Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Aug 1.
Published in final edited form as: Cytokine. 2015 Jul 10;76(1):13–24. doi: 10.1016/j.cyto.2015.07.005

Origin and functions of pro-inflammatory cytokine producing Foxp3+ regulatory T cells

Pushpa Pandiyan a,*, Jinfang Zhu b
PMCID: PMC4969074  NIHMSID: NIHMS741772  PMID: 26165923

Abstract

CD4+CD25+Foxp3+ regulatory cells (Tregs) are a special lineage of cells central in the maintenance of immune homeostasis, and are targeted for human immunotherapy. They are conventionally associated with the production of classical anti-inflammatory cytokines such as IL-10, TGF-β and IL-35, consistent to their anti-inflammatory functions. However, emerging evidence show that they also express effector cytokines such as IFN-γ and IL-17A under inflammatory conditions. While some studies reveal that these pro-inflammatory cytokine producing Foxp3+ regulatory cells retain their suppressive ability, others believe that these cells are dys-regulated and are associated with perpetuation of immunopathology. Therefore the development of these cells may challenge the efficacy of human Treg therapy. Mechanistically, toll-like receptor (TLR) ligands and the pro-inflammatory cytokine milieu have been shown to play important roles in the induction of effector cytokines in Tregs. Here we review the mechanisms of development and the possible functions of pro-inflammatory cytokine producing Foxp3+ Tregs.

Keywords: Treg, Th17, IL-17, Foxp3, Suppression, Inflammation, Cytokine, Reprogramming

1. Introduction

As CD4+Foxp3+ Tregs are central to immune tolerance [17], the potential of these cells for the treatment of T-cell-mediated diseases in humans has gained momentum in recent years. Therefore it is essential to understand precisely how they respond under steady state conditions and inflammatory conditions. Although Tregs are conventionally known to be non-producers of pro-inflammatory cytokines, emerging evidence shows that Foxp3+ Tregs are reprogrammed into T helper (Th) like cells producing pro-inflammatory cytokines in mice as well as in various human diseases [817] (Fig. 1). Zhou et al. first described the potential pathogenicity of reprogrammed Tregs that lose Foxp3 in murine model of Type 1 Diabetes [18]. Besides reprogrammed Tregs that lose Foxp3, T helper (Th) like Tregs that still express Foxp3 have also been reported to produce inflammatory cytokines. Th1 like Tregs co-express Foxp3, T-cell-specific T-box transcription factor (T-bet) and CXCR3 [19]. Th17 like Tregs express Foxp3, retinoic acid-related orphan receptor (ROR)-γt and IL-17A [11]. Such effector like Foxp3+ `Treg cells producing pro-inflammatory cytokines are readily found at very low levels in tissues, but their presence is exaggerated during inflammation [11,2023]. Th effector like Tregs share phenotypic and functional features with conventional Th cells, such as the expression of tissue homing receptors such as CCR4, CXCR5, CCR6 and CXCR3 [24,25]. These Tregs, with a possibility of losing Foxp3 and transforming into pathogenic cells under inflammatory conditions in vivo, temper the interests of using Tregs as cellular therapeutics, at least in the context of certain diseases. Since Treg cells are enriched for autoreactive TCR specificities, the possibility of them converting into pathogenic effector T cells could be a critical threat to the host in the context of autoimmunity [18]. While Th17 like Tregs are shown by some studies to retain suppressive function [8,11], some others suggest that they may contribute to immunopathology [20,26]. Human studies on whether these pro-inflammatory cytokine producing Tregs lose their suppressive functions and cause autoimmunity are only beginning to emerge. Although these cells are prominently seen during inflammatory conditions, under steady state conditions, the development of Th effector like Tregs must be tightly regulated on several levels. Therefore studies investigating (1) how Tregs are dys-regulated or reprogrammed, (2) what contributes to their pathogenic cytokine production and, (3) if these reprogrammed cells contribute to, or control immunopathology are essential, so that we can gain a complete understanding of this process in order to account for and regulate it when considering employing Tregs as treatment. In this review, we will focus on the recent studies that provide varying answers, and unresolved questions about the development and possible functions of Foxp3+ cells co-expressing inflammatory cytokines [11,27]. Especially, we will focus on the markers and mechanisms of development of these cells in the context of various diseases.

Fig. 1.

Fig. 1

Origin and functions of pro-inflammatory cytokine producing Tregs in various diseases.

2. Conventional view on Tregs

CD4+CD25+Foxp3+ Tregs that constitute about 5–10% of peripheral CD4 T cells are endowed with potent suppressive activity [28]. These cells are identified by markers that include CTLA-4, GITR, Neuropilin (Nrp)-1, besides CD25 and Foxp3. In humans, Tregs are identified also by CD127low expression. The use of Helios as a marker for natural Tregs is controversial, especially in humans [29]. Tregs are considered “anergic” because they proliferate poorly in vitro, when stimulated through T cell receptor in the absence of γ-chain cytokine, and do not express the CD4 cell signature cytokine IL-2 [30,31]. Their anergic state is attributed to the expression of Foxp3 itself, which is a repressive forkhead or winged-helix family transcription factor. On the one hand, Foxp3 directly binds to the regulatory elements of IL-2 and IFN-γ genes and induces active deacetylation of histone H3, thereby inhibiting chromatin remodeling and controlling gene transcription [3235]. On the other, Foxp3 binds to GITR, CD25, and CTLA-4 genes increases histone acetylation and contributes to increased expression of these proteins in Tregs. Foxp3 also interacts and/or collaborates with dozens of other transcription factors, including NFAT, Runx1, Eos and Blimp1 in promoting classical Treg functions, as well as repressing the transcription of Th effector genes [36]. Thus Foxp3 is the master regulator for Treg generation and their suppressive functions. Mutations of the FOXP3 gene in humans result in Treg deficiencies, and are responsible for immunodysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome [37,38]. In mice, disruption of the Foxp3 gene, including Scurfy mutation, also results in the loss of Tregs leading to fatal systemic autoimmune disease [39]. Similar symptoms of autoimmunity both in mice and humans highlight the parallelism between mice and humans in terms of functional importance of Tregs in suppressing severe autoimmune reactions [38].

