted member of the Irf family of transcription factors [130] that is critical for the function of mature B and T cells [131]. In T cells, Irf-4 binds to the regulatory regions of cytokine genes, notably IL-2, IL-4, IL-10 and IL-13, and enhances their expression [132]. Involvement of Irf-4 in Th17 polarization in mice is suggested by a failure of Th17 skewing in Thp from mice that are Irf-4-deficient [133]. T cells from these mice do not respond to Th17 polarizing conditions (TGF-β plus IL-6) in the same manner as their wild-type counterparts, maintaining low levels of RORγt, and fail to induce experimental allergic encephalomyelitis (EAE) in vivo[133]. Of particular note, while exposure of Thp from Irf-4−/− animals to TGF-β up-regulates FoxP3 in a normal manner, these cells are subsequently resistant to down-regulation of FoxP3 by IL-6, resulting in failure of Th17 differentiation [133]. Irf-4 is therefore a critical factor in the reciprocal differentiation of Tregs and Th17 cells from common precursors. This assertion is reinforced by the promotion, by Irf-4, of IL-21 [134,135], a stabilizing factor for the Th17 phenotype, and the development of IL-17 driven diseases (such as inflammatory arthropathies) in Irf-4-overexpressing animals [134]. As a result, there is the possibility that Irf-4 may also be an important transcription factor for the conversion of Treg-committed cells to a Th17 phenotype under the influence of inflammatory cytokines. This notion is enhanced by the recent finding that IL-1 induces the expression of Irf-4 during early stages of murine Th17 polarization [79].

Conclusions

The potent suppressive nature of Tregs and their ability to ameliorate a wide array of inflammatory conditions in animals has led to considerable efforts directed towards their utilization as therapeutic tools in humans. However, the propensity of Tregs to convert to a Th17 phenotype in the presence of inflammatory mediators is a real concern, as they would have the potential to exacerbate conditions they are intended to treat. To date, only the rudimentary mechanisms of this phenomenon have been identified, but a greater understanding of the mechanisms underlying Treg to Th17 conversion may identify targets for modification and pharmacological intervention that might stabilize Tregs intended for clinical use and inhibit their proinflammatory potential in vivo.

Disclosure

There are no conflicts of interest: the authors have been supported by grants from the Medical Research Council and the British Heart Foundation.

References

  • 1.Peterson P, Org T, Rebane A. Transcriptional regulation by AIRE: molecular mechanisms of central tolerance. Nat Rev Immunol. 2008;8:948–57. doi: 10.1038/nri2450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gallegos AM, Bevan MJ. Central tolerance to tissue-specific antigens mediated by direct and indirect antigen presentation. J Exp Med. 2004;200:1039–49. doi: 10.1084/jem.20041457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Palmer E. Negative selection – clearing out the bad apples from the T-cell repertoire. Nat Rev Immunol. 2003;3:383–91. doi: 10.1038/nri1085. [DOI] [PubMed] [Google Scholar]
  • 4.Grazia Roncarolo M, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K, Levings MK. Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol Rev. 2006;212:28–50. doi: 10.1111/j.0105-2896.2006.00420.x. [DOI] [PubMed] [Google Scholar]
  • 5.Weiner HL. Induction and mechanism of action of transforming growth factor-beta-secreting Th3 regulatory cells. Immunol Rev. 2001;182:207–14. doi: 10.1034/j.1600-065x.2001.1820117.x. [DOI] [PubMed] [Google Scholar]
  • 6.Lu L, Werneck MBF, Cantor H. The immunoregulatory effects of Qa-1. Immunol Rev. 2006;212:51–9. doi: 10.1111/j.0105-2896.2006.00418.x. [DOI] [PubMed] [Google Scholar]
  • 7.Cortesini R, LeMaoult J, Ciubotariu R, Cortesini NSF. CD8+CD28− T suppressor cells and the induction of antigen-specific, antigen-presenting cell-mediated suppression of Th reactivity. Immunol Rev. 2001;182:201–6. doi: 10.1034/j.1600-065x.2001.1820116.x. [DOI] [PubMed] [Google Scholar]
