Abstract
Acquired and genetic immunodeficiencies have revealed an indispensable role for CD4+ T cells in the induction of protective host immune responses against a myriad of microbial pathogens. Influenced by the cytokines present in the microenvironment, activated CD4+ T cells may differentiate into several highly-specialized helper subsets defined by the production of distinct signature cytokines tailored to combat diverse classes of pathogens. The process of specification and differentiation is controlled by networks of core, master, and accessory transcription factors, which ensure that CD4+ T helper (TH) cell responses mounted against an invading microbe are of the correct specificity and type. However, aberrant activation or inactivation of transcription factors can result in sustained and elevated expression of immune-related genes, leading to chronic activation of CD4+ TH cells and organ-specific autoimmunity. In this review, we provide an overview of the molecular basis of CD4+ TH cell differentiation and examine how combinatorial expression of transcription factors, which promotes genetic plasticity of CD4+ TH cells, can contribute to immunological dysfunction of CD4+ TH responses. We also discuss recent studies which highlight the potential of exploiting the genetic plasticity of CD4+ TH cells in the treatment of autoimmune and other immune-mediated disorders.
Keywords: CD4+ T helper cells, transcription factors, functional plasticity, pathogenic T cells
1 |. INTRODUCTION
After encountering a specific antigen, CD4+ T cells undergo clonal expansion and differentiate into functionally distinct TH effector subsets, which orchestrate immune responses against diverse microbial pathogens (Fig. 1). Interferon-γ (IFN-γ)-producing TH1 cells specialize in activating cell-mediated immune responses against intracellular bacteria. TH2 cells target helminths through the production of interleukin (IL)-4, IL-5, and IL-13. Finally, TH17 cell-derived cytokines IL-17A, IL-17F, IL-21, and IL-22 stimulate broad tissue responses that defend the host from extracellular bacterial and fungal infections (reviewed in Christie and Zhu,1 Zhu and Paul,2 and Zhu3). In addition, T follicular helper cells (TFH), TH9, TH22, and TGM-CSF may constitute unique TH cell subsets or potentially represent differentiation states of the effector cells described above. Overactive CD4+ TH responses carry the risk of initiating deleterious proinflammatory responses that can result in collateral tissue damage. Thus, the maintenance of immune homeostasis and prevention of immunopathology requires tight regulatory mechanisms of Treg cells.
FIGURE 1. Transcriptional regulation of CD4+ T helper (TH) cells.
When a naïve CD4+ T cell gets activated by an antigen presenting cell, it has a choice to develop into several different functional TH cells. The decision-making process is influenced by the cytokines in the microenvironment, which activate signal transducers and activators of transcription (STAT) proteins whose primary function is to establish TH lineage-specific enhancer landscape and induce the expression of master transcription factors. Master transcription factors establish and maintain TH cell lineage identity. CD4+ TH1 differentiation is under the control of the master transcription factor T-bet, which is induced by the IFN-γ-STAT1 and IL-12-STAT4 signaling pathways. The master transcription factor Gata3 is upregulated in response to IL-4-STAT6 and IL-2-STAT5 signaling, and it is responsible for coordinating the TH2 differentiation program. TH9 cells are an exception because their development is not controlled by a single lineage-specifying transcription factor, but rather by a complex network of transcription factors, which ensure Il9 gene expression and suppression of TH2- and Treg-cell-specific genes. Proinflammatory cytokines IL-6, IL-21, and IL-23 preferentially activate STAT3, which in conjunction with TGF-β-induced SMADs, induce the expression of RORγt, the master transcription factor of TH17 cell-specific developmental program. TFH cell differentiation is controlled by the master transcription factor Bcl6, whose expression is induced by IL-6-STAT3 and IL-21-STAT3 signaling. Differentiation of regulatory T (Treg) cells is under the control of Foxp3, whose expression is induced and maintained in response to TGF-β-SMAD and IL-2-STAT5 signaling
In a traditionally accepted view of cell fate determination, CD4+ TH differentiation involves several coordinated steps controlled at the transcriptional level: (i) specification, (ii) commitment, and (iii) terminal differentiation.4 Lineage specification of recently activated CD4+ T cells is initiated by TCR stimulation and cytokine signaling, two signals that lead to the activation of core transcription factors: NFAT-AP-1 or BATF-AP-1-IRF-4 and signal transducers and activators of transcription (STAT) proteins.1 Initiation of TH1 cell differentiation is contingent on IFN-γ-mediated STAT1 and IL-12-mediated STAT4 activation, while IL-2 and IL-4 signaling activate STAT5 and STAT6, respectively, to promote T 2 development.5–11 Proinflammatory cytokines IL-6, IL-21, and IL-23 preferentially activate STAT3 and drive the transcriptional regulation of TH17 lineage specification, whereas Treg lineage commitment is dependent on IL-2-STAT5 signaling.12,13
In addition to promoting targeted chromatin remodeling and establishing the TH cell-specific enhancer landscape, core transcription factors also induce the expression of master transcription factors that control lineage commitment.14 Master transcription factors are necessary and sufficient to establish cell identity by coordinating and maintaining established cellular differentiation programs. T-bet, Gata3, RORγt, and Foxp3 are master transcription factors of TH1,TH 2, TH 17 and Treg differentiation programs, respectively.15–20 Generally, master transcription factors facilitate lineage commitment by promoting the gene expression program of one TH lineage while simultaneously repressing the developmental programs of opposing TH lineages. Transcriptional regulation of TH cell differentiation programs is further reinforced by a complex network of accessory transcription factors, which cooperate in the fine-tuning of feedforward or cross-inhibitory transcriptional circuits that modulate the duration, magnitude or specificity of CD4+ TH responses.2
In mounting effective host immunity towards diverse microbial pathogens, transcriptional regulation of CD4+ TH cell responses ensures the effective elimination of pathogens, while preventing robust CD4+ T cell activity from causing excessive self-damage. Here, we review the current understanding of molecular mechanisms that regulate CD4+ TH cell differentiation and their functional plasticity in health and in the context of immune-mediated diseases.
2 |. TRANSCRIPTIONAL REGULATION OF TH 1 CELLS
2.1 |. Molecular basis of TH1 polarization
The immune response activities of CD4+ TH1 cells are largely mediated through the production of their signature cytokine, IFN-γ.21 The importance of IFN-γ in the immune system stems from its ability to enhance immunogenicity of tumor cells, directly inhibit viral replication, upregulate MHC Class I and MHC Class II protein expression, activate microbicidal mechanisms in macrophages, and recruit inflammatory cells to the site of inflammation. Thus, through IFN-γ production, TH1 cells simultaneously regulate multiple facets of immune system activation and immunoregulation. Differentiation of CD4+ T cells into IFN-γ-producing TH1 cells is under the control of T-bet, the master regulator of the T 1 differentiation program.15 Initially, T-bet is induced in response to TCR stimulation and IFN-γ-STAT1 signaling.5,6,8 Because T-bet transactivates the Ifng gene, it establishes an IFN-γ-STAT1-T-bet feedforward loop that ensures more IFN-γ and T-bet expression. In this aspect, IFN-γ functions not only as an effector cytokine, but also as an autocrine TH1-polarizing signal.8 Even though IFN-γ is a potent inducer of T-bet, it cannot drive TH1 differentiation in the absence of IL-12.22 Following termination of TCR signaling and under the influence of IL-2, T-bet, and STAT5 induce the expression of Il12rb (encoding IL-12Rβ), enabling developing TH1 cells to respond to IL-12. Full stabilization of the polarized TH1 phenotype is then established by IL-12-induced STAT4 activation, which induces a second wave of sustained T-bet expression.8,23 Thus, a two-step model of positive feedback regulation ensures that only CD4+ T cells which received proinflammatory signals that continue beyond initial antigen-recognition, go on to fully commit to the TH 1 cell lineage.24 T-bet-mediated transactivation of the Ifng gene is enhanced by accessory transcription factors, Runx3 and HLX, which interact with T-bet to promote heritable TH 1 gene expression.25,26 T-bet also controls the expression of genes encoding CXCR3 and chemokines responsible for the mobilization of leukocytes to the site of inflammation.27 Accordingly, T-bet-deficient mice show increased susceptibility to infections with intracellular pathogens due to impaired TH1 cell differentiation and diminished recruitment of effector cells to the site of challenge.21
In addition to promoting the expression of TH1 cell-specific genes, T-bet reinforces the TH1 cell differentiation program by concomitantly inhibiting alternative TH cell differentiation pathways. T-bet accomplishes this either by suppressing the induction of other lineage specifying transcription factors or by interfering with their transcriptional activity.28 For example, T-bet heterodimerizes with the TFH cell specific master regulator Bcl6 and hijacks its transcriptional repressor activities for effective suppression of alternative helper T cell gene programs.29 T-bet inhibits the TH2 developmental program by binding directly to the TH2 cell-specific master transcription factor, Gata3, and preventing it from transactivating TH 2 cell-specific genes.30 T-bet can also directly repress de novo expression of Gata3 by binding directly to the regulatory region in the Gata3 locus and promoting the deposition of repressive epigenetic marks.31 Additionally, T-bet-Runx3 transcriptional complexes silence Il4 gene expression and, thus, prevent expression of the TH2 cell-polarizing cytokines during TH1 differentiation.25 Likewise, T-bet effectively inhibits commitment to the TH17 cell lineage by blocking Runx1-mediated induction of the TH17 cell-specific master transcription factor, RORγt.32 However, T-bet expression in already committed TH17 cells results in their conversion into IFN-γ-producing TH17 cells, which are strong drivers of TH17 cell-mediated pathology (discussed in below sections).
Although in vitro studies have identified IL-12 and IFN-γ as central cytokine regulators of the TH1 differentiation program, not all TH1 cell responses require IL-12 and IFN-γ in vivo. For example, IL-12 is not required for the generation of TH1 cells following infections with Mycobacterium avium, lymphocytic choriomeningitis virus (LCMV), vesicular stomatitis virus (VSV), and mouse hepatitis virus (MHV) infections while IFN-γ-STAT1 signaling is not essential for the generation of TH1 cell responses following Toxoplasma gondii infection.33,34 These studies suggest that signals other than IL-12 and IFN-γ can instruct differentiation of TH1 cells in vivo. In this context, it has been shown that microbial products can induce the expression of Delta-like ligands (DLLs) on antigen presenting cells, which upon binding to Notch3 on CD4+ T cells promote translocation of the intracellular Notch to the nucleus where it can directly activate the transcription of Tbx21 gene in an recombination-signal-binding protein for immunoglobulin-κ region (RBPJ)-dependent or NF-κB (p50/p65) dependent manner.35,36
2.2 |. Transcriptional regulation of pathogenic TH1 cells
Many of the immune-mediated diseases that were previously associated with TH1 effector cells are, in fact, caused by IL-23-dependent TH17 cells. In this context, the pathogenicity of TH17 cells was shown to be tightly linked to the expression of the TH1-specific master transcription factor, T-bet, and the acquisition of TH1-specific effector functions. The confusion in the field was caused by the studies which showed that p40-deficiency results in resistance of mice to autoimmunity.37 Since p40 encodes a shared subunit of IL-12 and IL-23, it was not until the generation of p19-deficient mice that it was convincingly established that IL-23 – and not IL-12 – drives autoimmune inflammation and generation of pathogenic TH17 cells.38,39 Nevertheless, there are a few diseases that develop due to strong TH1 cell responses caused by dysregulated expression or transcriptional activity of T-bet (Fig. 2). For example, T-bet expression in TH1 cells contributes to the pathogenesis of Crohn’s disease.40–43 Several groups have detected higher T-bet expression and augmented IFN-γ production in the lamina propria of CD4+ T cells isolated from patients with Crohn’s disease, but not in those of patients with ulcerative colitis or healthy controls.41–43 Higher T-bet expression is also detected in mouse models of chronic intestinal inflammation.40 Additionally, T-bet-expressing TH1 cells play a major role in the pathogenesis of autoimmune type 1 diabetes and chronic obstructive pulmonary disease (COPD).44–46
FIGURE 2. Transcriptional regulation of CD4+ T helper (TH) cells that mediate tissue inflammation.