Whereas Foxp3 is essential for Treg cell development and function, most recent studies show that some Treg cell signature genes can be expressed in the absence of functional Foxp3 [4042]. Besides Foxp3, Foxo family members are also involved in classical Treg induction by binding to and remodeling many Foxp3-binding sites even before Foxp3 actually binds to these sites [43,44]. Thus Foxp3 by largely exploiting the preformed enhancer network, defines the Treg landscape and functionality. Moreover, it was recently shown the demethylation profile (nTreg-Me) of nTreg signature genes such as Foxp3, Ctla-4, Ikzf4 (encoding Eos) and Tnfrsf18 (encoding GITR), and not the Foxp3 expression alone, is a crucial component of Treg identity. As this hypomethylation signature is independent of Foxp3, it is clear that though Foxp3 is required, it is not sufficient for the establishment of a robust and stable Treg cell program [45]. Thus, although Foxp3 is the most reliable marker for Tregs suppression to date, hypomethylation status of Treg signature genes should also be taken into consideration in studies that identify changes in number and frequency of Tregs in vivo.

2.1. Natural Foxp3+ cells vs Induced Foxp3+ cells

Foxp3+ Treg cells include both natural or thymus-derived Treg (nTreg or tTreg) cells and peripherally induced Treg (pTreg) cells [4649]. nTreg cells develop in the thymus and constitute the majority of the peripheral Treg cell pool in naïve mice and in human cord blood. In contrast, pTreg cells arise predominantly at sites of inflammation and environmental interfaces, such as the gut. They can also be generated in vitro (induced Treg (iTreg)) after T-cell-receptor (TCR) stimulation of conventional CD4+ T cells in the presence of transforming growth factor-β (TGFβ) and interleukin-2 (IL-2). Both Tregs require Foxp3 for their development and function. In terms of Foxp3 stability and suppressive functions, nTregs constitute a relatively more stable population than pTregs [5052].

2.2. Treg’s suppressive functions and their anti-inflammatory cytokine production

Treg cells exert their immunoregulatory functions through various mechanisms requiring CTLA-4, direct killing of antigen presenting cells or T cells, consumption of IL-2, and the production of immunosuppressive cytokines such as IL-10, transforming growth factor (TGF)-β, IL-35 and galectin-1 [53]. Although a myriad of suppressive mechanisms have been proposed for Tregs, two basic modes of suppression, (1) deprivation mode under non-inflammatory and steady state conditions, and (2) active suppression mode during inflammatory reactions, have been established [1,54]. In the context of non-inflammatory conditions or at minimal levels of IL-2 and other inflammatory cytokines, natural Treg cells through CD25 and CTLA-4, deprive IL-2 and CD28 signals from antigen-reactive T cells resulting in blockade of progression of naïve T cells into effector states, and apoptosis of the differentiating T effectors. In highly inflammatory environments, for example, during a microbial infection, these deprivation mechanisms are transiently abrogated allowing T cells to respond to antigen. With excess of IL-2 and other cytokines in the milieu, Tregs also get activated acquire an armor of suppressive arsenal to kill or inactivate effector T cells and antigen-presenting cells through (1) granzyme/perforin pathways [55] and CD39 mediated ATP hydrolysis (2) IL-10 secretion, and (3) infectious tolerance mediated by TGF-β and IL-35. These mechanisms together constitute dampening of excessive immune responses. Disruption of one of these mechanisms alone cannot abrogate suppressive capacity of Tregs, as shown in earlier studies [5659]. Currently there seems to be a consensus in the literature that Treg mechanisms differ depending on the disease setting, inflammatory status of the local environment, and their anatomical localization [1,2,60]. Thus it is well known that anti-inflammatory cytokines such as IL-10, TGF-and IL-35 are produced by Treg cells, and are critical for some of their suppressive functions. However, Treg’s ability to undergo reprogramming into helper-like cells and produce inflammatory cytokines without the loss of Foxp3 was discovered only recently.

3. Pro-inflammatory cytokine production in Foxp3+ Tregs is associated with human diseases

3.1. Psoriasis

Psoriasis is an autoimmune chronic inflammatory skin disease that is associated with hyperactive IL-23 signaling and Th17 effector cytokines [61]. In this disease Tregs are known to be dysfunctional and lacking suppressive capacities [23]. Although IL-17A producing Treg cells are readily found in small numbers in the tissues such as tonsils and skin, the association of Th17-like Tregs with an human inflammatory condition was first identified in psoriasis patients [11,20,22,62]. The qualitative defect in Tregs is believed to be an underlying mechanism for unrestrained pathogenic effector T cell proliferation resulting in skin inflammation [20,62]. In human psoriatic skin the Foxp3+ cells have a memory phenotype expressing CD45RO, showing high capacity to proliferate and produce IL-17A [20,22]. However, they are bona fide Tregs expressing Foxp3 and CD25 and low levels of CD127, with no expression of IL-2 and IFN-γ [20,26]. TCRβ sequences of these Tregs show a very little overlap with those of effector T cells from the skin, suggesting that IL-17A+Foxp3+cells do not arise from T effector cells transiently expressing Foxp3 [22]. It is believed that these dys-regulated cells may represent Foxp3+Tregs that pass through a transient Foxp3+IL-17A+ stage and may progressively lose Foxp3 expression to become Ex-Tregs [18,20]. Examining TCR repertoires and nTreg-Me signature of specifically the Foxp3+IL-17A+ cells in psoriasis human tissue samples will offer more information about their origin and functions.

3.2. GVHD

Graft-versus-host disease (GvHD) is a major complication of allogeneic hematopoietic stem cell transplantation in which the transplanted immune system attacks recipient tissues as non-self. Acute GvHD is predominantly related to Th1 responses, where migration of T-bet+ and CXCR3+ cytotoxic effectors direct and type I interferon-mediated pathogenic processes in mucosa [63,64]. Th17 effector cells are also implicated in pathogenesis and Tregs are well established to have protective roles. While numerical deficiency in mucosal FOXP3+ Tregs correlates with acute and chronic GVHD [65], increase in FOXP3+ regulatory T cells in GVHD skin biopsies is associated with lesser severity of the disease and better treatment response [66]. In murine models of bone marrow transplantation, direct Treg infusion as well as agents that induce Tregs are shown to inhibit activation and proliferation of alloreactive donor T cells, thereby reducing GVHD [6,6769]. However recent reports show that activated Treg cells that migrate to inflamed sites in the skin tissues of cutaneous GVHD have an effector/memory cell phenotype. They are Th1-like Tregs expressing ICOS, CD39 and CXCR3, and also high levels of Th1 transcription factor T-bet. Although whether these Tregs express IFN-γ and contribute to exacerbation of the disease was not examined, it is speculated that T-bet expression in Tregs found in GvHD is not associated with the loss of suppression. Supporting this tenet, T-bet+ Tregs producing IFN-γ in fact have been shown to be required for regulatory T cell homeostasis and function during type 1 inflammation [70,71].