  • 8.Rifa'i M, Kawamoto Y, Nakashima I, Suzuki H. Essential roles of CD8+CD122+ regulatory T cells in the maintenance of T cell homeostasis. J Exp Med. 2004;200:1123–34. doi: 10.1084/jem.20040395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Liu J, Liu Z, Witkowski P, et al. Rat CD8+ FOXP3+ T suppressor cells mediate tolerance to allogeneic heart transplants, inducing PIR-B in APC and rendering the graft invulnerable to rejection. Transpl Immunol. 2004;13:239–47. doi: 10.1016/j.trim.2004.10.006. [DOI] [PubMed] [Google Scholar]
  • 10.Suciu-Foca N, Manavalan JS, Scotto L, et al. Molecular characterization of allospecific T suppressor and tolerogenic dendritic cells: review. Int Immunopharmacol. 2005;5:7–11. doi: 10.1016/j.intimp.2004.09.003. [DOI] [PubMed] [Google Scholar]
  • 11.Liu Z, Tugulea S, Cortesini R, Suciu-Foca N. Specific suppression of T helper alloreactivity by allo-MHC class I-restricted CD8+ Int Immunol. 1998;10:775–83. doi: 10.1093/intimm/10.6.775. [DOI] [PubMed] [Google Scholar]
  • 12.Kapp JA, Honjo K, Kapp LM, Xu XY, Cozier A, Bucy RP. TCR transgenic CD8+ T cells activated in the presence of TGF{beta} express FoxP3 and mediate linked suppression of primary immune responses and cardiac allograft rejection. Int Immunol. 2006;18:1549–62. doi: 10.1093/intimm/dxl088. [DOI] [PubMed] [Google Scholar]
  • 13.Zhang ZX, Young K, Zhang L. CD3+CD4-CD8− alphabeta-TCR+ T cell as immune regulatory cell. J Mol Med. 2001;79:419–27. doi: 10.1007/s001090100238. [DOI] [PubMed] [Google Scholar]
  • 14.Zhang ZX, Yang L, Young KJ, DuTemple B, Zhang L. Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression. Nat Med. 2000;6:782–9. doi: 10.1038/77513. [DOI] [PubMed] [Google Scholar]
  • 15.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155:1151–64. [PubMed] [Google Scholar]
  • 16.Shevach EM. Regulatory T cells in autoimmmunity. Annu Rev Immunol. 2000;18:423–49. doi: 10.1146/annurev.immunol.18.1.423. [DOI] [PubMed] [Google Scholar]
  • 17.Horwitz DA, Zheng SG, Gray JD. Natural and TGF-beta-induced Foxp3(+)CD4(+) CD25(+) regulatory T cells are not mirror images of each other. Trends Immunol. 2008;29:429–35. doi: 10.1016/j.it.2008.06.005. [DOI] [PubMed] [Google Scholar]
  • 18.Sakaguchi S, Toda M, Asano M, Itoh M, Morse SS, Sakaguchi N. T cell-mediated maintenance of natural self-tolerance: its breakdown as a possible cause of various autoimmune diseases. J Autoimmun. 1996;9:211–20. doi: 10.1006/jaut.1996.0026. [DOI] [PubMed] [Google Scholar]
  • 19.Chatila TA, Blaeser F, Ho N, et al. JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. J Clin Invest. 2000;106:R75–81. doi: 10.1172/JCI11679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wildin RS, Ramsdell F, Peake J, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20. doi: 10.1038/83707. [DOI] [PubMed] [Google Scholar]
  • 21.Dieckmann D, Bruett CH, Ploettner H, Lutz MB, Schuler G. Human CD4(+)CD25(+) regulatory, contact-dependent T cells induce interleukin 10-producing, contact-independent type 1-like regulatory T cells [corrected] J Exp Med. 2002;196:247–53. doi: 10.1084/jem.20020642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Longhi MS, Hussain MJ, Mitry RR, et al. Functional study of CD4+CD25+ regulatory T cells in health and autoimmune hepatitis. J Immunol. 2006;176:4484–91. doi: 10.4049/jimmunol.176.7.4484. [DOI] [PubMed] [Google Scholar]
  • 23.Collison LW, Workman CJ, Kuo TT, et al. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature. 2007;450:566–9. doi: 10.1038/nature06306. [DOI] [PubMed] [Google Scholar]
  • 24.Ly D, Mi QS, Hussain S, Delovitch TL. Protection from type 1 diabetes by invariant NK T cells requires the activity of CD4+CD25+ regulatory T cells. J Immunol. 2006;177:3695–704. doi: 10.4049/jimmunol.177.6.3695. [DOI] [PubMed] [Google Scholar]
  • 25.Lu LF, Lind EF, Gondek DC, et al. Mast cells are essential intermediaries in regulatory T-cell tolerance. Nature. 2006;442:997–1002. doi: 10.1038/nature05010. [DOI] [PubMed] [Google Scholar]