CD4+ TH cells are a heterogeneous population of effector cells with a significant degree of genetic plasticity that enables them to acquire effector functions and the cytokine profiles of the opposing TH lineages. Generally, CD4+ TH plasticity is associated with more aggressive CD4+ TH responses and destructive inflammation-associated pathology. For example, TH17 cells trans-differentiate into TH1-like TH17 cells in an IL-23-dependent manner and drive tissue pathology in multiple sclerosis, inflammatory bowel disease, diabetes, and autoimmune arthritis. Similarly, uncommitted Treg cells, characterized by low Foxp3 and CD25 expression, can convert into IL-17A-producing effector cells and upregulate RANKL expression, which increases their osteoclastogenic activity. Such TH17-like Treg cells not only lost their suppressive functions, but also accelerated arthritis pathology. Future studies should be focused to elaborate and functionally characterize transcription factors that regulate the generation and functional plasticity of pathogenic CD4+ TH effector cells
In contrast to other TH cells, TH1 cells appear to be terminally differentiated due to the dominant nature of the T-bet-STAT4 and Runx3 transcriptional regulatory network in maintaining the stability of the TH 1 differentiation program.21 For this reason, effective modulation of TH1-mediated autoimmunity represents a considerable clinical challenge as TH1 cells may not be as malleable as other TH subsets to manipulation. Pharmacological targeting of transcription factors is a daunting task since nuclear delivery of small molecule inhibitors is technically challenging to achieve. Furthermore, targeting the master regulator of a type 1 immune response may render patients immunocompromised and susceptible to infections. Careful analyses of immune responses against Toxoplasma gondii and Leishmania major have revealed TH1 cells that co-express IFN-γ and IL-10 in the late stage of infection.47,48 The appearance of IFN-γ and IL-10 co-producing TH1 cells was induced by the parasites, and in the case of T. gondii, IL-10-producing TH1 cells reduced immunopathology while in the case of L. major they promoted parasite persistence due to the immunosuppressive effects of IL-10 on the maintenance of TH1 responses.47,48 Therefore, induction of IL-10 production or exogenous administration of recombinant IL-10 could be the most effective strategy in the treatment of TH1 mediated pathologies. In this context, a recent study reporting that the transcription factor Bhlhe40 acts a suppressor of the Il10 gene in TH1 cells is of high significance.49 CD4+ T cell-specific deletion of Bhlhe40 resulted in the reprograming of proinflammatory TH1 cells into anti-inflammatory, IL-10-producing TH1 cells.49 Thus, understanding the molecular mechanisms that silence Bhlhe40 gene expression in TH1 cells may offer new drug targets aimed at limiting TH1-mediated inflammation.
3 |. TRANSCRIPTIONAL REGULATION OF TH 2 CELLS
3.1 |. Molecular basis of TH2 polarization
Shortly after activation in the lymph nodes, newly differentiated CD4+ TH2 cells migrate to the site of parasite invasion, where they initiate protective anti-helminth immune responses primarily through the production of TH2-associated cytokines: IL-4, IL-5, IL-9, and IL-13.TH2 cell-derived cytokines have pleotropic effects, which include stimulation of immunoglobulin E secretion by B cells (through IL-4), recruitment of eosinophils and mast cells (through IL-5 and IL-9, respectively), and increased mucus production, smooth muscle cell contractility, and vascular permeability (through IL-4, IL-9, and IL-13). Collectively, these type 2 effector mechanisms culminate in the “weep and sweep” response leading to parasite expulsion.50–52 Because extensive tissue damage may occur during infection or parasite expulsion, TH2 cell-specific cytokines also promote tissue repair by inducing differentiation of wound-healing (M2) macrophages, which stimulate myofibroblast activation, angiogenesis, and extracellular matrix deposition.53 However, the same TH2 cell-specific effector responses, if inappropriately activated or insufficiently controlled during chronic exposure to innocuous allergens, can induce deleterious structural changes in affected tissues, characterized by pronounced collagen deposition and fibrosis. In this context, overzealous TH2 cells have an established role as key instigators and drivers of immunopathology in asthma, allergy, and atopic dermatitis. Thus, dissecting the molecular basis of TH2 polarization is important for understanding the pathogenesis of TH2-mediated diseases and for finding new avenues for therapeutic targeting of TH2 cells in asthma and allergy.51
CD4+ TH2 differentiation is initiated following T cell receptor stimulation in response to IL-2 and IL-4 cytokines. Signals emanating from the TCR induce the assembly of core transcription factors BATF-AP-1-IRF4 at the AP-1-IRF4 composite elements (AICE) in the Il4 and Gata3 promoters, leading to the initiation of Il4 and Gata3 expression in CD4+ TH precursors.54–56 Importance of this transcriptional event is demonstrated by limited IL-4 production and TH2 differentiation in mice that lack Irf4, c-Maf, and Junb expression.54,55,57,58 Furthermore, IL-2 signaling activates STAT5 transcriptional activity, which induces Il4 and Il4ra gene expression, thus, increasing the responsiveness of developing TH2 cells to autocrine and paracrine IL-4 stimulation.10,11,59 The key function of IL-4 signaling is the activation of a core transcription factor, STAT6, which changes the genetic landscape of TH2-specific loci and induces expression of the TH2-specific master transcription factor, Gata3.9,60,61 Studies demonstrating that ectopic expression of Gata3 is necessary and sufficient to trans-differentiate polarized TH1 cells into IL-4-producing TH2 cells and that Gata3-deficiency results in defective TH2 cell commitment have placed Gata3 on the throne of the TH 2 differentiation program.20,62,63 Gata3 orchestrates diverse transcriptional events during TH2 differentiation, including augmentation of IL-4 expression by binding to the Il4 enhancer.64 Increased IL-4 signaling further enhances STAT6-mediated transcription of Gata3, thus generating a positive feedback loop that reinforces Gata3 expression in developing TH2 cells. Once a critical level of Gata3 is achieved, Gata3 autoactivates its own expression and ensures sustained expression of Gata3 in TH2 cells even when IL-4 signaling ceases.65 Furthermore, Gata3 is solely responsible for transcription of Il5 and Il13 genes, as Gata3-deficient CD4+ T cells do not produce IL-5 and IL-13 cytokines.62,66,67 Stability and maintenance of the TH2 phenotype is dependent on Gata3-mediated suppression of the TH1 lineage developmental program by inhibiting Stat4 and Il12rb gene expression, rendering TH2 cells unresponsive to IL-12 signaling.68 Additionally, Gata3 interacts with Runx3 and prevents Runx3-mediated upregulation of Eomesodermin and Ifng genes.69 Although Gata3 executes pleotropic functions, it does not act alone during TH2 differentiation. Accessory transcription factors such as Dec-2 and Tcf1 aid in establishing the genetic program of TH2 cells by promoting expression of positive regulators of the TH2 differentiation program (Gata3, Junb, Il2, and Il4) while transcription factors Tcf1, Gfi, and Ikaros inhibit induction of TH1 and TH17 cell-specific genes during TH2 differentiation.70–75
Although IL-4-STAT6 signaling pathway is essential for TH2 polarization in vitro, CD4+ TH2 responses can be generated in IL-4Rα-deficient and STAT6-deficient mice under certain inflammatory conditions.76–80 Analogous to the differentiation of IL-12-STAT4 independent TH1 cells, studies have demonstrated that the Notch pathway can provide signals for the differentiation of IL-4-STAT6 independent TH2 cells. The induction of TH2 cell differentiation depends on RBPJ and Notch1 or Notch2, which directly bind to the Gata3 promoter and a 3′ enhancer of Il4.35,81,82 Mice that lack either RBPJ or Notch1 and Notch2 expression have impaired TH2 responses to parasite antigens.82 Collectively, these studies demonstrate the importance of the Notch signaling pathway in promoting the generation of cytokine-independent TH2 cells in vivo.
3.2 |. Transcriptional regulation of pathogenic TH2 cells
Dysregulated CD4+ TH2 cell responses are responsible for the pathogenesis of asthma, which affects 300 million people worldwide.83 Specifically, CD62LloCXCR3lo memory TH2 cells, which produce excessive amounts of IL-5, and amphiregulin-expressing ST2hi memory TH2 cells have recently been shown to drive severe pathology in mouse models of allergic airway inflammation by stimulating eosinophils to produce increased amounts of a key profibrotic factor osteopontin.84,85 Although TH2 cell-specific transcription factors generally suppress expression of TH1-specific transcription factors, CD4+ TH2 cells with intermediate levels of Gata3 express low amounts of T-bet and Eomesodermin, which appear to have important immunoregulatory functions. By binding to Gata3, T-bet sequesters Gata3 away from Gata3 DNA binding sites in the TH 2 cytokine locus.30 Thus, T-bet-deficient mice develop spontaneous airway changes and exhibit more severe IL-13-induced fibrosis in the lungs than wild-type mice during allergic airway inflammation due to increased expression of TH 2 signature cytokines.86,87 Additionally, low levels of Eomesodermin are sufficient to prevent Gata3 binding to the Il5 promoter and inhibit development of IL-5-hyperproducing pathogenic TH2 cells.85 Interestingly, CD62LloCXCR3lo memory TH2 cells, that produce excessive levels of IL-5, have selectively silenced Eomes gene expression.85 Pathogenicity of TH2 cells is further enhanced by their functional plasticity and acquisition of effector functions normally associated with TH17 cells.88,89 Such TH2/TH17 cells, that co-express Gata3 and RORγt and co-produce TH2 and TH17 signature cytokines, are readily detected in the bronchoalveolar lavage samples of patients with severe forms of asthma that is refractory to steroid treatment.89 Thus, the transcriptional state of pathogenic TH2 cells in asthmatic patients is characterized by complete silencing of TH1 cell-specific transcription factors, T-bet and Eomesodermin, which may or may not be accompanied by upregulation of the TH17 cell-specific transcription factor, RORγt (Fig. 2).