3.3. Inflammatory bowel disease (IBD)

Lack of intestinal tolerance to commensals and other innocuous antigens can lead to inflammatory bowel diseases (IBD) such as Crohn’s disease and ulcerative colitis. Both nTreg cells and pTreg cells are critical for commensal tolerance in the intestine [7274]. A number of studies have shown protective roles of Tregs in IBD. It is also well established that Th17 like Treg cells are present at elevated levels in the intestinal mucosa and in circulation of IBD patients, compared to healthy controls [10,7577]. Although one study showed that these cells are defective in their suppressive functions [77], other studies suggest that Th17 like Treg cells from CD tissues and colitic samples are immunosuppressive. These cells suppress T-cell proliferation in vitro, supporting the interpretation that these are not reprogrammed to lose their suppressive capacity [8,75]. Similarly, in the mouse model, although a fraction of Tregs produce IL-17A under Th17 conditions in vitro, when adoptively transferred into immunodeficient mice, they still modulate IBD and weight loss during Th17 IBD inflammation in vivo [78,79]. This is observed despite some of these Tregs may have additionally lost Foxp3 upon transfer into Rag−/− mice. Moreover, Th1 like Foxp3+ Treg cells that produce IFN-γ have also been reported to retain their regulatory functions and inhibit colitis in mice [80]. Taken together, these data demonstrate that inflammatory conditions in IBD can induce IFN-γ or IL-17A production in a fraction of Foxp3+ Tregs in vivo, but do not affect their immunomodulatory functions.

3.4. Periodontitis

The progression and severity of the periodontal disease are strongly governed by an imbalance between oral commensal bacteria and host immunity. T-cells, especially CD4+ T helper cells, play an essential role in aberrant responses and local inflammation. Increase in Th17 cells along with the decrease in Tregs, i.e., Th17)/Treg imbalance, has been suggested recently to be critical in the development of periodontitis. Okui et al. analyzed the association between functional plasticity in Tregs and the pathogenesis of periodontitis [81]. Their study revealed the presence of Th17 like Treg cells co-expressing Foxp3 and IL-17A in periodontitis lesions as well as in gingival tissue T-cell lines from patients. This finding indicates that Treg reprogramming may be another feature of periodontitis lesions. However it remains to be seen whether these IL-17A producing Treg cells contribute to inflammatory responses against periodontopathic bacteria.

3.5. Arthritis

Th17 cells are the dominant initiators of inflammation in several murine models of autoimmune arthritis. In humans, while Th17 cells play a pathological role in juvenile idiopathic arthritis [82,83] ankylosing, spondylitis and psoriatic arthritis [84], Th1/Th17 cells with intermediate phenotype are pathogenic in rheumatoid arthritis (RA) [82,85]. RA patients have been reported to exhibit a higher frequency of Th17 like Tregs compared to healthy subjects [86]. These Tregs also produce IFN-γ, IL-2, ROR-γt and Tbet, along with IL-17A [87,88]. However, the ability of these cells to suppress proliferation, production of IFN-γ and IL-2 in effector T cells was comparable to those of conventional Treg subpopulation from healthy controls, suggesting that these Tregs are not reprogrammed to lose their suppressive capacities.

3.6. Multiple Sclerosis

Multiple Sclerosis (MS) is characterized by the damage in myelin sheaths surrounding axons in the brain and spinal cord. Although myelin-antigen reactive Th1 cells were previously thought to drive MS, it now appears that pathogenic Th17 cells play an important role in disease pathogenesis [89]. Murine models of experimental autoimmune encephalomyelitis (EAE), show that the factors including IL-23 [90], GMCSF [91], a high-salt diet [92] and microbiota [93], all have impact on the development and severity of Th17 mediated EAE. A higher frequency of Th1-like Tregs secreting IFN-γ has been reported in subjects with relapsing remitting multiple sclerosis (RRMS), compared to healthy control individuals [21]. Analysis of the Foxp3+ Treg cell-specific demethylated region (TSDR) in isolated IFN-γ+ Foxp3+ (Th1-like Tregs) and IFN-γ Foxp3+ (conventional Tregs) in multiple sclerosis patients reveals that Th1-like Treg cells possess a similar pattern of demethylation in the TSDR region to that of conventional Tregs from patients and healthy controls. These studies suggest that Th1-like Tregs may have arisen from conventional Treg cells and not from effector cells expressing Foxp3 transiently. Also, while there is a direct correlation between Th1-like Tregs and pathology, an inverse correlation has been reported for between Th17-like Tregs and inflammation. Moreover, Th1 like Treg cells show reduced suppressive activity in vitro, which can partially be reversed by IFN-γ-specific antibodies or by removal of IL-12. Consistent to this, in subjects undergoing IFN-β immunomodulatory treatment, lower frequency of Th1 like Treg cells correlates with reduced inflammation. These data point to a general functional defect in the suppressive activity of Th1-like Tregs that might contribute to inflammation instead of regulating Th1 inflammation in MS. This is consistent to experimental autoimmune encephalomyelitis (EAE) mouse model study that demonstrated that T-bet expression in Tregs is not required for the resolution of central nervous system (CNS) inflammation [94]. However the functions of Th17 like Treg cells, whose frequency increases in IFN-β treated patients remains to be investigated in MS patients.

3.6.1. Type 1 diabetes

Autoimmune type 1diabetes (T1D) is a T cell-mediated autoimmune disease characterized by the destruction and dysfunction of pancreatic β cells. Numerous studies have demonstrated the beneficial role of Tregs in controlling T1D. In patients with type 1diabetes, the frequency of Tregs is comparable to healthy controls in peripheral blood. However, these patients reveal an increase in Th1-effector-like Tregs enriched in CD45RO+ memory cells. These FOXP3+IFN-γ+ Tregs are significantly increased in peripheral blood compared to the healthy controls [95]. Because Ex-Tregs that lose Foxp3 can be pathogenic in murine model of T1D [18], careful epigenetic analyses were performed in these human cells. These data showed that FOXP3+IFN-γ+ cells are predominately methylated at the TSDR, in contrast to conventional nTregs, suggesting a potential instability and loss of Foxp3 expression. However, consistent with nTreg anergic properties, IFN-γ+ Tregs do not produce IL-2. Most importantly, these Th1-effector-like Tregs retain their suppressive function, suggesting that these reprogrammed Tregs may not contribute to T1D pathogenesis.