  • 26.Dieckmann D, Plottner H, Berchtold S, Berger T, Schuler G. Ex vivo isolation and characterization of CD4(+)CD25(+) T cells with regulatory properties from human blood. J Exp Med. 2001;193:1303–10. doi: 10.1084/jem.193.11.1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Thornton AM, Shevach EM. CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med. 1998;188:287–96. doi: 10.1084/jem.188.2.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Jonuleit H, Schmitt E, Stassen M, Tuettenberg A, Knop J, Enk AH. Identification and functional characterization of human CD4(+)CD25(+) T cells with regulatory properties isolated from peripheral blood. J Exp Med. 2001;193:1285–94. doi: 10.1084/jem.193.11.1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA. CD4+CD25high regulatory cells in human peripheral blood. J Immunol. 2001;167:1245–53. doi: 10.4049/jimmunol.167.3.1245. [DOI] [PubMed] [Google Scholar]
  • 30.Wing K, Lindgren S, Kollberg G, et al. CD4 T cell activation by myelin oligodendrocyte glycoprotein is suppressed by adult but not cord blood CD25+ T cells. Eur J Immunol. 2003;33:579–87. doi: 10.1002/eji.200323701. [DOI] [PubMed] [Google Scholar]
  • 31.Taams LS, van Amelsfort JM, Tiemessen MM, et al. Modulation of monocyte/macrophage function by human CD4+CD25+ regulatory T cells. Hum Immunol. 2005;66:222–30. doi: 10.1016/j.humimm.2004.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Baecher-Allan C, Hafler DA. Human regulatory T cells and their role in autoimmune disease. Immunol Rev. 2006;212:203–16. doi: 10.1111/j.0105-2896.2006.00417.x. [DOI] [PubMed] [Google Scholar]
  • 33.Umetsu DT, DeKruyff RH. The regulation of allergy and asthma. Immunol Rev. 2006;212:238–55. doi: 10.1111/j.0105-2896.2006.00413.x. [DOI] [PubMed] [Google Scholar]
  • 34.Izcue A, Coombes JL, Powrie F. Regulatory T cells suppress systemic and mucosal immune activation to control intestinal inflammation. Immunol Rev. 2006;212:256–71. doi: 10.1111/j.0105-2896.2006.00423.x. [DOI] [PubMed] [Google Scholar]
  • 35.Aluvihare VR, Kallikourdis M, Betz AG. Regulatory T cells mediate maternal tolerance to the fetus. Nat Immunol. 2004;5:266–71. doi: 10.1038/ni1037. [DOI] [PubMed] [Google Scholar]
  • 36.O'Connor RA, Taams LS, Anderton SM. CD4+ T helper cells: functional plasticity and differential sensitivity to regulatory T cell-mediated regulation. Clin Exp Immunol. 2009 doi: 10.1111/j.1365-2249.2009.04040.x. doi: 10.1111/j.1365-2249.2009.04040.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rouse BT, Sarangi PP, Suvas S. Regulatory T cells in virus infections. Immunol Rev. 2006;212:272–86. doi: 10.1111/j.0105-2896.2006.00412.x. [DOI] [PubMed] [Google Scholar]
  • 38.Belkaid Y, Blank RB, Suffia I. Natural regulatory T cells and parasites: a common quest for host homeostasis. Immunol Rev. 2006;212:287–300. doi: 10.1111/j.0105-2896.2006.00409.x. [DOI] [PubMed] [Google Scholar]
  • 39.Belkaid Y, Piccirillo CA, Mendez S, Shevach EM, Sacks DL. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature. 2002;420:502–7. doi: 10.1038/nature01152. [DOI] [PubMed] [Google Scholar]
  • 40.Hisaeda H, Maekawa Y, Iwakawa D, et al. Escape of malaria parasites from host immunity requires CD4+ CD25+ regulatory T cells. Nat Med. 2004;10:29–30. doi: 10.1038/nm975. [DOI] [PubMed] [Google Scholar]
  • 41.Scott-Browne JP, Shafiani S, Tucker-Heard G, et al. Expansion and function of Foxp3-expressing T regulatory cells during tuberculosis. J Exp Med. 2007;204:2159–69. doi: 10.1084/jem.20062105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Lundgren A, Stromberg E, Sjoling A, et al. Mucosal FOXP3-expressing CD4+ CD25high regulatory T cells in Helicobacter pylori-infected patients. Infect Immun. 2005;73:523–31. doi: 10.1128/IAI.73.1.523-531.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Andersson J, Boasso A, Nilsson J, et al. The prevalence of regulatory T cells in lymphoid tissue is correlated with viral load in HIV-infected patients. J Immunol. 2005;174:3143–7. doi: 10.4049/jimmunol.174.6.3143. [DOI] [PubMed] [Google Scholar]