Uncontrolled persistence of TH2 cell responses exacerbates the pathology of allergy and asthma. Since even low levels of TH1 cell-specific transcription factors, T-bet and Eomesodermin, can reduce the pathogenicity of TH2 cells, one could harness functional plasticity of TH2 cells to reprogram them into nonpathogenic TH1-like TH2 cells. However, a major challenge to this specific therapeutic strategy is the lost sensitivity of TH2 cells to signaling by the TH1 inducing cytokine IL-12 due to Gata3-mediated downregulation of Stat4 and Il12rb.68,90 Although challenging, functional reprograming of unfavorably differentiated TH2 cells into TH1-like TH2 cells can be achieved by simultaneous TCR stimulation and concerted Type I and Type II IFN and IL-12 signaling.90 In this context, stable TH2/TH1 cells can be maintained in vivo for months and co-existence of TH2/TH1 cell differentiation programs within CD4+ TH cells has beneficial effects by providing a fine balance between protective immune responses while preventing immunopathology.90 Administration of specific Toll-like receptor (TLR) ligands, such as TLR7 and TLR9 agonists, could be employed to repolarize asthma-inducing TH2 cells into anti-inflammatory TH1-like TH2 cells.51 Furthermore, pharmacological targeting of TH2 cell-specific transcription factors, such as Gata3 and STAT6, could have profound beneficial therapeutic effects by preventing differentiation of TH2 cells with pathogenic effector functions.51
4 |. TRANSCRIPTIONAL REGULATION OF TH 9 CELLS
4.1 |. Molecular basis of TH9 polarization
Mouse studies have provided strong evidence for potent anti-helminth and anti-tumor activity of TH9 cells, mediated primarily by its signature cytokine IL-9. IL-9 drives the expulsion of Trichuris muris,Nippostrongylus brasiliensis, and Trichinella spiralis by increasing the intestinal muscle contraction, mast cell activation, and epithelial cell mucus production.91 Adoptive transfer of TH9 cells is sufficient to protect susceptible Il9-deficient mice from N. brasiliensis infection, demonstrating the importance of TH9 cell-derived IL-9 in driving anti-helminth immune responses.92 Interestingly, TH9 cells also exhibit effective anti-tumor activity against melanoma and lung adenocarcinoma.93,94 The anti-tumor activity of TH9 cells is dependent on IL-3, IL-9, and IL-21.91 TH9 cell-derived IL-3 prolongs dendritic cell survival while IL-9 increases the anti-tumor activity of mast cells.93,95 Additionally, exposure of TH9 cells to IL-1β results in IRF-1-mediated augmentation of IL-21 secretion from TH9 cells, which enhances IFN-γ production and the anti-tumor activity of NK cells and CD8+ T cells.94
It is becoming increasingly clear that in addition to these beneficial activities, TH9 cells can have detrimental functions in allergic, skin and intestinal inflammatory diseases (Fig. 2). Genetic studies have identified the link between the genes related to TH9 cell function and increased incidence of asthma and allergies in humans.91 Mouse studies indicate that TH9 cells are superior to TH2 cells in stimulating cytokine production by type 2 innate lymphoid cells (ILC2), promoting mast cell and eosinophil activation and increasing bronchial hyperresponsiveness.91 Increased IL-9 production and the frequency of circulating TH9 cells are also detected in patients with atopic dermatitis.96 Neutralization of IL-9 abrogates the ability of TH9 cells to induce atopic disease in mice, suggesting that TH9 cells promote disease primarily through IL-9 production.97,98 Likewise, there is a strong association between enhanced TH9 responses and a more severe form of inflammatory bowel disease (IBD).96,99,100 In mice, TH9 cells are sufficient to cause severe colitis following the adoptive transfer while Il9 deficiency or neutralization of IL-9 effectively reduces intestinal pathology.99 Functionally, it was demonstrated that TH9 cell-derived IL-9 disrupts the epithelial cell barrier functions and impairs mucosal wound healing in vivo.99 Thus, TH9 cells have a strong colitogenic potential to drive intestinal inflammation.
A complex regulatory network of transcription factors controls TH9 cell differentiation. None of the following transcription factors are TH9-specific; however, their combinatorial effects culminate into a unique TH9 gene expression program that shares very few similarities with TH1, TH2, TH17, and Treg cells. Differentiation of IL-9-secreting TH cells is dependent on IL-2-STAT5, IL-4-STAT6, and TGF-β-signaling.91 IL-2 promotes TH9 differentiation by activating STAT5, which binds directly to the Il9 promoter and blocks the binding of the TFH-specific transcription factor Bcl6.101 Accordingly, loss of IL-2 or pharmacological inhibition of STAT5 transcriptional activity results in impaired TH9 differentiation.101,102 IL-4 and STAT6 promote TH9 differentiation by blocking the generation of TGF-β-induced Foxp3+ Treg cells and T-bet-expressing TH1 cells.103,104 Additionally, IL-4-STAT6 signaling induces the expression of transcription factors BATF and IRF4, both of which bind directly to the Il9 locus and coordinate the expression of STAT6-regulated genes in TH9 cells.97,105 Genetic deletion of STAT6, IRF4, or BATF results in impaired differentiation of TH9 cells and increased expression of T 1 cell-associated genes.97,103,105 Since IL-2-STAT5 and IL-4-STAT6 signaling pathways drive TH2 cell differentiation, TGF-β is critically important for the suppression of TH2 cell-specific genes in TH 9 cells.104,106 Furthermore, TGF-β-activated SMAD2/3/4 transcription factors can promote TH9 cell differentiation by binding independently or in a complex with IRF4 to the Il9 locus. Another critical function of TGF-β is induction of the transcription factor PU.1.107 PU.1 deficiency in CD4+ T cells results in markedly impaired IL-9 production, whereas ectopic expression of PU.1 in either TH2 or TH9 cells increases IL-9 production.107 Mechanistically, PU.1 binds directly to the Il9 promoter and recruits the histone acetyltransferase GCN5 to induce Il9 promoter activity.108 Recently, three independent studies have reported a novel function of the transcription factor Foxo1 in regulating the development of TH 9 cells.109–111 Activation of the PI3K/AKT pathway results in Foxo1 upregulation in CD4+ T cells, in which Foxo1 directly induces IRF4 and IL-9 expression while simultaneously inhibiting the expression of TH17-cell specific genes.110
Since none of the aforementioned transcription factors are uniquely expressed in TH9 cells, the differentiation of TH9 cells is not under the control of a defined lineage-specifying master transcription factor, but rather an end-product of synergistic and antagonistic actions of multiple transcription factors, which ensure Il9 gene expression and concomitant suppression of alternative TH fates.
4.2 |. Transcriptional regulation of pathogenic TH9 cells
Since proinflammatory functions of TH9 cells map to the expression of IL-9, transcription factors which transactivate Il9 gene directly support the development of pathogenic TH9 cells. As expected, deficiency of IRF4, BATF, PU.1, or Foxo1 in CD4+ T cells results in ameliorated TH9 dependent asthma, allergic inflammation, and colitis.97,99,105,107,110,111 Additionally, administration of neutralizing IL-9 antibody protected the recipient mice from TH9-mediated pathologies, demonstrating that IL-9-targeted therapy may be particularly successful in limiting TH9 driven inflammatory responses.
5 |. TRANSCRIPTIONAL REGULATION OF TH 17 CELLS
5.1 |. Molecular basis of TH17 polarization
Although TH17 cells can be induced in multiple tissues under different inflammatory conditions, in the steady state, they are most commonly found at barrier sites such as intestine, lung, and skin. Here, TH17 cells play an important role in providing protective barrier functions against fungal and bacterial pathogens and for maintaining host–commensal homeostasis. The prototypic barrier functions of TH17 cells are mediated by the production of TH17 cell signature cytokines (IL-17A, IL-17F, IL-21, and IL-22), which induce a broad range of tissue responses including the expression of anti-microbial peptides (lipocalin-2, Reg3β, Reg3γ, S100A8, S100A9), proinflammatory cytokines (IL-1β, IL-6,TNF-α, GM-CSF), CXC chemokines (CXCL8, CXCL1, CXCL2, CXCL10), and metalloproteases.112 Accordingly, TH17 cell-mediated effector mechanisms are indispensable in providing protection against Klebsiella pneumoniae, Staphylococcus aureus, Propionibacterium acnes, Bacteroides spp., Borrelia spp., Candida albicans, and Pneumocystis carinii.113 The broad distribution of IL-17R and IL-22R results in a massive tissue response to TH17 cell-derived effector cytokines, which may explain the prominent ability of TH17 cells to cause severe tissue inflammation.113 Indeed, TH17 cells have emerged as an important mediator in inflammatory and autoimmune diseases. Strong clinical evidence points to the pathogenic functions of TH17 cells in psoriasis, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, and asthma.113 Systems biology approaches have been instrumental in identifying novel transcriptional networks that regulate the differentiation of protective and pathogenic TH17 cells.114,115
Because both TH17 cells and Treg cells require TGF-β for their development, the TH17 cell transcriptional program is functionally coupled to the transcriptional program of regulatory (Treg) cells, a feature unique to the TH17 lineage.116–120 When activated in the presence of TGF-β, CD4+ T cells transiently co-express two antagonizing transcription factors, Foxp3 and RORγt, which direct the differentiation program of regulatory T (Treg) cells and TH17 cells, respectively.16,17,121,122 From this intermediate Foxp3+RORγt+ stage, a CD4+ T cell has the potential to develop into either the Treg or the TH17 lineage. Their ultimate fate is determined by the relative abundance of TGF-β and IL-6 in the microenvironment. At high TGF-β concentrations, Foxp3 inhibits RORγt transcriptional activity, thereby tipping the balance away from the TH17 transcriptional program towards Treg cell differentiation. At low TGF-β concentrations, TGF-β synergizes with signals initiated by IL-6, which relieves RORγt from Foxp3-mediated suppression through STAT3 transcriptional activity.121–123 Thus, IL-6-mediated activation of STAT3 and RORγt is critical for the initiation of the TH17 lineage specification. Another important aspect of IL-6-STAT3 signaling is the induction of IL-21, which, in a STAT3-dependent manner, induces its own transcription and promotes the expression of the IL-23 receptor. IL-23, produced by activated antigen presenting cells, acts in synergy with IL-6, IL-21, and TGF-β to amplify RORγt-dependent terminal differentiation of TH17 cells.13,124 An extensive network of transcription factors, including IRF4-AP-1-BATF transcriptional complexes, IκBζ, Runx1, Ahr, and HIF-1α, is activated to stabilize the TH17 cell transcriptional program by ensuring optimal induction of RORγt or by promoting RORγt transcriptional activity at TH 17 cell-specific loci.125–127
Neither IL-6 nor IL-23 alone efficiently generate TH17 cells in the absence of TGF-β, demonstrating the importance of TGF-β signaling in murine TH17 cell differentiation. However, a combination of IL-6, IL-23, and IL-1β effectively supports the development of TGF-β-independent TH17 effector cells with enhanced pathogenic potential.128 These findings point to the alternative modes of TH17 cell differentiation in vivo, which can contribute to the heterogeneity of TH17 cells and their differing abilities to cause autoimmune pathologies.128
5.2 |. Transcriptional regulation of pathogenic TH17 cells
CD4+ T cells activated in the presence of IL-6 and TGF-β differentiate into IL-10-producing TH17 cells that lack pathogenic potential.129 For TH17 cells to become pathogenic, they must receive IL-23 signals or be differentiated in the presence of alternative cytokine combinations such IL-6, IL-1β, and IL-23.115,128,130 IL-23R signaling results in the repression of anti-inflammatory IL-10 production and induction of genes within the “proinflammatory transcriptional module” of TH17 cells.115,129 Thus, transcription factors that control IL-23R expression will also affect the pathogenic potential of TH17 cells. In addition to STAT3, the canonical Notch signaling mediator, RBPJ, was shown to directly enhance expression of the IL-23R and pathogenicity of TH17 cells.131