3.7. Cancer

Treg infiltration in tumors, and the resulting suppression of anti-tumor immunity is considered to be one of the mechanisms of immune evasion in several tumors [96,97]. Human tumor-infiltrating lymphocytes (TIL) isolated from tumor tissue samples of melanoma, ovarian, breast and colon cancers revealed that a substantial fraction of these cells can develop into FOXP3+ cells expressing pro-inflammatory cytokines [98]. This population of cells, arising after rounds of in vitro expansion of TIL, can co-produce IFN-γ and IL-17A, [99] and are shown to develop from effector Th17 cells. However, the expanded population exhibits stable expression and demethylation of FOXP3 and retain potent suppressive function in vitro. These data are consistent to another study, which showed a strong suppressive activity of IL-17A+Foxp3+ cells isolated from colon carcinoma [75]. This study further showed although the Treg population isolated tumor tissues of melanoma, renal cell carcinoma, colon carcinoma patients can show up to 31% of IL-17A-producers ex vivo. However, they exhibit a strong suppressive function in vitro, which was comparable to Tregs that had little or no IL-17 producers. Taken together, these data suggest that plasticity in Th17 cells may be one of the mechanisms by which effector-like Tregs develop in cancer. Although the precise role of these cells in tumor progression remains to be seen, these cells may contribute to promoting active inflammation and tumor development, while still retaining their suppressive capacity on T cells.

3.8. Allergy

Th1-like- Treg cells were first identified in the airway hyper-reactivity (AHR) setting in mice [19]. Studies by Scott et al., showed that the IFN-γ producing Foxp3+ cells arise from naïve CD4+CD25 T cells during Th1 polarized immune responses, upon stimulation with CD8α+ dendritic cells (DCs) in vivo. These Th1-like-Tregs expressing Foxp3, T-bet and inducible costimulator (ICOS) are antigen-specific. Moreover, these cells co-produce both IL-10, IFN-γ, and potently inhibit the development of allergen induced AHR, a function that can be inhibited by neutralization of IL-10, but not IFN-γ in mice. In human allergy there are no reports on Th1 like Tregs. However, ex vivo analyses of surgically removed nasal mucosa in allergic rhinitis and nasal polyposis patients show the presence of Th17-like Tregs cells co-expressing FOXP3, RORC, and IL-17A. These cells are found at higher frequency in patients with polyps than in those with allergic rhinitis alone. Further studies are required to delineate whether they play pathogenic roles in human allergy. Most recently, GATA-3+IRF-4 + IL-4 + IL-13 + Th2 like Tregs have been reported in a mouse allergic model [100]. They arise due to selective augmentation of IL-4 receptor signaling in Treg cells. Failure of Treg cells to maintain oral tolerance in food allergy has been directly attributed to the production of Th2 cytokines and impairment of suppressive function in these cells. Such allergen-specific Th2 like Treg cells are also found in the peripheral-blood of children with food allergy [100].

3.9. Infections

Although Tregs carry out specialized functions in various infections, their ability to produce pro-inflammatory cytokines was reported only in two infections listed below. During an acute and lethal infection by Toxoplasma gondii, a strong Th1 environment triggered by this microbe induces T-bet and IFN-γ expression in Treg cells in mice. This is associated with IL-2 exhaustion, and decline in Foxp3+ Treg cell numbers, coinciding with immunopathology and death in mice. These data support the idea that during an infection, Th1 environment can subvert regulatory networks and become pathogenic by promoting Th1-like Tregs [101]. In the context of oropharyngeal candidiasis (OPC), a Th17 infection model, Th17-like Tregs arise transiently in infected mice [78,102]. The increase is pronounced among Tregs in mouse oral lamina propria and intraepithelial cells (MOIL) isolated from tongue and palatal tissues of the infected mice [103]. Both Th17 effectors and nTregs contribute to the induction of Th17-like Tregs in vitro, likely reflecting the same scenario during candidal infection in vivo [78]. Functionally, it is clear that the development of these cells does not contribute to pathology during acute candida infection in mice. In fact, Tregs as a whole, play dual but protective roles in enhancing IL-17A and decreasing TNF-α, to respectively control acute infection and reduce immunopathology in mice [79,104]. It remains to be seen how Th17-like Tregs are specifically involved in each of these processes during infections.

4. Markers and mechanisms of development of pro-inflammatory cytokine producing Foxp3+ Tregs

4.1. Pro-inflammatory cytokine production in natural Foxp3+ cells vs induced Foxp3+ cells

One of the key questions about pro-inflammatory cytokine Foxp3+ cells is whether these cells develop from conventional effector T cells expressing promiscuous Foxp3, or reprogrammed natural regulatory T cells [50,52]. Human CD4+ cells are known to express FOXP3 transiently upon TCR activation [105]. Unlike nTreg cells, these activation induced FOXP3+ cells eventually lose FOXP3 expression, and do not suppress proliferation or cytokine production in effector cells. This is consistent to spontaneous up-regulation of FOXP3 in a proportion of human Th17 cells expanded from blood CD4+ T cells [106]. Therefore it is likely that non-suppressive Foxp3+ cells that are found in some diseases, and those that can be easily reprogrammed to lose Foxp3 expression, are derived from the effector cells in the periphery. However, effector like Tregs that retain their suppressive properties may have arisen from nTregs and pTregs. Although mouse CD4 cells do not spontaneously express Foxp3, TGF-β and IL-2 together can induce Foxp3+ cells in vitro. In contrast, during Th17 inducing conditions in the presence of TGF-β and IL-6, although Foxp3 induction is substantially reduced because of the inhibitory effect of IL-6, a few cells express Foxp3. Interestingly, a small proportion of these induced Foxp3+ cells also co-express IL-17A [78]. Using mixed cultures where CD45.2 nTregs are co-cultured with CD45.1 naïve cells under Th17 inducing conditions, it has been shown that both nTregs and induced Foxp3+ cells are capable of producing IL-17A in a IL-6 dependent manner [78]. These data support the notion that the majority of effector-like Treg cells develop from effector cells, while reprogrammed nTregs may also have a contribution to this population that arises in vivo. Moreover, the expression of Foxp3 in these cells may also confer them with suppressive activity, as shown in some instances where effector derived Foxp3+ cells expressing pro-inflammatory cytokines are shown to possess potent suppressive capacity [15,99].