  • 44.Kinter AL, Hennessey M, Bell A, et al. CD25(+)CD4(+) regulatory T cells from the peripheral blood of asymptomatic HIV-infected individuals regulate CD4(+) and CD8(+) HIV-specific T cell immune responses in vitro and are associated with favorable clinical markers of disease status. J Exp Med. 2004;200:331–43. doi: 10.1084/jem.20032069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Pereira LE, Villinger F, Onlamoon N, et al. Simian immunodeficiency virus (SIV) infection influences the level and function of regulatory T cells in SIV-infected rhesus macaques but not SIV-infected sooty mangabeys. J Virol. 2007;81:4445–56. doi: 10.1128/JVI.00026-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Peng G, Guo Z, Kiniwa Y, et al. Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science. 2005;309:1380–4. doi: 10.1126/science.1113401. [DOI] [PubMed] [Google Scholar]
  • 47.Liu H, Komai-Koma M, Xu D, Liew FY. Toll-like receptor 2 signaling modulates the functions of CD4+ CD25+ regulatory T cells. Proc Natl Acad Sci USA. 2006;103:7048–53. doi: 10.1073/pnas.0601554103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Pasare C, Medzhitov R. Toll pathway-dependent blockade of CD4+CD25+ T cell-mediated suppression by dendritic cells. Science. 2003;299:1033–6. doi: 10.1126/science.1078231. [DOI] [PubMed] [Google Scholar]
  • 49.Kubo T, Hatton RD, Oliver J, Liu X, Elson CO, Weaver CT. Regulatory T cell suppression and anergy are differentially regulated by proinflammatory cytokines produced by TLR-activated dendritic cells. J Immunol. 2004;173:7249–58. doi: 10.4049/jimmunol.173.12.7249. [DOI] [PubMed] [Google Scholar]
  • 50.Sutmuller RP, den Brok MH, Kramer M, et al. Toll-like receptor 2 controls expansion and function of regulatory T cells. J Clin Invest. 2006;116:485–94. doi: 10.1172/JCI25439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Yamazaki S, Iyoda T, Tarbell K, et al. Direct expansion of functional CD25+ CD4+ regulatory T cells by antigen-processing dendritic cells. J Exp Med. 2003;198:235–47. doi: 10.1084/jem.20030422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.O'Sullivan BJ, Thomas HE, Pai S, et al. IL-1 beta breaks tolerance through expansion of CD25+ effector T cells. J Immunol. 2006;176:7278–87. doi: 10.4049/jimmunol.176.12.7278. [DOI] [PubMed] [Google Scholar]
  • 53.Valencia X, Stephens G, Goldbach-Mansky R, Wilson M, Shevach EM, Lipsky PE. TNF downmodulates the function of human CD4+CD25hi T-regulatory cells. Blood. 2006;108:253–61. doi: 10.1182/blood-2005-11-4567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Nadkarni S, Mauri C, Ehrenstein MR. Anti-TNF-alpha therapy induces a distinct regulatory T cell population in patients with rheumatoid arthritis via TGF-beta. J Exp Med. 2007;204:33–9. doi: 10.1084/jem.20061531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Koenders MI, van den Berg WB. Are T helper 17 cells really pathogenic in autoimmunity? Clin Exp Immunol. 2009 doi: 10.1111/j.1365-2249.2009.04039.x. doi: 10.1111/j.1365-2249.2009.04039.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ye P, Rodriguez FH, Kanaly S, et al. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. J Exp Med. 2001;194:519–27. doi: 10.1084/jem.194.4.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Nakae S, Komiyama Y, Nambu A, et al. Antigen-specific T cell sensitization is impaired in IL-17-deficient mice, causing suppression of allergic cellular and humoral responses. Immunity. 2002;17:375–87. doi: 10.1016/s1074-7613(02)00391-6. [DOI] [PubMed] [Google Scholar]
  • 58.Grimbacher B, Holland SM, Gallin JI, et al. Hyper-IgE syndrome with recurrent infections – an autosomal dominant multisystem disorder. N Engl J Med. 1999;340:692–702. doi: 10.1056/NEJM199903043400904. [DOI] [PubMed] [Google Scholar]
  • 59.Ma CS, Chew GY, Simpson N, et al. Deficiency of Th17 cells in hyper IgE syndrome due to mutations in STAT3. J Exp Med. 2008;205:1551–7. doi: 10.1084/jem.20080218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Afzali B, Lombardi G, Lechler RI, Lord GM. The role of T helper 17 (Th17) and regulatory T cells (Treg) in human organ transplantation and autoimmune disease. Clin Exp Immunol. 2007;148:32–46. doi: 10.1111/j.1365-2249.2007.03356.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Crome SQ, Wang AY, Levings MK. Function and regulation of human T helper 17 cells in health and disease. Clin Exp Immunol. 2009 doi: 10.1111/j.1365-2249.2009.04037.x. doi: 10.1111/j.1365-2249.2009.04037.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–8. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