Interestingly, propagation of committed TH17 cells in the presence of IL-23 but without TGF-β induces the expression of the TH1-specific master transcription factor T-bet and promotes the emergence of IFN-γ-producing TH17 cells.128,132 Indeed, under chronic inflammatory conditions during experimental autoimmune encephalomyelitis, inflammatory bowel disease, autoimmune arthritis, and diabetes, TH17 cells trans-differentiate into TH1-like TH17 cells in an IL-23 dependent manner and drive tissue pathology (Fig. 2).132–137 Transition of TH17 cells into TH1-like effector cells is critically dependent on the transcription factors T-bet, Runx1, and STAT4. Accordingly, T-bet, Runx1, or STAT4 deficiencies impair the development of TH1-like TH17 cells and protect the mice from autoimmunity.135,138 However, not all TH17-mediated diseases are driven by the functional plasticity of TH17 cells. For example, TH17 cells exhibit exceptional phenotypic stability in the lungs, where they contribute to allergen-induced airway hyper-responsiveness by promoting neutrophilic airway inflammation and enhancing airway smooth muscle contractions.139–141 Similarly, psoriasis, a chronic inflammatory disease of the skin, is caused specifically by the TH17 cell-derived cytokine, IL-22. In psoriasis, IL-23 stimulates TH17 cells to secrete IL-22, which promotes dermal inflammation through STAT3 activation in keratinocytes.142 The pathology of dry eye disease, autoimmune uveitis, and scleritis is also driven by memory TH17 cells.143–146 Neutralization of IL-17A and IL-27/STAT1-mediated antagonism of the TH17 cell phenotype have demonstrated significant therapeutic benefits in the treatment of eye inflammation.143–145
IL-1β plays a unique and nonredundant role during differentiation of pathogenic TH17 cells. IL-1R signaling is essential for TH17 cell production of GM-CSF, which is the only cytokine proven to be critical for the encephalitogenicity of TH17 cells in the experimental autoimmune encephalomyelitis.147,148 IL-1β strongly induces the expression of the transcription factor Bhlhe40, which acts as a transcriptional activator of Csf2 (encoding GM-CSF) in TH17 cells. Additionally, Bhlhe40 represses IL-10 production by TH17 cells, demonstrating the importance of this IL-1β-induced transcription factor in regulating TH 17 cell pathogenicity.149,150 IL-1R expression is under strict negative control of a transcriptional repressor, Foxo1. Therefore, downregulation of Foxo1 by the miR-183-96-182 cluster is required to unleash the full pathogenic potential of TH17 cells in response to IL-1 signaling.151
Given the strong association of TH17 cells with multiple immunemediated diseases, it is not surprising that a substantial amount of research has been devoted to devising novel therapeutic strategies that can target TH17 differentiation. Since TH17 cells also have barrier protective functions, it is important that new therapies do not interfere with beneficial functions of TH17 cells that safeguard the intestine from potential pathogens.112 Although TH17 cell functional plasticity has been linked to TH17 cell pathogenicity, one could exploit this property of TH17 cells as a therapeutic opportunity for the treatment of inflammatory diseases.152 Indeed, TH17 cells can be redirected to adopt immunoregulatory functions and contribute to the resolution of intestinal inflammation in response to Ahr and TGF-β signaling.152 TH17 cells can also deviate towards TFH cell phenotype in Peyer’s patches where they support the generation of high-affinity antigen-specific IgA responses. In this way, TH17 cell-derived TFH cells contribute to the maintenance of the host/commensals equilibrium and constitute an effective arm of mucosal immune responses in the gut.153 Understanding the precise molecular mechanisms that regulate TH17 cell genetic and functional plasticity will broaden the repertoire of therapeutic approaches, which can harness T cell plasticity for the benefit of patients.154
6 |. TRANSCRIPTIONAL REGULATION OF TFH CELLS
6.1 |. Molecular basis of TFH polarization
TFH cells can be distinguished from all other TH subsets by elevated IL-21 production and high expression of surface receptors that are critical for their development and function, including CXCR5, PD-1, ICOS, CD40L, BTLA, and CD84.155 TFH cells control humoral immunity by supporting the formation of germinal centers in which TFH cells provide help to cognate B cells. Productive TFH–B cell interactions result in the development of long-lived, antigen-specific memory B cells and plasma cells. Proper regulation of TFH differentiation is necessary to enable generation of high-affinity neutralizing antibodies following vaccinations and infections; however, dysregulated TFH responses can lead to pathogenic autoantibody production and autoimmunity. Aberrant activation of TFH cells and increased autoantibody production are frequently observed in patients with systemic lupus erythematosus (SLE), Sjogren’s syndrome, rheumatoid arthritis, autoimmune thyroid diseases, type 1 diabetes, multiple sclerosis, neuromyelitis optica, Guillain-Barré syndrome, myasthenia gravis, and idiopathic thrombocytopenic purpura.156 Therefore, investigations of transcriptional mechanisms that control generation of TFH cells will provide us with a unique opportunity to identify molecules that could be targeted to attenuate TFH responses for the treatment of immunological diseases caused by excessive autoantibody production.155
Differentiation of TFH cells is a multistage process spanning three spatiotemporal phases: initial priming, T-B border commitment phase, and germinal center phase (reviewed in Ma et al.,155 Crotty,157 Qi,158, and King159). The initial priming of naïve CD4+ T cells occurs in T cell zones of lymphoid organs after recognition of peptide-MHC Class II complexes on dendritic cells. Expression of high-affinity T cell-receptor or activation of CD4+ T cells in the presence of high doses of antigen favors the differentiation of TFH cells.157,160,161 The initiation of TFH differentiation program is dependent on ICOS-ICOSL and IL-6-STAT3 signaling pathways, which induce Bcl6, c-maf and IL-21 expression in TFH precursors.162–165 IL-6 and TFH cell-derived IL-21 ensure continuous STAT3 activation necessary for the maintenance of TFH phenotype during different stages of differentiation.163,165 These initial interactions between dendritic cells and TFH precursors also produce phenotypic changes characterized by CXCR5 upregulation and CCR7 downregulation, which license TFH precursors to move out of the T cell zone towards T cell–B cell border where the critical B cell dependent phase of TFH differentiation occurs.166 A continuous dialogue between TFH cells and B cells, mediated by TCR-MHC Class II,ICOS-ICOSL, CD40-CD40L, and SAP-SLAM interactions, further stabilizes Bcl6 expression in the intermediate TFH cells. After entry into the B cell follicle, TFH cells complete their differentiation program in a B cell and ICOS-dependent manner.162 At this stage, germinal center TFH cells acquire their most polarized state characterized by the highest level of Bcl6, CXCR5, PD-1, ICOS, and SAP expression.157 Here,CXCR5hiPD-1hiBcl6hi TFH cells are needed to maintain germinal centers and facilitate differentiation of germinal center B cells into plasma cells and memory B cells.157
Although Bcl6 is the master regulator of the TFH cell differentiation program, the development of TFH cells requires hierarchical and integrated functions of numerous transcription factors.155,167–169 STAT3-activating cytokines, IL-6 and IL-21, induce the expression of BATF, which subsequently binds to and transactivates Bcl6 and c-maf genes.170,171 While primary function of c-maf is to induce Il21 expression, Bcl6 employs multiple mechanisms to imprint the TFH phenotype. Specifically, Bcl6 suppresses expression and/or function of transcriptional regulators of alternative TH lineages (T-bet, Gata3, and RORγt), Blimp-1, and the miR-17–92 cluster, which represses CXCR5 expression.167–169
6.2 |. Transcriptional regulation of pathogenic TFH cells
Resolution of the germinal center response after pathogen clearance is essential for the prevention of autoimmunity. Aberrant expression of TFH cell-associated cytokines and co-stimulatory molecules, particularly IL-21 and ICOS, has been directly linked to the development of lupus.172 Antibody-mediated neutralization of IL-21 or Il21r gene deficiency in different strains of lupus-prone mice significantly ameliorated autoimmune pathology, suggesting that IL-21-targeted therapies have a great therapeutic potential in terminating TFH-driven inflammatory processes.172 In mice, homozygosity for the “san” allele of Roquin, which encodes a post-transcriptional negative regulator of Icos, results in increased accumulation of TFH cells, spontaneous germinal center formation and lupus pathology.173 The pathogenic accumulation of TFH cells in Roquinsan/san mice is caused by excessive IFN-γ signaling, which promoted Bcl6 overexpression and hyperproliferation of TFH cells, suggesting that elevated IFN-γ production during infections carries a risk of triggering germinal center-driven autoimmunity.173
CD4+ T cells isolated from the salivary glands of Sjogren’s syndrome patients express bona fide TFH phenotype, characterized by high expression of BCL6, CXCR5, ICOS, and IL-21.174 Similarly, increased frequency of PD-1hiCXCR5+ TFH-like cells was detected in the circulation of multiple sclerosis patients during the relapse of disease.175 Using the experimental autoimmune encephalomyelitis model of multiple sclerosis, the authors have shown that CXCR5+ TFH-like cells and B cells infiltrate the CNS parenchyma where they form organized ectopic lymphoid structures. In these structures, TFH-like cells are juxtaposed with mature IgD+ B cells and CD138+ plasma cells, suggesting that ectopic lymphoid structures may be the precise location of continuous TFH-like cell–B cell interactions driving a germinal center-like response and chronic inflammation.175 In contrast, CD4+ T cells isolated from the synovium of rheumatoid arthritis patients expressed some, but not all, TFH-specific markers (PD-1hiICOS+SAP+MAF+ IL-21+CXCR5−BCL6−).176 These pathogenic PD-1hiCXCR5− CD4+ T cells provided B cell help in an IL-21 and SLAMF5-dependent manner.176 Although they have demonstrated helper functions normally associated with TFH cells, peripherally-expanded pathogenic TFH-like cells were negative for BCL6 expression,176 suggesting that they may not be susceptible to the negative regulation by the IL-2-STAT5-Blimp1 pathway like bona fide TFH cells.177,178 In this case, antibody-mediated blockade of PD-1, SLAMF5, and IL-21 may be the only therapeutic strategy for targeting tissue-specific T-B cell interactions.176
7 |. TRANSCRIPTIONAL REGULATION OF Treg CELLS
7.1 |. Molecular basis of Treg polarization
The “central tolerance” purging mechanisms of autoreactive T cells are imperfect, and a certain fraction of self-reactive T cells will escape thymic negative selection. The presence of self-reactive T cells in the circulation is a constant threat to the host. Aberrant activation of self-reactive T cells will inevitably result in fatal autoimmunity unless such a response is suppressed by regulatory T (Treg) cells.179 Like-wise, activated CD4+ TH cell responses may provoke an overexuberant, deleterious inflammatory cascade, unless the magnitude or duration of such a response is curtailed by Treg cells. Therefore, the primary function of CD4+ Treg cells is to maintain immunological homeostasis by suppressing excessive and destructive immune responses against innocuous, self and foreign antigens. Treg cells can be grouped into thymic (natural) Treg cells or peripheral (induced) Treg cells, depending on where they originate. Thymic selection of Treg cells requires strong TCR (upon recognition of MHC Class II-restricted self-peptides), CD28 co-stimulation and IL-2-STAT5 signaling.12 Gut-associated lymphoid tissue is a preferential site for the differentiation of peripheral T cells, where CD103+ reg dendritic cells induce Treg cell differentiation via a mechanism that is dependent on TGF-β and retinoic acid signaling.180,181
Development of thymic and peripheral Treg cells remains under the control of the master transcription factor, Foxp3, which plays a critical role in the establishment and maintenance of Treg lineage identity and suppressor functions.18,182–185 Ectopic expression of Foxp3 in CD4+ T cells is sufficient to endow them with Treg-suppressive functions, and conversely, genetic deletion of Foxp3 results in the loss of Treg cells and fatal autoimmunity.16,18,182 Because proper functioning of the immune system requires balanced activation of CD4+ TH effector responses, Foxp3 expression must be stringently regulated. A network of transcription factors, including Smad3, NFAT, c-Rel, Foxo1, Foxo3, Runx1, Stat5, CREB, and the Nr4a family of transcription factors, binds to regulatory regions in the Foxp3 gene locus; ensuring the timely, stable, and heritable cell-restricted expression of Foxp3 in Treg cells (reviewed in Lee and Lee186). Rather than generating a Treg-specific chromatin landscape, Foxp3 exploits a preformed network of accessible enhancers established by TCR signaling for promoting Treg cell differentiation.187 Only a small fraction of genes (~6%) in the entire Foxp3-dependent gene expression program is directly regulated by Foxp3, while the expression of the remaining genes is controlled indirectly through interactions with other transcription factors.183 Thus, Foxp3 forms transcriptional complexes with Eos, Runx1, and NFAT to activate (Il2ra, Ctla4) or repress (Il2, Ifng) target gene expression.188–190