4.2. Markers of effector-like Tregs

The most important markers of effector-like Tregs are the relevant lineage specific transcription factors: T-bet for Th1 like effector Tregs, GATA-3 for Th2 like Tregs, and ROR-γt for Th17 like Tregs. Although they express key transcription factors of effector cells, it is important to remember that not all the T-bet, GATA3, and ROR-γt -expressing Foxp3+ cells produce cytokines. Besides these intracellular markers, there are attempts to further classify these cells based on the cell surface markers. Recently, human Tregs are classified into three populations based on their stability, FOXP3, CD25 and CD45RA expression, namely CD4+CD25+CD45RA+ (stable resting Tregs), CD4+CD25+++Foxp3high CD45RA (activated Tregs), and CD4+CD25+Foxp3lowCD45RA (unstable Tregs) [107]. It has been shown that the cells with pro-inflammatory cytokine producing ability are enriched among unstable CD4+CD25+Foxp3lowCD45RA Treg cells. Specifically for IL-17A+Foxp3+ cells, CCR6, CD49d, HLA-DRneg and IL1R-β have also been described as markers [8,26,108,109]. Most recent studies further defined CD161, a natural killer cell marker, as a reliable marker for the Th17-like Tregs [11,24,86,87,108,110]. Taken together, effector like Tregs expressing proinflammatory cytokines can be detected by the expression of CD4+CD25hi CD127lo CD45RA HLA-DRneg CD161+Foxp3low markers, along with the specific lineage transcription factors and chemokine receptors. Although the markers for Th1 like Tregs are not well established, they may also express the above markers, and can be distinguished from Th17 like Tregs by the expression of T-bet. Not surprisingly, there are variations in the expression of some of these markers, which can be explained by variations in the milieu of different inflammatory conditions.

4.3. Mechanisms of induction of proinflammatory cytokine production in Tregs

4.3.1. Proinflammatory cytokines

The development of pro-inflammatory cytokine producing Tregs is strongly determined by the cytokine microenvironment. Foxp3+ Tregs have similar requirements like naïve cells to differentiate into respective lineages. For example, they require the cytokine IL-12 to develop into Th1-like Tregs. IL-6, IL-23 and IL1-β promote the differentiation of Tregs into Th17-like Tregs [26,78,86,111113]. Mechanistically, the conversion of Tregs into Th17-like Tregs relies on IL-6 and IL-1β dependent epigenetic modifications [8,26,109]. When this phenomenon was first reported in mice, IL-6 alone was shown to be able to induce Th17 producing cells in Foxp3+ Tregs in the presence of TGF-β [114]. Thus, as in Th17 cells, IL-6 dependent STAT3 activation, along with ROR-γt and ROR-α are required for IL-17A expression in Treg cells [26,112,115]. Most recent report also suggests that IL-6 promotes PIM1 kinase expression in in vitro expanded human Tregs. This kinase, by specifically phosphorylating FOXP3 at Ser(422), negatively regulates FOXP3 chromatin binding activity, likely reprogramming the cells [116]. While knockdown of PIM1 promoted FOXP3-induced suppressor functions, the role of PIM1 in the development of effector like Tregs remains to be studied. Because IL-2 and IL-15 are known suppressors of IL-17A production [117,118], and Tregs express higher levels of the receptors for these cytokines, it is probable that these cytokines control the differentiation of Th17-like Tregs. It remains to be seen whether these cytokines can be employed for therapeutic intervention to control IL-17A expression in Tregs. Indoleamine 2,3-dioxygenase (IDO), as well as Eos (Ikzf4), an obligate co-repressor for Foxp3, are also implicated in controlling the development of Th17 like Tregs in vitro and in vivo [119121]. Interestingly, in most situations, only a small population of Tregs becomes effector like and most Tregs do not. It is not clear whether it is due to very low frequencies of Ag-specific Tregs that respond in the tissue during inflammation. Suppressor of cytokine signaling 1 (SOCS1) plays an important role in Treg cell integrity and function, by protecting the cells from excessive inflammatory cytokines [16]. Although the underlying mechanisms of why only a small fraction of Tregs become effector like Tregs are unknown, SOCS1 may prevent most of the cells from becoming pro-inflammatory cytokine producers, while a few cells are relieved from these signals to become effector like Tregs.

4.3.2. TLR signaling

Another important signal that has been implicated in pro-inflammatory cytokine production in Tregs emanate from microbial stimuli that activate Toll like receptors (TLRs) [78,122]. TLRs are not only expressed by the innate immune cells and antigen presenting cells (APC) [123,124], they are also expressed in CD4 T cells, including Foxp3+ Tregs [123,125131]. It is now clear that majority of the tissue Tregs express high levels of TLR-2, and thus have an ability to respond to TLR-2 ligands directly [78]. TLR-2 signaling activation in the presence of APC in vitro, reverses Treg’s suppressive functions [112,122,129,132137], because of the pro-inflammatory cytokines produced by the APC responding to TLR-2 ligands [138]. Also, loss of Foxp3 is a prominent feature [139] in a heterogenous mixture of stable and unstable Tregs that were isolated only on the basis of CD25 marker [128,140]. However, when highly purified CD4+CD25highFoxp3GFP+Tregs isolated from Foxp3GFP mice are stimulated with TLR-2 ligands and IL-2 in the absence of APC, they show moderate proliferation without losing Foxp3 expression [31,78,141,142]. Most recent reports support the idea that Tregs proliferate in response to TLR-2 ligands, but they retain their regulatory activity under TLR activation conditions [8,78,139,140,143,144]. The latter tenet is more consistent with the view that TLR-2 ligands promote tolerance by inducing peripheral Tregs (pTregs), and do not reverse suppression [145]. Thus while TLR-9 signaling from commensal bacteria may negatively control the development of Tregs [146], TLR-2 signaling may promote the homeostasis of Tregs under steady-state conditions [78]. This is in line with the association of TLR-2 polymorphisms with changes in neonatal Treg numbers as well as allergies and atopic dermatitis [147]. However, under inflammatory Th17 conditions, when nTregs are stimulated with TLR-2 ligands in the presence of APC, IL-6, IL-23 and IL-1β, a minor fraction (<20%) of nTregs lose Foxp3. Interestingly, among nTregs that do not lose Foxp3, TLR-2 ligands also induce proliferation and IL-17A production [12,78]. Importantly, when mice are infected with a TLR-2 activating fungus, C. albicans, Foxp3+ cells expand and produce IL-17A in vivo [12,78]. Myd88 [148] [129,138] and TLR-2 are required, both in Tregs and APC for Foxp3+ cells to produce IL-17A [78]. TLR-2 dependent increase in Foxp3+IL-17A+ROR-γt+ cells is an additive effect of direct proliferation of Tregs, as well as IL-6 mediated induction of IL-17A. These findings are consistent with the role of TLR-2 signaling in promoting IL-17A in effector CD4 cells [149]. As C. albicans also activates dectin signaling, which has been implicated in IL-17A induction in Foxp3+ cells [115,150], it is possible that dectin1 is also involved in inducing Th17-like Treg cells during OPC. Moreover, the role of IL-1β dependent Myd88 signaling in Tregs cannot be excluded in the induction of Th17-like Treg cells. Thus excess of TLR-2 signaling from commensal bacteria, infections and endogenous ligands may all contribute to the development of Foxp3+ IL-17A+ cells in various inflammatory disease conditions. In this context, Staphylococcal enterotoxin B is also implicated in the differentiation of Tregs into RORC(+)Tregs that may be involved in the pathogenesis of eosinophilic nasal polyposis [151].