  • 63.Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24:179–89. doi: 10.1016/j.immuni.2006.01.001. [DOI] [PubMed] [Google Scholar]
  • 64.Mangan PR, Harrington LE, O'Quinn DB, et al. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. 2006;441:231–4. doi: 10.1038/nature04754. [DOI] [PubMed] [Google Scholar]
  • 65.de Jong E, Suddason T, Lord GM. Development of mouse and human T helper 17 cells. Clin Exp Immunol. 2009 doi: 10.1111/j.1365-2249.2009.04041.x. doi: 10.1111/j.1365-2249.2009.04041.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mathur AN, Chang HC, Zisoulis DG, et al. T-bet is a critical determinant in the instability of the IL-17-secreting T-helper phenotype. Blood. 2006;108:1595–601. doi: 10.1182/blood-2006-04-015016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Bending D, De La PH, Veldhoen M, et al. Highly purified Th17 cells from BDC2.5NOD mice convert into Th1-like cells in NOD/SCID recipient mice. J Clin Invest. 2009 doi: 10.1172/JCI37865. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.O'Connor RA, Prendergast CT, Sabatos CA, et al. Cutting edge: Th1 cells facilitate the entry of Th17 cells to the central nervous system during experimental autoimmune encephalomyelitis. J Immunol. 2008;181:3750–4. doi: 10.4049/jimmunol.181.6.3750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wilson CB, Rowell E, Sekimata M. Epigenetic control of T-helper-cell differentiation. Nat Rev Immunol. 2009;9:91–105. doi: 10.1038/nri2487. [DOI] [PubMed] [Google Scholar]
  • 70.Xu L, Kitani A, Fuss I, Strober W. 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;178:6725–9. doi: 10.4049/jimmunol.178.11.6725. [DOI] [PubMed] [Google Scholar]
  • 71.Beriou G, Costantino CM, Ashley CW, et al. IL-17 producing human peripheral regulatory T cells retain suppressive function. Blood. 2009;113:4240–9. doi: 10.1182/blood-2008-10-183251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Osorio F, LeibundGut-Landmann S, Lochner M, et al. DC activated via dectin-1 convert Treg into IL-17 producers. Eur J Immunol. 2008;38:3274–81. doi: 10.1002/eji.200838950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Koenen HJ, Smeets RL, Vink PM, van RE, Boots AM, Joosten I. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17-producing cells. Blood. 2008;112:2340–52. doi: 10.1182/blood-2008-01-133967. [DOI] [PubMed] [Google Scholar]
  • 74.Yang XO, Nurieva R, Martinez GJ, et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity. 2008;29:44–56. doi: 10.1016/j.immuni.2008.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zheng SG, Wang J, Horwitz DA. Cutting edge: Foxp3+CD4+CD25+ regulatory T cells induced by IL-2 and TGF-beta are resistant to Th17 conversion by IL-6. J Immunol. 2008;180:7112–6. doi: 10.4049/jimmunol.180.11.7112. [DOI] [PubMed] [Google Scholar]
  • 76.Deknuydt F, Bioley G, Valmori D, Ayyoub M. IL-1beta and IL-2 convert human Treg into T(H)17 cells. Clin Immunol. 2009;131:298–367. doi: 10.1016/j.clim.2008.12.008. [DOI] [PubMed] [Google Scholar]
  • 77.Voo KS, Wang YH, Santori FR, et al. Identification of IL-17-producing FOXP3+ regulatory T cells in humans. Proc Natl Acad Sci USA. 2009;106:4793–8. doi: 10.1073/pnas.0900408106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Radhakrishnan S, Cabrera R, Schenk EL, et al. Reprogrammed FoxP3+ T regulatory cells become IL-17+ antigen-specific autoimmune effectors in vitro and in vivo. J Immunol. 2008;181:3137–47. doi: 10.4049/jimmunol.181.5.3137. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 79.Chung Y, Chang SH, Martinez GJ, et al. Critical regulation of early Th17 cell differentiation by interleukin-1 signaling. Immunity. 2009;30:576–87. doi: 10.1016/j.immuni.2009.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Singh SP, Zhang HH, Foley JF, Hedrick MN, Farber JM. Human T cells that are able to produce IL-17 express the chemokine receptor CCR6. J Immunol. 2008;180:214–21. doi: 10.4049/jimmunol.180.1.214. [DOI] [PubMed] [Google Scholar]