Clearly, maintaining Treg cell identity is critically important for immune homeostasis. Because the TCR repertoire of thymic Treg cells is heavily biased toward autoreactivity,191 conversion of Treg cells into inflammatory effector cells could have dire consequences. Several transcriptional mechanisms are put in place to ensure that Treg cells are phenotypically and functionally stable under inflammatory conditions. In addition to regulating the induction of Foxp3 gene expression,Smad2/3 and Foxo1-mediated control of Treg cell specification includes active suppression of T-bet and IFN-γ.192–194 The transcription factor, musclin, promotes the unidirectional development of peripheral Treg cells by suppressing the TH 2 transcriptional program,195 while the transcriptional repressor Bach2 stabilizes Treg lineage identity by antagonizing differentiation programs of multiple TH effector subsets.196
Nevertheless, a certain degree of phenotypic and functional heterogeneity within the Treg population must be permitted for proper immunoregulation of qualitatively different types of inflammation induced by distinct TH1, TH2, and TH17 effector subsets. For example, a single gene deletion of either T-bet or Gata3 specifically in Treg cells does not affect Treg cell functionality and immune homeostasis in the steady state; however, combined deletion of T-bet and Gata3 leads to the loss of Foxp3 expression, Treg cell instability and spontaneous autoimmune lymphoproliferative disease.197 Several elegant studies have demonstrated that during polarized TH1-type inflammatory responses, Treg cells selectively upregulate T-bet expression, which is necessary for efficient Treg cell trafficking, proliferation, and survival.19,198 Following selective depletion of T-bet− Treg cells, the remaining T-bet-expressing Treg cells were fully sufficient to control TH 1 and CD8+ T cell responses while specific elimination of T-bet-expressing Treg cells resulted in severe TH1 autoimmunity.19 Similarly, in an autoimmune model of diabetes, CXCR3+ T-bet-expressing Treg cells played a crucial role in controlling autoimmune inflammation in the pancreas in a manner that could not be compensated by T-bet− Treg cells.199 These studies demonstrate that Treg cells may employ certain components of the T-bet/TH1-specific transcriptional machinery to fine-tune their functional properties under type 1 response inflammatory conditions.19 Similarly, Treg cell expression of IRF4, a transcription factor essential for TH2 effector cell differentiation, is required for Treg-mediated suppression of TH 2-type inflammatory responses,200 while the control of pathogenic TH17 cells is dependent upon Treg cell-restricted expression of STAT3, a transcription factor that controls TH17 effector cell differentiation.201 Furthermore, individual intestinal symbionts induce a distinct population of RORγt-expressing Treg cells, which contribute substantially to the regulation of intestinal TH2- and TH1/TH17 mediated inflammatory responses.202,203 Development and immune suppressive functions of RORγt-expressing Treg cells are dependent on the expression of another transcription factor c-maf, which integrates TGF-β receptor signals with pathobiont-induced cytokine dependent STAT3 activation to promote the differentiation of RORγt+ Treg cells.204 From these studies, a model has emerged in which selective expression of TH1, TH2-, and TH17-specific transcriptional regulators is necessary for functional and phenotypical specialization of Treg cells that are better equipped to restrain specific types of CD4+ T cell responses.205
7.2 |. Transcriptional regulation of pathogenic Treg cells
Recent studies have challenged the stability of the Treg cell lineage, demonstrating that Treg cells could be fully converted into inflammatory TH effector cells and become major drivers of immunopathology (Fig. 2). For example, CD25loFoxp3+ Treg cells isolated from the pancreas of diabetic mice have an activated memory phenotype, express TH1-like effector functions, and upon adoptive transfer into näive recipient mice cause a rapid onset of autoimmune type 1 diabetes, demonstrating that pathogenic ex-Treg cells could be generated under certain inflammatory conditions.206 In the context of food allergic responses, augmented IL-4R-STAT6 signaling is sufficient to promote the reprogramming of Treg cells towards a TH2-cell-like lineage. Consistent with this observation, peripheral Treg cells isolated from children with food allergies demonstrate increased expression of TH2-specific transcription factors (Gata3 and IRF4) and TH2 cellassociated cytokines.207 Treg conversion into TH2-cell-like lineage was critical for the development of food allergy and could be reversed by the blockade of IL-4/IL-4Rα signaling pathway in Treg cells.207 Additionally, in rheumatoid arthritis patients, CD4+ T cells isolated from the inflamed joints of patients are characterized by co-expression of Foxp3, RORγt, and IL-17A.208 Fate-mapping studies have revealed that a substantial proportion of arthritogenic CD4+ TH17 cells originated from CD25loFoxp3+ Treg cells.208 A local IL-6-rich inflammatory milieu, created by joint synovial fibroblasts, shifts the balance, not only by promoting de novo TH17 differentiation, but also by converting existing Foxp3+ Treg cells into IL-17A-producing effector cells. Interestingly, the Treg-derived TH17 cells express high levels of RANKL, a central cytokine for osteoclast activation and differentiation, and are more potent osteoclastogenic T cells than de novo differentiated TH17 cells.208 Thus, CD25loFoxp3+ Treg cells fail to suppress collagen induced arthritis in mice and accelerate arthritis pathology more efficiently, probably due to their higher degree of self-reactivity. Collectively, these studies demonstrate that under specific inflammatory conditions, Foxp3+ Treg cells can give rise to pathogenic TH1, TH2, and TH17 cells.
While expression of TH1-, TH2-, and TH17-specific transcriptional regulators promotes functional specialization of Treg cells that improves their immunoregulatory abilities, in other contexts, expression of the same transcriptional regulators can transform Treg cells into aggressive effector TH cell subsets. This paradox has raised the question of why certain Foxp3+ Treg cells cannot maintain their lineage identity under inflammatory conditions. Genetic fate-mapping approaches have revealed that there is a degree of heterogeneity within Treg cell population with respect to the level of Foxp3 and CD25 (IL-2Rα subunit) expression.206,208,209 Fully committed Treg cells, defined by high expression of Foxp3 and CD25, will remain stable and retain their suppressive functions in inflammatory environments, whereas uncommitted Treg cells, which express low or transient expression of Foxp3 and CD25, will acquire effector functions and become pathogenic.206,208,209 The availability of IL-2 and IL-2/STAT5 signaling is critically important for the maintenance of Foxp3 expression.210 Inevitably, immune system activation will result in limited IL-2 availability due to IL-2 consumption by CD4+ TH effector cells. This turned out to be the key destabilizing factor of Treg cells in a strong TH1-polarizing environment following Toxoplasma gondii infection.211 Importantly, the collapse of Treg cells can be rescued by IL-2 treatment, which stabilizes Foxp3 and CD25 expression in Treg cells.211,212 Thus, therapeutic approaches aimed at inducing or transferring Treg cells during the onset of autoimmune disease should consider administration of biologics that will shift the cytokine balance in favor of Treg cells. For instance, administration of IL-2 with concomitant blockade of IL-6 signaling might allow for the stabilization of Foxp3+ Treg cells in tissue during an ongoing pathogenic TH 17 cell response.208
8 |. CONCLUDING REMARKS
Originally, CD4+ TH cell differentiation was viewed as a linear and irreversible event whereupon each TH cell subset expressed its lineage specific transcription factors and unique signature cytokines. This functional specialization was deemed to be a necessary adaptation of the immune response to ensure that the appropriate effector mechanisms of each CD4+ TH subset are tailored to respond to a specific challenge. However, with the emergence of more sophisticated technologies, it became apparent that CD4+ TH cells are a heterogeneous population of effector cells with a significant degree of genetic plasticity that enables them to acquire effector functions and the cytokine profiles of the opposing TH lineages. Generally, CD4+ TH plasticity is associated with more aggressive CD4+ TH responses and destructive inflammation-associated pathology. In this perspective, CD4+ TH17 cells have been notoriously detrimental; responsible for causing and sustaining tissue damage in various models of organ-specific autoimmunity. Despite the indisputable role of TH17 cells in driving chronic inflammation, a complete knowledge of what makes TH17 cells so destructive is lacking. There is a substantial gap in our understanding of the origin of pathogenic TH17 cells. Are all TH17 cells capable of acquiring pathogenic effector functions if they receive appropriate signals in a specific tissue microenvironment, and is this phenotype reversible? Are there two different functional subsets of TH17 cells – protective TH17 cells at mucosal sites and pathogenic TH17 cells as key drivers of autoimmunity? With these questions in mind, it will be necessary to further elaborate and functionally characterize transcription factors that regulate the generation and functional plasticity of pathogenic CD4+ TH effector cells. Furthermore, ongoing developments in understanding the transcriptional regulation of CD4+ TH responses will foster new translational opportunities for therapeutic interventions in the treatment of immune-mediated diseases.
REFERENCES
- 1.Christie D, Zhu J. Transcriptional regulatory networks for CD4 T cell differentiation. Curr Top Microbiol Immunol. 2014;381:125–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Zhu J, Paul WE. Peripheral CD4+ T-cell differentiation regulated by networks of cytokines and transcription factors. Immunol Rev. 2010;238:247–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations (*). Annu Rev Immunol. 2010;28:445–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.O’Shea JJ, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science. 2010;327:1098–1102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Afkarian M, Sedy JR, Yang J, et al. T-bet is a STAT1-induced regulator of IL-12R expression in naive CD4+ T cells. Nat Immunol. 2002;3:549–557. [DOI] [PubMed] [Google Scholar]
- 6.Lighvani AA, Frucht DM, Jankovic D, et al. T-bet is rapidly induced by interferon-gamma in lymphoid and myeloid cells. Proc Natl Acad Sci U S A. 2001;98:15137–15142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mullen AC, High FA, Hutchins AS, et al. Role of T-bet in commitment of TH1 cells before IL-12-dependent selection. Science. 2001;292:1907–1910. [DOI] [PubMed] [Google Scholar]
- 8.Schulz EG, Mariani L, Radbruch A, Hofer T. Sequential polarization and imprinting of type 1 T helper lymphocytes by interferon-gamma and interleukin-12. Immunity. 2009;30:673–683. [DOI] [PubMed] [Google Scholar]
- 9.Kaplan MH, Schindler U, Smiley ST, Grusby MJ. Stat6 is required for mediating responses to IL-4 and for development of Th2 cells. Immunity. 1996;4:313–319. [DOI] [PubMed] [Google Scholar]
- 10.Liao W, Schones DE, Oh J, et al. Priming for T helper type 2 differentiation by interleukin 2-mediated induction of interleukin 4 receptor alpha-chain expression. Nat Immunol. 2008;9:1288–1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhu J, Cote-Sierra J, Guo L, Paul WE. Stat5 activation plays a critical role in Th2 differentiation. Immunity. 2003;19:739–748. [DOI] [PubMed] [Google Scholar]
- 12.Littman DR, Rudensky AY. Th17 and regulatory T cells in mediating and restraining inflammation. Cell. 2010;140:845–858. [DOI] [PubMed] [Google Scholar]
- 13.Zhou L, Ivanov II, Spolski R, et al. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol. 2007;8:967–974. [DOI] [PubMed] [Google Scholar]
- 14.Sundrud MS, Nolan MA. Synergistic and combinatorial control of T cell activation and differentiation by transcription factors. Curr Opin Immunol. 2010;22:286–292. [DOI] [PubMed] [Google Scholar]