5. Effector transcription factors contribute to effector cytokine production in Tregs

The development of naive CD4 T cells into T helper cell lineages such as Th1, Th2, Treg and Th17 cells rely on lineage specific transcription such as T-bet, GATA-3, Foxp3 and ROR-γt respectively. Each Th lineage develops as a result of specific response to unique set of cytokines that instruct a specific differentiation program, and in turn exhibits unique profile of cytokines [152154]. Along with lineage specific master transcription factors, these lineages are also associated with other factors including signal transducer and activator of transcription (STAT) factors. While STAT1 and STAT4 are critical for Th1 regulation, STAT6 directs Th2 cells. While Th17-cell differentiation is dependent on a balance between signaling through STAT3 and interferon regulatory factor 4 (IRF-4) [155], Treg differentiation is dependent on STAT-5 and SMAD factors. Emerging concepts of Th lineage commitment suggest that Th-cell fate is not as irreversible as previously thought, and that lineage reprogramming can occur through the inducible expression of key transcription factors in all the Th subsets [152]. Similarly, the expression of effector Th lineage transcription factors exemplifies the plasticity in Tregs, and is a prerequisite for their effector characteristics.

Besides the classical Th subsets are the T follicular helper (Tfh) cells, which are required for survival, differentiation and class-switching of B cells in the germinal centers. Tfh cells are vital for the generation and maintenance of germinal center reactions and the long-lived humoral immunity. B-cell lymphoma (Bcl)-6, CXCR5, PD-1, SAP (SH2D1A), IL-4, IL-21, and ICOS, and the absence of Blimp-1 (prdm1) are the characteristic markers of Tfh cells [156158]. Similar to Tfh cells, CD4+CD25+ Foxp3+CD69 Treg cells are able to downregulate CCR7 and upregulate Bcl-6, CXCR5, programmed cell death 1(PD)-1 and ICOS. Functionally, these Treg cells migrate into the B-cell zone and suppress germinal center reactions [159161]. Although a fraction of Tfh like Treg cells lose Foxp3 expression and produce IL-17A in gut Peyer’s patches, they are incapable of producing the characteristic Tfh cytokines, namely, the IL-4 and IL-21 [17,162164].

5.1. Th effector lineage specific transcription factors

In many of the above mentioned diseases, IL-17A+Foxp3+ Th17 like Tregs also co-express ROR-γt, suggesting that ROR-γt is involved in IL-17A production in Foxp3+ cells. Most recent data reveal also the requirement of RUNX transcription factors for the induction of a Th17-like phenotype in Tregs [165,166]. Th17-like Tregs represent either early stages of cells undergoing Th17 and Treg differentiation, or plastic Tregs losing their activity during Th17 inflammation. As Th17 cells and Tregs have evolved greater developmental plasticity than Th1 and Th2 subsets, it is believed that these subsets can interconvert. This is because of the involvement of TGFβ, a cytokine that is common to both Th17 and Treg cell differentiation [167169]. While TGF-β receptor I signaling is required for both Foxp3 and IL-17A induction, the differential up-regulation of Foxp3 and IL-17A is dependent on its differential ability to activate mothers against decapentaplegic homologue (SMAD)4. Also, TGFβ induces Foxp3 in collaboration with IL-2 dependent STAT5 activation, whereas it induces ROR-γt in collaboration with IL-6 dependent STAT3 activation. As a result, ROR-γt and Foxp3 are often co-expressed at early stages of T cell differentiation, and these two transcription factors antagonize each other partly through protein–protein interaction in steady state conditions [170,171]. In the absence of IL-6 in the milieu, Foxp3 inhibits Th17 differentiation by antagonizing the function of the transcription factors ROR-γt and ROR-α. However, IL-6 overcomes this suppressive effect of Foxp3 and promotes Th17-like Tregs in a ROR-γt and STAT3 dependent manner [8,115]. These data suggest that during immune imbalance caused by inflammatory conditions, despite Foxp3 expression, the antagonistic effects of the protein may be lost. Studies in human Tregs have shown that, compared to IL-17Aneg Tregs and conventional T cells, IL-17A+Tregs express higher levels of ROR-γt after co-culture with conventional T cells and APCs, suggesting a complete absence of Foxp3 mediated antagonism [113]. It is clear that IL-6 mediated STAT3 activation is also critical in this phenomenon, as Treg cells from patients with STAT3 mutations are unable to produce IL-17A [86]. Interestingly, STAT-3 signaling is not only essential for the development of Th17-like Tregs, it has also been shown to be required in Tregs to suppress Th17 immunopathology in mice [172,173]. However, it is unknown whether Th17 like Tregs with active STAT-3 signaling, are programmed to engage in distinct effector-specific suppression of Th17 immunopathology in an inflammatory environment.

In the context of Th1-like Tregs, T-bet has been implicated in CXCR3 expression and IFN-γ production [70,71]. To delineate whether T-bet in Tregs causes IFN-γ dependent Th1 inflammation instead of suppressing it, mice with Treg-specific T-bet ablation were made. These mice do not develop autoimmune diseases at steady state suggesting that T-bet is dispensable for Tregs to suppress auto-reactive Th1 cells at steady state [174]. However, T-bet-deficient Tregs do not thrive in a Th1 environment and these cells are unable to suppress Scurfy effector Th1 cells in a transfer model. It appears that the actions of T-bet expressing Tregs at steady state are different from those under inflammatory conditions [54,175]. However, the role of antigen-specific T-bet-expressing Tregs and the function of T-bet and IFN-γ in these cells during Th1 immune responses are unknown and require future investigation.