  • 81.Yamazaki T, Yang XO, Chung Y, et al. CCR6 regulates the migration of inflammatory and regulatory T cells. J Immunol. 2008;181:8391–401. doi: 10.4049/jimmunol.181.12.8391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lim HW, Lee J, Hillsamer P, Kim CH. Human Th17 cells share major trafficking receptors with both polarized effector T cells and FOXP3+ regulatory T cells. J Immunol. 2008;180:122–9. doi: 10.4049/jimmunol.180.1.122. [DOI] [PubMed] [Google Scholar]
  • 83.Liao F, Rabin RL, Smith CS, Sharma G, Nutman TB, Farber JM. CC-chemokine receptor 6 is expressed on diverse memory subsets of T cells and determines responsiveness to macrophage inflammatory protein 3 alpha. J Immunol. 1999;162:186–94. [PubMed] [Google Scholar]
  • 84.Pene J, Chevalier S, Preisser L, et al. Chronically inflamed human tissues are infiltrated by highly differentiated Th17 lymphocytes. J Immunol. 2008;180:7423–30. doi: 10.4049/jimmunol.180.11.7423. [DOI] [PubMed] [Google Scholar]
  • 85.Ayyoub M, Deknuydt F, Raimbaud I, et al. Human memory FOXP3+ Tregs secrete IL-17 ex vivo and constitutively express the TH17 lineage-specific transcription factor ROR{gamma}t. Proc Natl Acad Sci USA. 2009;106:8635–40. doi: 10.1073/pnas.0900621106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Nakae S, Saijo S, Horai R, Sudo K, Mori S, Iwakura Y. IL-17 production from activated T cells is required for the spontaneous development of destructive arthritis in mice deficient in IL-1 receptor antagonist. Proc Natl Acad Sci USA. 2003;100:5986–90. doi: 10.1073/pnas.1035999100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Cho ML, Kang JW, Moon YM, et al. STAT3 and NF-kappaB signal pathway is required for IL-23-mediated IL-17 production in spontaneous arthritis animal model IL-1 receptor antagonist-deficient mice. J Immunol. 2006;176:5652–61. doi: 10.4049/jimmunol.176.9.5652. [DOI] [PubMed] [Google Scholar]
  • 88.Horai R, Saijo S, Tanioka H, et al. Development of chronic inflammatory arthropathy resembling rheumatoid arthritis in interleukin 1 receptor antagonist-deficient mice. J Exp Med. 2000;191:313–20. doi: 10.1084/jem.191.2.313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Abdollahi-Roodsaz S, Joosten LA, Koenders MI, et al. Stimulation of TLR2 and TLR4 differentially skews the balance of T cells in a mouse model of arthritis. J Clin Invest. 2008;118:205–16. doi: 10.1172/JCI32639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Ivanov II, McKenzie BS, Zhou L, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126:1121–33. doi: 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
  • 91.Yang XO, Pappu BP, Nurieva R, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity. 2008;28:29–39. doi: 10.1016/j.immuni.2007.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Manel N, Unutmaz D, Littman DR. The differentiation of human T(H)-17 cells requires transforming growth factor-beta and induction of the nuclear receptor RORgammat. Nat Immunol. 2008;9:641–9. doi: 10.1038/ni.1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Russell WL, Russell LB, Gower JS. Exceptional inheritance of a sex-linked gene in the mouse explained on the basis that the x/o sex-chromosome constitution is female. Proc Natl Acad Sci USA. 1959;45:554–60. doi: 10.1073/pnas.45.4.554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Blair PJ, Bultman SJ, Haas JC, Rouse BT, Wilkinson JE, Godfrey VL. CD4+CD8− T cells are the effector cells in disease pathogenesis in the scurfy (sf) mouse. J Immunol. 1994;153:3764–74. [PubMed] [Google Scholar]
  • 95.Kanangat S, Blair P, Reddy R, et al. Disease in the scurfy (sf) mouse is associated with overexpression of cytokine genes. Eur J Immunol. 1996;26:161–5. doi: 10.1002/eji.1830260125. [DOI] [PubMed] [Google Scholar]
  • 96.Brunkow ME, Jeffery EW, Hjerrild KA, et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. 2001;27:68–73. doi: 10.1038/83784. [DOI] [PubMed] [Google Scholar]
  • 97.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–61. doi: 10.1126/science.1079490. [DOI] [PubMed] [Google Scholar]
  • 98.Bennett CL, Christie J, Ramsdell F, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–1. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