- 15.Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, Glimcher LH. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell. 2000;100:655–669. [DOI] [PubMed] [Google Scholar]
- 16.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 2003;4:330–336. [DOI] [PubMed] [Google Scholar]
- 17.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;126:1121–1133. [DOI] [PubMed] [Google Scholar]
- 18.Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol. 2003;4:337–342. [DOI] [PubMed] [Google Scholar]
- 19.Levine AG, Mendoza A, Hemmers S, et al. Stability and function of regulatory T cells expressing the transcription factor T-bet. Nature. 2017;546:421–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Zheng W, Flavell RA. The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells. Cell. 1997;89:587–596. [DOI] [PubMed] [Google Scholar]
- 21.Lazarevic V, Glimcher LH. T-bet in disease. Nat Immunol. 2011;12:597–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Seder RA, Gazzinelli R, Sher A, Paul WE. Interleukin 12 acts directly on CD4+ T cells to enhance priming for interferon gamma production and diminishes interleukin 4 inhibition of such priming. Proc Natl Acad Sci U S A. 1993;90:10188–10192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Liao W, Lin JX, Wang L, Li P, Leonard WJ. Modulation of cytokine receptors by IL-2 broadly regulates differentiation into helper T cell lineages. Nat Immunol. 2011;12:551–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Leavy O The TH1 two step. Nat Rev Immunol. 2009;9:387. [Google Scholar]
- 25.Djuretic IM, Levanon D, Negreanu V, Groner Y, Rao A, Ansel KM. Transcription factors T-bet and Runx3 cooperate to activate Ifng and silence Il4 in T helper type 1 cells. Nat Immunol. 2007;8:145–153. [DOI] [PubMed] [Google Scholar]
- 26.Mullen AC, Hutchins AS, High FA, et al. Hlx is induced by and genetically interacts with T-bet to promote heritable T(H)1 gene induction. Nat Immunol. 2002;3:652–658. [DOI] [PubMed] [Google Scholar]
- 27.Lord GM, Rao RM, Choe H, et al. T-bet is required for optimal proinflammatory CD4+ T-cell trafficking. Blood. 2005;106:3432–3439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Lazarevic V, Glimcher LH, Lord GM. T-bet: a bridge between innate and adaptive immunity. Nat Rev Immunol. 2013;13:777–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Oestreich KJ, Huang AC, Weinmann AS. The lineage-defining factors T-bet and Bcl-6 collaborate to regulate Th1 gene expression patterns. J Exp Med. 2011;208:1001–1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hwang ES, Szabo SJ, Schwartzberg PL, Glimcher LH. T helper cell fate specified by kinase-mediated interaction of T-bet with GATA-3. Science. 2005;307:430–433. [DOI] [PubMed] [Google Scholar]
- 31.Zhu J, Jankovic D, Oler AJ, et al. The transcription factor T-bet is induced by multiple pathways and prevents an endogenous Th2 cell program during Th1 cell responses. Immunity. 2012;37: 660–673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lazarevic V, Chen X, Shim JH, et al. T-bet represses T(H)17 differentiation by preventing Runx1-mediated activation of the gene encoding RORgammat. Nat Immunol. 2011;12:96–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lieberman LA, Banica M, Reiner SL, Hunter CA. STAT1 plays a critical role in the regulation of antimicrobial effector mechanisms, but not in the development of Th1-type responses during toxoplasmosis. J Immunol. 2004;172:457–463. [DOI] [PubMed] [Google Scholar]
- 34.Szabo SJ, Sullivan BM, Peng SL, Glimcher LH. Molecular mechanisms regulating Th1 immune responses. Annu Rev Immunol. 2003;21: 713–758. [DOI] [PubMed] [Google Scholar]
- 35.Amsen D, Antov A, Flavell RA. The different faces of Notch in T-helper-cell differentiation. Nat Rev Immunol. 2009;9:116–124. [DOI] [PubMed] [Google Scholar]
- 36.Minter LM, Turley DM, Das P, et al. Inhibitors of gamma-secretase block in vivo and in vitro T helper type 1 polarization by preventing Notch upregulation of Tbx21. Nat Immunol. 2005;6:680–688. [PubMed] [Google Scholar]
- 37.Segal BM, Dwyer BK, Shevach EM. An interleukin (IL)-10/IL-12 immunoregulatory circuit controls susceptibility to autoimmune disease. J Exp Med. 1998;187:537–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Cua DJ, Sherlock J, Chen Y, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. 2003;421:744–748. [DOI] [PubMed] [Google Scholar]
- 39.Langrish CL, Chen Y, Blumenschein WM, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201:233–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Neurath MF, Weigmann B, Finotto S, et al. The transcription factor T-bet regulates mucosal T cell activation in experimental colitis and Crohn’s disease. J Exp Med. 2002;195:1129–1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Fuss IJ, Neurath M, Boirivant M, et al. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn’s disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J Immunol. 1996;157:1261–1270. [PubMed] [Google Scholar]
- 42.Matsuoka K, Inoue N, Sato T, et al. T-bet upregulation and subsequent interleukin 12 stimulation are essential for induction of Th1 mediated immunopathology in Crohn’s disease. Gut. 2004;53:1303–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Plevy SE, Landers CJ, Prehn J, et al. A role for TNF-alpha and mucosal T helper-1 cytokines in the pathogenesis of Crohn’s disease. J Immunol. 1997;159:6276–6282. [PubMed] [Google Scholar]
- 44.Trembleau S, Penna G, Bosi E, Mortara A, Gately MK, Adorini L. Interleukin 12 administration induces T helper type 1 cells and accelerates autoimmune diabetes in NOD mice. J Exp Med. 1995;181:817–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang B, Andre I, Gonzalez A, et al. Interferon-gamma impacts at multiple points during the progression of autoimmune diabetes. Proc Natl Acad Sci U S A. 1997;94:13844–13849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol. 2008;8:183–192. [DOI] [PubMed] [Google Scholar]
- 47.Anderson CF, Oukka M, Kuchroo VJ, Sacks D. CD4(+)CD25(−)Foxp3 (−) Th1 cells are the source of IL-10-mediated immune suppression in chronic cutaneous leishmaniasis. J Exp Med. 2007;204:285–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Jankovic D, Kullberg MC, Feng CG, et al. Conventional T-bet(+)Foxp3(−) Th1 cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection. J Exp Med. 2007;204:273–283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Yu F, Sharma S, Jankovic D, et al. The transcription factor Bhlhe40 is a switch of inflammatory versus antiinflammatory Th1 cell fate determination. J Exp Med. 2018;215:1813–1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Paul WE, Zhu J. How are T(H)2-type immune responses initiated and amplified? Nat Rev Immunol. 2010;10:225–235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Walker JA, McKenzie ANJ. TH2 cell development and function. Nat Rev Immunol. 2018;18:121–133. [DOI] [PubMed] [Google Scholar]
- 52.Anthony RM, Rutitzky LI, Urban JF Jr, Stadecker MJ, Gause WC. Protective immune mechanisms in helminth infection. Nat Rev Immunol. 2007;7:975–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gause WC, Wynn TA, Allen JE. Type 2 immunity and wound healing: evolutionary refinement of adaptive immunity by helminths. Nat Rev Immunol. 2013;13:607–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lohoff M, Mittrucker HW, Prechtl S, et al. Dysregulated T helper cell differentiation in the absence of interferon regulatory factor 4. Proc Natl Acad Sci U S A. 2002;99:11808–11812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Rengarajan J, Mowen KA, McBride KD, Smith ED, Singh H, Glimcher LH. Interferon regulatory factor 4 (IRF4) interacts with NFATc2 to modulate interleukin 4 gene expression. J Exp Med. 2002;195: 1003–1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Li B, Tournier C, Davis RJ, Flavell RA. Regulation of IL-4 expression by the transcription factor JunB during T helper cell differentiation. EMBO J. 1999;18:420–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hartenstein B, Teurich S, Hess J, Schenkel J, Schorpp-Kistner M, Angel P. Th2 cell-specific cytokine expression and allergeninduced airway inflammation depend on JunB. EMBO J. 2002;21: 6321–6329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kim JI, Ho IC, Grusby MJ, Glimcher LH. The transcription factor c-Maf controls the production of interleukin-4 but not other Th2 cytokines. Immunity. 1999;10:745–751. [DOI] [PubMed] [Google Scholar]
- 59.Kagami S, Nakajima H, Suto A, et al. Stat5a regulates T helper cell differentiation by several distinct mechanisms. Blood. 2001;97: 2358–2365. [DOI] [PubMed] [Google Scholar]
- 60.Wei L, Vahedi G, Sun HW, et al. Discrete roles of STAT4 and STAT6 transcription factors in tuning epigenetic modifications and transcription during T helper cell differentiation. Immunity. 2010;32: 840–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kurata H, Lee HJ, O’Garra A, Arai N. Ectopic expression of activated Stat6 induces the expression of Th2-specific cytokines and transcription factors in developing Th1 cells. Immunity. 1999;11:677–688. [DOI] [PubMed] [Google Scholar]
- 62.Pai SY, Truitt ML, Ho IC. GATA-3 deficiency abrogates the development and maintenance of T helper type 2 cells. Proc Natl Acad Sci U S A. 2004;101:1993–1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhu J, Min B, Hu-Li J, et al. Conditional deletion of Gata3 shows its essential function in T(H)1-T(H)2 responses. Nat Immunol. 2004;5:1157–1165. [DOI] [PubMed] [Google Scholar]
- 64.Agarwal S, Avni O, Rao A. Cell-type-restricted binding of the transcription factor NFAT to a distal IL-4 enhancer in vivo. Immunity. 2000;12:643–652. [DOI] [PubMed] [Google Scholar]
- 65.Ouyang W, Lohning M, Gao Z, et al. Stat6-independent GATA-3 autoactivation directs IL-4-independent Th2 development and commitment. Immunity. 2000;12:27–37. [DOI] [PubMed] [Google Scholar]
- 66.Yamashita M, Ukai-Tadenuma M, Kimura M, et al. Identification of a conserved GATA3 response element upstream proximal from the interleukin-13 gene locus. J Biol Chem. 2002;277:42399–42408. [DOI] [PubMed] [Google Scholar]
- 67.Zhang DH, Cohn L, Ray P, Bottomly K, Ray A. Transcription factor GATA-3 is differentially expressed in murine Th1 and Th2 cells and controls Th2-specific expression of the interleukin-5 gene. J Biol Chem. 1997;272:21597–21603. [DOI] [PubMed] [Google Scholar]
- 68.Usui T, Nishikomori R, Kitani A, Strober W. GATA-3 suppresses Th1 development by downregulation of Stat4 and not through effects on IL-12Rbeta2 chain or T-bet. Immunity. 2003;18:415–428. [DOI] [PubMed] [Google Scholar]
- 69.Yagi R, Junttila IS, Wei G, et al. The transcription factor GATA3 actively represses RUNX3 protein-regulated production of interferon-gamma. Immunity. 2010;32:507–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yang XO, Angkasekwinai P, Zhu J, et al. Requirement for the basic helix-loop-helix transcription factor Dec2 in initial TH2 lineage commitment. Nat Immunol. 2009;10:1260–1266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yu Q, Sharma A, Oh SY, et al. T cell factor 1 initiates the T helper type 2 fate by inducing the transcription factor GATA-3 and repressing interferon-gamma. Nat Immunol. 2009;10:992–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Quirion MR, Gregory GD, Umetsu SE, Winandy S, Brown MA. Cutting edge: ikaros is a regulator of Th2 cell differentiation. J Immunol. 2009;182:741–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Thomas RM, Chen C, Chunder N, et al. Ikaros silences T-bet expression and interferon-gamma production during T helper 2 differentiation. J Biol Chem. 2010;285:2545–2553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhu J, Davidson TS, Wei G, et al. Down-regulation of Gfi-1 expression by TGF-beta is important for differentiation of Th17 and CD103+ inducible regulatory T cells. J Exp Med. 2009;206:329–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zhu J, Jankovic D, Grinberg A, Guo L, Paul WE. Gfi-1 plays an important role in IL-2-mediated Th2 cell expansion. Proc Natl Acad Sci U S A. 2006;103:18214–18219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Grunewald SM, Teufel M, Erb K, et al. Upon prolonged allergen exposure IL-4 and IL-4Ralpha knockout mice produce specific IgE leading to anaphylaxis. Int Arch Allergy Immunol. 2001;125:322–328. [DOI] [PubMed] [Google Scholar]
- 77.Mohrs M, Holscher C, Brombacher F. Interleukin-4 receptor alphadeficient BALB/c mice show an unimpaired T helper 2 polarization in response to Leishmania major infection. Infect Immun. 2000;68:1773–1780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.van Panhuys N, Tang SC, Prout M, et al. In vivo studies fail to reveal a role for IL-4 or STAT6 signaling in Th2 lymphocyte differentiation. Proc Natl Acad Sci U S A. 2008;105:12423–12428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Jankovic D, Kullberg MC, Noben-Trauth N, Caspar P, Paul WE, Sher A. Single cell analysis reveals that IL-4 receptor/Stat6 signaling is not required for the in vivo or in vitro development of CD4+ lymphocytes with a Th2 cytokine profile. J Immunol. 2000;164:3047–3055. [DOI] [PubMed] [Google Scholar]