Similarly, Th2 like Treg cells express GATA3, the master transcription factors of Th2 cells [166,174,176,177,178]. Genome-wide pattern of GATA3 binding in various T cells has been profiled through chromatin immune-precipitation followed by high throughput sequencing (ChIP-Seq) analysis. Although GATA3 binds to the CGRE site in the Il13 distal promoter in regulatory T cells as in Th2 cells, GATA3-expressing Tregs do not produce IL-13. It is possible that Foxp3 suppresses the function of GATA3 to induce the expression of Th2 cytokines. In accordance to this tenet, a modest reduction of Foxp3 expression in Tregs results in the production of Th2 cytokines in these cells [179]. GATA3 also regulates many Th2-specific genes including Il1rl1, which encodes the IL-33 receptor T1/ST2, and chemokine receptor gene Ccr8 in Tregs [180]. Although Treg-specific deletion of GATA3 in mice result in spontaneous autoimmunity in one study [177], three other studies report that mice with Treg-specific GATA3 deletion do not develop any disease up to 6 months of age [166,174,176]. Therefore, GATA3 expression in Tregs may not be required to for these Tregs to control auto-reactive Th2 cells at steady state. It seems that the suppressive effect of Foxp3 on GATA3 is strong enough to restrain the production of Th2 cytokines in Tregs. However, whether GATA3 can overcome the restraining effect of Foxp3 and contribute to production of these cytokines in Tregs during a strong Th2 inflammation remains to be seen.

Although T-bet- and GATA3-expressing Tregs can be found at steady state, these cells do not represent stable Treg subsets in mice [174]. The expression of T-bet and/or GATA3 in Tregs is highly dynamic and is often associated with TCR activation of the Tregs. When Tregs are stimulated by TCR and/or IL-2 stimulation, they can either up-regulate T-bet or GATA3, each contributing to the suppression of ROR-γt expression and stabilization of Foxp3 [176]. T-bet-expressing Tregs are enriched among CD44hi population, and express higher levels of Treg-specific genes such as GITR and CTLA-4 than T-bet-non-expressing Tregs [71]. Interestingly, although specific deletion of either Tbx21 or Gata3 genes alone in Tregs does not alter the functions and phenotype of Tregs, combined deletion of both the genes in these cells results in the development of autoimmune-like diseases even at steady state. T-bet-GATA3 double-deficient Tregs lose their suppressive functions. Strikingly, this is accompanied by the up-regulation of ROR-γt expression and down-regulation of Foxp3 expression. Without T-bet and GATA3, Foxp3 expression cannot be maintained in Tregs, possibly because of the up-regulation of antagonistic protein, RORγt. Most importantly, T-bet-GATA3 double-deficient Tregs also produce IL-17A, suggesting that Treg cells may utilize several cross-regulatory mechanisms that are adopted by T effector cells.

Taken together, a balance between the expression and cross-regulation of hallmark transcription factors T-bet, GATA3 and RORγt in Tregs may determine their suppressive functions and pro-inflammatory cytokine production [174]. It is similar to T helper cells whose lineage transcription factors can cross-suppress other lineages during differentiation [181187]. Similarly, under steady state conditions, a delicate balance between the transcription factors would normally stabilize Foxp3 and suppress the function of effector transcription factors in Tregs. However, an inflammatory situation may disrupt this balance and cause dys-regulation in Tregs. Thus inflammation driven activation of one or more effector transcription factors in Tregs contributes to the reprogramming Tregs into effector-like Tregs in vivo.

5.2. IRF4

In effector T cells, IRF4 plays a critical role during the differentiation of Th2, Th17 and T follicular helper (Tfh) cells [188]. Although Tregs acquire effector Tfh cell characteristics to regulate Tfh and germinal center responses, it is dependent on Bcl-6 and CXCR5 expression but not IRF4 [159,160]. In Tregs, IRF4 in collaboration with Foxp3, regulates a subset of Treg-specific genes. It is well known that, to gain full suppressive activity, Tregs need to be activated through their TCR [189]. Together with IL-2-mediated STAT5 activation, TCR stimulation induces the activation of several constitutively expressed transcription factors such as NFATs, NF-κb and AP-1 family members, along with the expression of many transcription factors including Egr1, Egr2 and NFATc1 and IRF4 in Tregs [190]. Though all of these transcription factors may contribute to Treg suppressive functions, it is believed that IRF4 is critical for maintenance of Treg functionality. Consistent to this idea, acute ablation of TCR in mature Tregs, accompanied by the down-regulation of IRF4 in activated Tregs, also induces autoimmune diseases [191,192]. Treg specific deletion of Irf4 results in severe Th2-related autoimmunity [193]. Since IRF4 is expressed in all activated Tregs [35] and IRF4 is involved in optimal Treg activation, it is speculated that the suppression of auto-reactive Th2 cells may require higher Treg suppressive activity than auto-reactive Th1 or Th17 cells. Thus, transcriptional regulatory network and IRF4 activity fueled by continuous TCR stimulation in Tregs is essential for maintaining immune tolerance. Whether IRF4 contributes to pro-inflammatory cytokine production in Tregs during immune dys-regulation has not been explored.