  • 99.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4:330–6. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 100.Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol. 2003;4:337–42. doi: 10.1038/ni909. [DOI] [PubMed] [Google Scholar]
  • 101.Fontenot JD, Rasmussen JP, Williams LM, Dooley JL, Farr AG, Rudensky AY. Regulatory T cell lineage specification by the forkhead transcription factor foxp3. Immunity. 2005;22:329–41. doi: 10.1016/j.immuni.2005.01.016. [DOI] [PubMed] [Google Scholar]
  • 102.Wan YY, Flavell RA. Identifying Foxp3-expressing suppressor T cells with a bicistronic reporter. Proc Natl Acad Sci USA. 2005;102:5126–31. doi: 10.1073/pnas.0501701102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ziegler SF. FOXP3: of mice and men. Annu Rev Immunol. 2006;24:209–26. doi: 10.1146/annurev.immunol.24.021605.090547. [DOI] [PubMed] [Google Scholar]
  • 104.Walker MR, Kasprowicz DJ, Gersuk VH, et al. Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+ J Clin Invest. 2003;112:1437–43. doi: 10.1172/JCI19441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Gavin MA, Torgerson TR, Houston E, et al. Single-cell analysis of normal and FOXP3-mutant human T cells: FOXP3 expression without regulatory T cell development. Proc Natl Acad Sci USA. 2006;103:6659–64. doi: 10.1073/pnas.0509484103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wang J, Ioan-Facsinay A, van der Voort EI, Huizinga TW, Toes RE. Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol. 2007;37:129–38. doi: 10.1002/eji.200636435. [DOI] [PubMed] [Google Scholar]
  • 107.Schubert LA, Jeffery E, Zhang Y, Ramsdell F, Ziegler SF. Scurfin (FOXP3) acts as a repressor of transcription and regulates T cell activation. J Biol Chem. 2001;276:37672–9. doi: 10.1074/jbc.M104521200. [DOI] [PubMed] [Google Scholar]
  • 108.Bettelli E, Dastrange M, Oukka M. Foxp3 interacts with nuclear factor of activated T cells and NF-kappa B to repress cytokine gene expression and effector functions of T helper cells. Proc Natl Acad Sci USA. 2005;102:5138–43. doi: 10.1073/pnas.0501675102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Allan SE, Passerini L, Bacchetta R, et al. The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. J Clin Invest. 2005;115:3276–84. doi: 10.1172/JCI24685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Du J, Huang C, Zhou B, Ziegler SF. Isoform-specific inhibition of ROR alpha-mediated transcriptional activation by human FOXP3. J Immunol. 2008;180:4785–92. doi: 10.4049/jimmunol.180.7.4785. [DOI] [PubMed] [Google Scholar]
  • 111.Zhou L, Lopes JE, Chong MM, et al. TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature. 2008;453:236–40. doi: 10.1038/nature06878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ichiyama K, Yoshida H, Wakabayashi Y, et al. Foxp3 inhibits RORgammat-mediated IL-17A mRNA transcription through direct interaction with RORgammat. J Biol Chem. 2008;283:17003–8. doi: 10.1074/jbc.M801286200. [DOI] [PubMed] [Google Scholar]
  • 113.Levy DE, Darnell JE., Jr Stats: transcriptional control and biological impact. Nat Rev Mol Cell Biol. 2002;3:651–62. doi: 10.1038/nrm909. [DOI] [PubMed] [Google Scholar]
  • 114.Boulay JL, O'shea JJ, Paul WE. Molecular phylogeny within type I cytokines and their cognate receptors. Immunity. 2003;19:159–63. doi: 10.1016/s1074-7613(03)00211-5. [DOI] [PubMed] [Google Scholar]
  • 115.Yao Z, Kanno Y, Kerenyi M, et al. Nonredundant roles for Stat5a/b in directly regulating Foxp3. Blood. 2007;109:4368–75. doi: 10.1182/blood-2006-11-055756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Burchill MA, Yang J, Vogtenhuber C, Blazar BR, Farrar MA. IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J Immunol. 2007;178:280–90. doi: 10.4049/jimmunol.178.1.280. [DOI] [PubMed] [Google Scholar]
  • 117.Suzuki H, Kundig TM, Furlonger C, et al. Deregulated T cell activation and autoimmunity in mice lacking interleukin-2 receptor beta. Science. 1995;268:1472–6. doi: 10.1126/science.7770771. [DOI] [PubMed] [Google Scholar]
  • 118.Willerford DM, Chen J, Ferry JA, Davidson L, Ma A, Alt FW. Interleukin-2 receptor alpha chain regulates the size and content of the peripheral lymphoid compartment. Immunity. 1995;3:521–30. doi: 10.1016/1074-7613(95)90180-9. [DOI] [PubMed] [Google Scholar]