- 80.King SB, Knorn AM, Ohnmacht C, Voehringer D. Accumulation of effector CD4 T cells during type 2 immune responses is negatively regulated by Stat6. J Immunol. 2008;180:754–763. [DOI] [PubMed] [Google Scholar]
- 81.Amsen D, Blander JM, Lee GR, Tanigaki K, Honjo T, Flavell RA. Instruction of distinct CD4 T helper cell fates by different notch ligands on antigen-presenting cells. Cell. 2004;117:515–526. [DOI] [PubMed] [Google Scholar]
- 82.Amsen D, Antov A, Jankovic D, et al. Direct regulation of Gata3 expression determines the T helper differentiation potential of Notch. Immunity. 2007;27:89–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Tumes DJ, Papadopoulos M, Endo Y, Onodera A, Hirahara K, Nakayama T. Epigenetic regulation of T-helper cell differentiation, memory, and plasticity in allergic asthma. Immunol Rev. 2017;278: 8–19. [DOI] [PubMed] [Google Scholar]
- 84.Morimoto Y, Hirahara K, Kiuchi M, et al. Amphiregulin-producing pathogenic memory T Helper 2 cells instruct eosinophils to secrete osteopontin and facilitate airway fibrosis. Immunity. 2018;49:134–150 e6. [DOI] [PubMed] [Google Scholar]
- 85.Endo Y, Iwamura C, Kuwahara M, et al. Eomesodermin controls interleukin-5 production in memory T helper 2 cells through inhibition of activity of the transcription factor GATA3. Immunity.2011;35:733–745. [DOI] [PubMed] [Google Scholar]
- 86.Finotto S, Neurath MF, Glickman JN, et al. Development of spontaneous airway changes consistent with human asthma in mice lacking T-bet. Science. 2002;295:336–338. [DOI] [PubMed] [Google Scholar]
- 87.Finotto S, Hausding M, Doganci A, et al. Asthmatic changes in mice lacking T-bet are mediated by IL-13. Int Immunol. 2005;17:993–1007. [DOI] [PubMed] [Google Scholar]
- 88.Wang YH, Voo KS, Liu B, et al. A novel subset of CD4(+) T(H)2 memory/effector cells that produce inflammatory IL-17 cytokine and promote the exacerbation of chronic allergic asthma. J Exp Med. 2010;207:2479–2491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Irvin C, Zafar I, Good J, et al. Increased frequency of dual-positive TH2/TH17 cells in bronchoalveolar lavage fluid characterizes a population of patients with severe asthma. J Allergy Clin Immunol. 2014;134:1175–1186 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Hegazy AN, Peine M, Helmstetter C, et al. Interferons direct Th2 cell reprogramming to generate a stable GATA-3(+)T-bet(+) cell subset with combined Th2 and Th1 cell functions. Immunity. 2010;32: 116–128. [DOI] [PubMed] [Google Scholar]
- 91.Kaplan MH, Hufford MM, Olson MR. The development and in vivo function of T helper 9 cells. Nat Rev Immunol. 2015;15:295–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Licona-Limon P, Henao-Mejia J, Temann AU, et al. Th9 cells drive host immunity against gastrointestinal worm infection. Immunity. 2013;39:744–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Purwar R, Schlapbach C, Xiao S, et al. Robust tumor immunity to melanoma mediated by interleukin-9-producing T cells. Nat Med. 2012;18:1248–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Vegran F, Berger H, Boidot R, et al. The transcription factor IRF1 dictates the IL-21-dependent anticancer functions of TH9 cells. Nat Immunol. 2014;15:758–766. [DOI] [PubMed] [Google Scholar]
- 95.Park J, Li H, Zhang M, et al. Murine Th9 cells promote the survival of myeloid dendritic cells in cancer immunotherapy. Cancer Immunol Immunother. 2014;63:835–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Yao W, Tepper RS, Kaplan MH. Predisposition to the development of IL-9-secreting T cells in atopic infants. J Allergy Clin Immunol. 2011;128:1357–1360 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Jabeen R, Goswami R, Awe O, et al. Th9 cell development requires a BATF-regulated transcriptional network. J Clin Invest. 2013;123:4641–4653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Yao W, Zhang Y, Jabeen R, et al. Interleukin-9 is required for allergic airway inflammation mediated by the cytokine TSLP. Immunity. 2013;38:360–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gerlach K, Hwang Y, Nikolaev A, et al. TH9 cells that express the transcription factor PU.1 drive T cell-mediated colitis via IL-9 receptor signaling in intestinal epithelial cells. Nat Immunol. 2014;15: 676–686. [DOI] [PubMed] [Google Scholar]
- 100.Nalleweg N, Chiriac MT, Podstawa E, et al. IL-9 and its receptor are predominantly involved in the pathogenesis of UC. Gut. 2015;64:743–755. [DOI] [PubMed] [Google Scholar]
- 101.Liao W, Spolski R, Li P, et al. Opposing actions of IL-2 and IL-21 on Th9 differentiation correlate with their differential regulation of BCL6 expression. Proc Natl Acad Sci U S A. 2014;111:3508–3513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Schmitt E, Germann T, Goedert S, et al. IL-9 production of naive CD4+ T cells depends on IL-2, is synergistically enhanced by a combination of TGF-beta and IL-4, and is inhibited by IFN-gamma. J Immunol. 1994;153:3989–3996. [PubMed] [Google Scholar]
- 103.Goswami R, Jabeen R, Yagi R, et al. STAT6-dependent regulation of Th9 development. J Immunol. 2012;188:968–975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Dardalhon V, Awasthi A, Kwon H, et al. IL-4 inhibits TGF-beta induced Foxp3+ T cells and, together with TGF-beta, generates IL-9+ IL-10+ Foxp3(−) effector T cells. Nat Immunol. 2008;9:1347–1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Staudt V, Bothur E, Klein M, et al. Interferon-regulatory factor 4 is essential for the developmental program of T helper 9 cells. Immunity. 2010;33:192–202. [DOI] [PubMed] [Google Scholar]
- 106.Veldhoen M, Uyttenhove C, van Snick J, et al. Transforming growth factor-beta ‘reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nat Immunol. 2008;9:1341–1346. [DOI] [PubMed] [Google Scholar]
- 107.Chang HC, Sehra S, Goswami R, et al. The transcription factor PU.1 is required for the development of IL-9-producing T cells and allergic inflammation. Nat Immunol. 2010;11:527–534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Goswami R, Kaplan MH. Gcn5 is required for PU.1-dependent IL-9 induction in Th9 cells. J Immunol. 2012;189:3026–3033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Bi E, Ma X, Lu Y, et al. Foxo1 and Foxp1 play opposing roles in regulating the differentiation and antitumor activity of TH9 cells programmed by IL-7. Sci Signal. 2017;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Malik S, Sadhu S, Elesela S, et al. Transcription factor Foxo1 is essential for IL-9 induction in T helper cells. Nat Commun. 2017;8:815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Buttrick TS, Wang W, Yung C, et al. Foxo1 promotes Th9 cell differentiation and airway allergy. Sci Rep. 2018;8:818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Stockinger B, Omenetti S. The dichotomous nature of T helper 17 cells. Nat Rev Immunol. 2017;17:535–544. [DOI] [PubMed] [Google Scholar]
- 113.Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Annu Rev Immunol. 2009;27:485–517. [DOI] [PubMed] [Google Scholar]
- 114.Gaublomme JT, Yosef N, Lee Y, et al. Single-cell genomics unveils critical regulators of Th17 cell pathogenicity. Cell. 2015;163: 1400–1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Lee Y, Awasthi A, Yosef N, et al. Induction and molecular signature of pathogenic TH17 cells. Nat Immunol. 2012;13:991–999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–238. [DOI] [PubMed] [Google Scholar]
- 117.Chen W, Jin W, Hardegen N, et al. Conversion of peripheral CD4+CD25-naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 2003;198:1875–1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Mangan PR, Harrington LE, O’Quinn DB, et al. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. 2006;441:231–234. [DOI] [PubMed] [Google Scholar]
- 119.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;24:179–189. [DOI] [PubMed] [Google Scholar]
- 120.Gutcher I, Donkor MK, Ma Q, Rudensky AY, Flavell RA, Li MO. Autocrine transforming growth factor-beta1 promotes in vivo Th17 cell differentiation. Immunity. 2011;34:396–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.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] [PMC free article] [PubMed] [Google Scholar]
- 122.Zhou L, Lopes JE, Chong MM, et al. TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature. 2008;453:236–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Weaver CT, Hatton RD. Interplay between the TH17 and TReg cell lineages: a (co-)evolutionary perspective. Nat Rev Immunol. 2009;9:883–889. [DOI] [PubMed] [Google Scholar]
- 124.Korn T, Bettelli E, Gao W, et al. IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells. Nature. 2007;448:484–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Ciofani M, Madar A, Galan C, et al. A validated regulatory network for Th17 cell specification. Cell. 2012;151:289–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Yosef N, Shalek AK, Gaublomme JT, et al. Dynamic regulatory network controlling TH17 cell differentiation. Nature. 2013;496: 461–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Kurebayashi Y, Nagai S, Ikejiri A, Koyasu S. Recent advances in understanding the molecular mechanisms of the development and function of Th17 cells. Genes Cells. 2013;18:247–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ghoreschi K, Laurence A, Yang XP, et al. Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling. Nature. 2010;467:967–971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.McGeachy MJ, Bak-Jensen KS, Chen Y, et al. TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol. 2007;8: 1390–1397. [DOI] [PubMed] [Google Scholar]
- 130.McGeachy MJ, Chen Y, Tato CM, et al. The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17 producing effector T helper cells in vivo. Nat Immunol. 2009;10: 314–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Meyer Z, Horste G, Wu C, et al. RBPJ controls development of pathogenic Th17 cells by regulating IL-23 receptor expression. Cell Rep. 2016;16:392–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Lee YK, Turner H, Maynard CL, et al. Late developmental plasticity in the T helper 17 lineage. Immunity. 2009;30:92–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Ahern PP, Schiering C, Buonocore S, et al. Interleukin-23 drives intestinal inflammation through direct activity on T cells. Immunity. 2010;33:279–288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Bending D, De la Pena H, 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;119:565–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Harbour SN, Maynard CL, Zindl CL, Schoeb TR, Weaver CT. Th17 cells give rise to Th1 cells that are required for the pathogenesis of colitis. Proc Natl Acad Sci U S A. 2015;112:7061–7066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Hirota K, Duarte JH, Veldhoen M, et al. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat Immunol. 2011;12:255–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Nistala K, Adams S, Cambrook H, et al. Th17 plasticity in human autoimmune arthritis is driven by the inflammatory environment. Proc Natl Acad Sci U S A. 2010;107:14751–14756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Wang Y, Godec J, Ben-Aissa K, et al. The transcription factors T-bet and Runx are required for the ontogeny of pathogenic interferon-gamma-producing T helper 17 cells. Immunity. 2014;40: 355–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Glosson-Byers NL, Sehra S, Stritesky GL, et al. Th17 cells demonstrate stable cytokine production in a proallergic environment. J Immunol. 2014;193:2631–2640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Kudo M, Melton AC, Chen C, et al. IL-17A produced by alphabeta T cells drives airway hyper-responsiveness in mice and enhances mouse and human airway smooth muscle contraction. Nat Med. 2012;18:547–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.McKinley L, Alcorn JF, Peterson A, et al. TH17 cells mediate steroid resistant airway inflammation and airway hyperresponsiveness in mice. J Immunol. 2008;181:4089–4097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Zheng Y, Danilenko DM, Valdez P, et al. Interleukin-22, a T(H)17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature. 2007;445:648–651. [DOI] [PubMed] [Google Scholar]
- 143.Amadi-Obi A, Yu CR, Liu X, et al. TH17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1. Nat Med. 2007;13:711–718. [DOI] [PubMed] [Google Scholar]
- 144.Chen Y, Chauhan SK, Lee HS, Saban DR, Dana R. Chronic dry eye disease is principally mediated by effector memory Th17 cells. Mucosal Immunol. 2014;7:38–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.De Paiva CS, Chotikavanich S, Pangelinan SB, et al. IL-17 disrupts corneal barrier following desiccating stress. Mucosal Immunol. 2009;2:243–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Luger D, Silver PB, Tang J, et al. Either a Th17 or a Th1 effector response can drive autoimmunity: conditions of disease induction affect dominant effector category. J Exp Med. 2008;205:799–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Codarri L, Gyulveszi G, Tosevski V, et al. RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol. 2011;12:560–567. [DOI] [PubMed] [Google Scholar]
- 148.El-Behi M, Ciric B, Dai H, et al. The encephalitogenicity of T(H)17 cells is dependent on IL-1and IL-23-induced production of the cytokine GM-CSF. Nat Immunol. 2011;12:568–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Lin CC, Bradstreet TR, Schwarzkopf EA, et al. IL-1-induced Bhlhe40 identifies pathogenic T helper cells in a model of autoimmune neuroinflammation. J Exp Med. 2016;213:251–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Lin CC, Bradstreet TR, Schwarzkopf EA, et al. Bhlhe40 controls cytokine production by T cells and is essential for pathogenicity in autoimmune neuroinflammation. Nat Commun. 2014;5:3551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Ichiyama K, Gonzalez-Martin A, Kim BS, et al. The microRNA-183-96-182 cluster promotes T Helper 17 cell pathogenicity by negatively regulating transcription factor Foxo1 expression. Immunity. 2016;44:1284–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Gagliani N, Amezcua Vesely MC, Iseppon A, et al. Th17 cells transdifferentiate into regulatory T cells during resolution of inflammation. Nature. 2015;523:221–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Hirota K, Turner JE, Villa M, et al. Plasticity of Th17 cells in Peyer’s patches is responsible for the induction of T cell-dependent IgA responses. Nat Immunol. 2013;14:372–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.DuPage M, Bluestone JA. Harnessing the plasticity of CD4(+) T cells to treat immune-mediated disease. Nat Rev Immunol. 2016;16: 149–163. [DOI] [PubMed] [Google Scholar]
- 155.Ma CS, Deenick EK, Batten M, Tangye SG. The origins, function, and regulation of T follicular helper cells. J Exp Med. 2012;209: 1241–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Ueno H T follicular helper cells in human autoimmunity. Curr Opin Immunol. 2016;43:24–31. [DOI] [PubMed] [Google Scholar]
- 157.Crotty S Follicular helper CD4 T cells (TFH). Annu Rev Immunol. 2011;29:621–663. [DOI] [PubMed] [Google Scholar]
- 158.Qi H T follicular helper cells in space-time. Nat Rev Immunol. 2016;16:612–625. [DOI] [PubMed] [Google Scholar]
- 159.King C New insights into the differentiation and function of T follicular helper cells. Nat Rev Immunol. 2009;9:757–766. [DOI] [PubMed] [Google Scholar]
- 160.Fazilleau N, McHeyzer-Williams LJ, Rosen H, McHeyzer-Williams MG. The function of follicular helper T cells is regulated by the strength of T cell antigen receptor binding. Nat Immunol. 2009; 10:375–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Tubo NJ, Pagan AJ, Taylor JJ, et al. Single naive CD4+ T cells from a diverse repertoire produce different effector cell types during infection. Cell. 2013;153:785–796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Choi YS, Kageyama R, Eto D, et al. ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity. 2011;34: 932–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Nurieva RI, Chung Y, Hwang D, et al. Generation of T follicular helper cells is mediated by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. Immunity. 2008;29:138–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Bauquet AT, Jin H, Paterson AM, et al. The costimulatory molecule ICOS regulates the expression of c-Maf and IL-21 in the development of follicular T helper cells and TH-17 cells. Nat Immunol. 2009;10:167–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Eto D, Lao C, DiToro D, et al. IL-21 and IL-6 are critical for different aspects of B cell immunity and redundantly induce optimal follicular helper CD4 T cell (Tfh) differentiation. PLoS One. 2011; 6:e17739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Hardtke S, Ohl L, Forster R. Balanced expression of CXCR5 and CCR7 on follicular T helper cells determines their transient positioning to lymph node follicles and is essential for efficient B-cell help. Blood. 2005;106:1924–1931. [DOI] [PubMed] [Google Scholar]
- 167.Johnston RJ, Poholek AC, DiToro D, et al. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science. 2009;325:1006–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Nurieva RI, Chung Y, Martinez GJ, et al. Bcl6 mediates the development of T follicular helper cells. Science. 2009;325: 1001–1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Yu D, Rao S, Tsai LM, et al. The transcriptional repressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity. 2009;31: 457–468. [DOI] [PubMed] [Google Scholar]
- 170.Betz BC, Jordan-Williams KL, Wang C, et al. Batf coordinates multiple aspects of B and T cell function required for normal antibody responses. J Exp Med. 2010;207:933–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Ise W, Kohyama M, Schraml BU, et al. The transcription factor BATF controls the global regulators of class-switch recombination in both B cells and T cells. Nat Immunol. 2011;12:536–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Shekhar S, Yang X. The darker side of follicular helper T cells: from autoimmunity to immunodeficiency. Cell Mol Immunol. 2012;9: 380–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Lee SK, Silva DG, Martin JL, et al. Interferon-gamma excess leads to pathogenic accumulation of follicular helper T cells and germinal centers. Immunity. 2012;37:880–892. [DOI] [PubMed] [Google Scholar]
- 174.Maehara T, Moriyama M, Hayashida JN, et al. Selective localization of T helper subsets in labial salivary glands from primary Sjogren’s syndrome patients. Clin Exp Immunol. 2012;169:89–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Guo J, Zhao C, Wu F, et al. T follicular helper-like cells are involved in the pathogenesis of experimental autoimmune encephalomyelitis. Front Immunol. 2018;9:944. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Rao DA, Gurish MF, Marshall JL, et al. Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature. 2017;542:110–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Ballesteros-Tato A, Leon B, Graf BA, et al. Interleukin-2 inhibits germinal center formation by limiting T follicular helper cell differentiation. Immunity. 2012;36:847–856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Johnston RJ, Choi YS, Diamond JA, Yang JA, Crotty S. STAT5 is a potent negative regulator of TFH cell differentiation. J Exp Med. 2012;209:243–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol. 2007;8:191–197. [DOI] [PubMed] [Google Scholar]
- 180.Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, 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;204:1757–1764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Sun CM, Hall JA, Blank RB, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204:1775–1785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.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;22:329–341. [DOI] [PubMed] [Google Scholar]
- 183.Gavin MA, Rasmussen JP, Fontenot JD, et al. Foxp3-dependent programme of regulatory T-cell differentiation. Nature. 2007;445: 771–775. [DOI] [PubMed] [Google Scholar]
- 184.Lin W, Haribhai D, Relland LM, et al. Regulatory T cell development in the absence of functional Foxp3. Nat Immunol. 2007;8:359–368. [DOI] [PubMed] [Google Scholar]
- 185.Williams LM, Rudensky AY. Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires continued expression of Foxp3. Nat Immunol. 2007;8:277–284. [DOI] [PubMed] [Google Scholar]
- 186.Lee W, Lee GR. Transcriptional regulation and development of regulatory T cells. Exp Mol Med. 2018;50:e456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Samstein RM, Arvey A, Josefowicz SZ, et al. Foxp3 exploits a preexistent enhancer landscape for regulatory T cell lineage specification. Cell. 2012;151:153–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Wu Y, Borde M, Heissmeyer V, et al. FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell. 2006;126: 375–387. [DOI] [PubMed] [Google Scholar]
- 189.Ono M, Yaguchi H, Ohkura N, et al. Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1. Nature. 2007;446: 685–689. [DOI] [PubMed] [Google Scholar]
- 190.Pan F, Yu H, Dang EV, et al. Eos mediates Foxp3-dependent gene silencing in CD4+ regulatory T cells. Science. 2009;325: 1142–1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Lio CW, Hsieh CS. Becoming self-aware: the thymic education of regulatory T cells. Curr Opin Immunol. 2011;23:213–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Kerdiles YM, Stone EL, Beisner DR, et al. Foxo transcription factors control regulatory T cell development and function. Immunity. 2010;33:890–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Ouyang W, Liao W, Luo CT, et al. Novel Foxo1-dependent transcriptional programs control T(reg) cell function. Nature. 2012;491: 554–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Takimoto T, Wakabayashi Y, Sekiya T, et al. Smad2 and Smad3 are redundantly essential for the TGF-beta-mediated regulation of regulatory T plasticity and Th1 development. J Immunol. 2010;185: 842–855. [DOI] [PubMed] [Google Scholar]
- 195.Wu C, Chen Z, Dardalhon V, et al. The transcription factor musculin promotes the unidirectional development of peripheral Treg cells by suppressing the TH2 transcriptional program. Nat Immunol. 2017;18:344–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Roychoudhuri R, Hirahara K, Mousavi K, et al. BACH2 represses effector programs to stabilize T(reg)-mediated immune homeostasis. Nature. 2013;498:506–510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Yu F, Sharma S, Edwards J, Feigenbaum L, Zhu J. Dynamic expression of transcription factors T-bet and GATA-3 by regulatory T cells maintains immunotolerance. Nat Immunol. 2015;16: 197–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Koch MA, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat Immunol. 2009;10:595–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Tan TG, Mathis D, Benoist C. Singular role for T-BET+CXCR3+ regulatory T cells in protection from autoimmune diabetes. Proc Natl Acad Sci U S A. 2016;113:14103–14108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Zheng Y, Chaudhry A, Kas A, et al. Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature. 2009;458:351–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Chaudhry A, Rudra D, Treuting P, et al. CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science. 2009;326:986–991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Ohnmacht C, Park JH, Cording S, et al. Mucosal immunology. The microbiota regulates type 2 immunity through RORgammat(+) T cells. Science. 2015;349:989–993. [DOI] [PubMed] [Google Scholar]
- 203.Sefik E, Geva-Zatorsky N, Oh S, et al. Mucosal immunology. Individual intestinal symbionts induce a distinct population of RORgamma(+) regulatory T cells. Science. 2015;349:993–997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Xu M, Pokrovskii M, Ding Y, et al. c-MAF-dependent regulatory T cells mediate immunological tolerance to a gut pathobiont. Nature. 2018;554:373–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Campbell DJ, Koch MA. Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat Rev Immunol. 2011;11:119–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Zhou X, Bailey-Bucktrout SL, Jeker LT, et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol. 2009;10:1000–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Noval Rivas M, Burton OT, Wise P, et al. Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity. 2015;42:512–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Komatsu N, Okamoto K, Sawa S, et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat Med. 2014;20:62–68. [DOI] [PubMed] [Google Scholar]
- 209.Miyao T, Floess S, Setoguchi R, et al. Plasticity of Foxp3(+) T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells. Immunity. 2012;36: 262–275. [DOI] [PubMed] [Google Scholar]
- 210.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;178:280–290. [DOI] [PubMed] [Google Scholar]
- 211.Oldenhove G, Bouladoux N, Wohlfert EA, et al. Decrease of Foxp3+ Treg cell number and acquisition of effector cell phenotype during lethal infection. Immunity. 2009;31:772–786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Tang Q, Adams JY, Penaranda C, et al. Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity. 2008;28:687–697. [DOI] [PMC free article] [PubMed] [Google Scholar]