  • 119.Passerini L, Allan SE, Battaglia M, et al. STAT5-signaling cytokines regulate the expression of FOXP3 in CD4+CD25+ regulatory T cells and CD4+ Int Immunol. 2008;20:421–31. doi: 10.1093/intimm/dxn002. [DOI] [PubMed] [Google Scholar]
  • 120.Cohen AC, Nadeau KC, Tu W, et al. Cutting edge: decreased accumulation and regulatory function of CD4+ CD25(high) T cells in human STAT5b deficiency. J Immunol. 2006;177:2770–4. doi: 10.4049/jimmunol.177.5.2770. [DOI] [PubMed] [Google Scholar]
  • 121.Wuest TY, Willette-Brown J, Durum SK, Hurwitz AA. The influence of IL-2 family cytokines on activation and function of naturally occurring regulatory T cells. J Leukoc Biol. 2008;84:973–80. doi: 10.1189/jlb.1107778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Yates J, Rovis F, Mitchell P, et al. The maintenance of human CD4+ CD25+ regulatory T cell function: IL-2, IL-4, IL-7 and IL-15 preserve optimal suppressive potency in vitro. Int Immunol. 2007;19:785–99. doi: 10.1093/intimm/dxm047. [DOI] [PubMed] [Google Scholar]
  • 123.Setoguchi R, Hori S, Takahashi T, Sakaguchi S. Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med. 2005;201:723–35. doi: 10.1084/jem.20041982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Yang XO, Panopoulos AD, Nurieva R, et al. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem. 2007;282:9358–63. doi: 10.1074/jbc.C600321200. [DOI] [PubMed] [Google Scholar]
  • 125.de Beaucoudrey L, Puel A, Filipe-Santos O, et al. Mutations in STAT3 and IL12RB1 impair the development of human IL-17-producing T cells. J Exp Med. 2008;205:1543–50. doi: 10.1084/jem.20080321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Milner JD, Brenchley JM, Laurence A, et al. Impaired T(H)17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature. 2008;452:773–6. doi: 10.1038/nature06764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Renner ED, Rylaarsdam S, Nover-Sombke S, et al. Novel signal transducer and activator of transcription 3 (STAT3) mutations, reduced T(H)17 cell numbers, and variably defective STAT3 phosphorylation in hyper-IgE syndrome. J Allergy Clin Immunol. 2008;122:181–7. doi: 10.1016/j.jaci.2008.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Laurence A, Tato CM, Davidson TS, et al. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity. 2007;26:371–81. doi: 10.1016/j.immuni.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 129.Huber M, Steinwald V, Guralnik A, et al. IL-27 inhibits the development of regulatory T cells via STAT3. Int Immunol. 2008;20:223–34. doi: 10.1093/intimm/dxm139. [DOI] [PubMed] [Google Scholar]
  • 130.Matsuyama T, Grossman A, Mittrucker HW, et al. Molecular cloning of LSIRF, a lymphoid-specific member of the interferon regulatory factor family that binds the interferon-stimulated response element (ISRE) Nucleic Acids Res. 1995;23:2127–36. doi: 10.1093/nar/23.12.2127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Mittrucker HW, Matsuyama T, Grossman A, et al. Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Science. 1997;275:540–3. doi: 10.1126/science.275.5299.540. [DOI] [PubMed] [Google Scholar]
  • 132.Hu CM, Jang SY, Fanzo JC, Pernis AB. Modulation of T cell cytokine production by interferon regulatory factor-4. J Biol Chem. 2002;277:49238–46. doi: 10.1074/jbc.M205895200. [DOI] [PubMed] [Google Scholar]
  • 133.Brustle A, Heink S, Huber M, et al. The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4. Nat Immunol. 2007;8:958–66. doi: 10.1038/ni1500. [DOI] [PubMed] [Google Scholar]
  • 134.Chen Q, Yang W, Gupta S, et al. IRF-4-binding protein inhibits interleukin-17 and interleukin-21 production by controlling the activity of IRF-4 transcription factor. Immunity. 2008;29:899–911. doi: 10.1016/j.immuni.2008.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Huber M, Brustle A, Reinhard K, et al. IRF4 is essential for IL-21-mediated induction, amplification, and stabilization of the Th17 phenotype. Proc Natl Acad Sci USA. 2008;105:20846–51. doi: 10.1073/pnas.0809077106. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical and Experimental Immunology are provided here courtesy of British Society for Immunology

RESOURCES