6. Do pro-inflammatory cytokine producing Foxp3+ Tregs retain suppressive capacities?

Induction of Foxp3+ effector-like Treg cells under strong pro-inflammatory conditions is either associated with or without the loss of Foxp3 [51,139,194]. In instances where they lose Foxp3 expression, they either fail to control inflammation or even exacerbate pathogenic immune responses [18,194,195]. This raises an interesting possibility that Treg functions are not limited solely to suppression but also to other effector functions that may be largely dependent on cytokine milieu or innate immune responses [79,196,197]. Although a small fraction of Tregs themselves loses Foxp3 in Th17 inflammatory conditions in vitro, the destabilization is only a transient phenomenon. Adoptive transfer of such Tregs, a proportion of which includes the destabilized cells, exhibits delayed but strong potential to suppress Th17 inflammation chronic Th17 IBD model [79]. The transient loss of Foxp3 and/or suppressive functions in Tregs is attributed to the inflammatory cytokines in the milieu [138], as they lose their suppressive capacity only upon strong activation in the presence of IL-1β and IL-6 [8,79]. Withdrawal of these inflammatory cytokines restores the suppressive functions in Tregs. This mechanism of transient silencing of Tregs, along with the strong Th effector responses might allow subset-specific protective immune responses against microbes to proceed normally in cytokine-rich environments, such as during acute infections [196,198,199]. Although a small proportion of Foxp3+ IL-17A+ cells develop in response to C. albicans in the oral cavity, the mice clear the infection and immunopathology normally in a Th17 and Treg dependent manner [79,102,200]. Although the fungal clearance and immunopathology required the presence of Tregs as a whole, the role of Foxp3+ IL-17A+ cells in mediating the protection is not known. Although these Foxp3+IL-17A+ cells do not cause immunopathology during acute infection, whether these cells contribute to protective functions of Tregs remains to be investigated. As the expression of lineage specific transcription factors are required in Tregs to control the respective lineages [201], it can be cautiously speculated that RORgt+IL-17A+Foxp3+ cells are the Th17-specific regulatory cells that may control infection associated Th17 immunopathology. These data support the possibility that effector like Foxp3+ cells found in tissues can retain the suppressive capacities. Thus the accumulation of these pro-inflammatory Treg cells in the inflammatory tissues could be a result of auto-antigen mediated proliferation and activation of Tregs. These effector/memory Tregs may be actively recruited to the tissue for controlling the immunopathology. This idea is consistent to the data that showed thymus derived regulatory T cells become activated, proliferate and differentiate into memory Tregs and serve as more potent suppressors mediating resolution of organ-specific autoimmunity in mice [202]. As pro-inflammatory cytokine producing Tregs are known to be enriched among Tregs with memory characteristics, it is tempting to speculate that these cells may express the respective inflammatory cytokines as well to establish “regulatory memory”. At least in regards to IBD, RA and multiple sclerosis, the pro-inflammatory cytokine producing Treg cells with memory characteristics are functionally as suppressive as conventional Treg cells. However, one cannot rule out that their increased persistence during chronic inflammation such as psoriasis may contribute to worsening of the immunopathology. The differences in the cytokines, metabolites and commensal diversity in the milieu are likely critical in determining whether Tregs may adopt suppressive or non-suppressive modes in different diseases.

7. Concluding remarks and perspective

Pro-inflammatory cytokine producing Foxp3+Treg cells expressing lineage-specific effector transcription factors are extensively documented in various inflammatory diseases. A growing number of recent studies from mouse models reveal that pro-inflammatory cytokines and TLR signaling promote the induction of these cells. The expression of effector transcription factors in Tregs does not necessarily imply that they lose their suppressive capacities. In fact Tregs require lineage specific effector transcription factors along with Foxp3 to suppress respective lineages and the resulting inflammation [71,173,193]. It appears that Tregs may adopt the transcriptional program of lineage-specific effector cells to selectively suppress cells of that same lineage. However, a considerable overlap between the transcriptional program, with a concomitant loss of FOXP3 expression presents the potential for conversion of Tregs into effector cells and Treg lineage instability [18]. This is seen in the context of a lethal infection by Toxoplasma gondii where T-bet and IFN-γ expressing Foxp3+ cells have been associated with immunopathogenesis. Similarly, Th17 like Tregs are associated with dys-regulation in psoriasis patients. However in other diseases such as IBD, MS, RA, allergy, candidal infection and cancer, effector like Tregs retain their suppressive properties. These Tregs, whether suppressive or non-suppressive, may be functionally specialized to their local environment that determines their phenotype, suppressive function and pro- and anti-inflammatory cytokine production [1,15]. Therefore the functions of the effector-like Foxp3+ cells is strongly dependent on environmental cues and local pro-inflammatory cytokines, metabolites, and possibly whether they originated from nTregs, pTregs or effector cells. Improved understanding of how Treg subsets respond to ranging environmental cues that direct the heterogeneity within these subsets is key to direct efforts to employ these cells for clinical intervention.

Acknowledgments

The project is supported by the startup funding to PP from the Department of Biological Sciences, SODM, Case Western Reserve University and the Skin disease research center pilot grant (from P30AR39750) to PP. J.Z. is supported by the Division of Intramural Research (DIR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), USA.

Footnotes

Conflicts of interest

The authors declare no conflict of interest.

References

  • 1.Pandiyan P, Zheng L, Lenardo MJ. The molecular mechanisms of regulatory T cell immunosuppression. Front Immunol. 2011;2:60. doi: 10.3389/fimmu.2011.00060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Rudensky AY, Campbell DJ. In vivo sites and cellular mechanisms of T reg cell-mediated suppression. J Exp Med. 2006;203:489–92. doi: 10.1084/jem.20060214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Brunstein CG, Miller JS, Cao Q, McKenna DH, Hippen KL, Curtsinger J, et al. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood. 2011;117:1061–70. doi: 10.1182/blood-2010-07-293795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Canavan JB, Scotta C, Vossenkamper A, Goldberg R, Elder MJ, Shoval I, et al. Developing in vitro expanded CD45RA+ regulatory T cells as an adoptive cell therapy for Crohn’s disease. Gut. 2015 doi: 10.1136/gutjnl-2014-306919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kasagi S, Zhang P, Che L, Abbatiello B, Maruyama T, Nakatsukasa H, et al. In vivo-generated antigen-specific regulatory T cells treat autoimmunity without compromising antibacterial immune response. Sci Translat Med. 2014;6:241ra78. doi: 10.1126/scitranslmed.3008895. [DOI] [PubMed] [Google Scholar]
  • 6.Koreth J, Matsuoka K, Kim HT, McDonough SM, Bindra B, Alyea EP, 3rd, et al. Interleukin-2 and regulatory T cells in graft-versus-host disease. New England J Med. 2011;365:2055–66. doi: 10.1056/NEJMoa1108188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Singer BD, King LS, D’Alessio FR. Regulatory T cells as immunotherapy. Front Immunol. 2014;5:46. doi: 10.3389/fimmu.2014.00046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Beriou G, Costantino CM, Ashley CW, Yang L, Kuchroo VK, Baecher-Allan C, et al. IL-17-producing human peripheral regulatory T cells retain suppressive function. Blood. 2009;113:4240–9. doi: 10.1182/blood-2008-10-183251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Du R, Zhao H, Yan F, Li H. IL-17+Foxp3+ T cells: an intermediate differentiation stage between Th17 cells and regulatory T cells. J Leukoc Biol. 2014;96:39–48. doi: 10.1189/jlb.1RU0114-010RR. [DOI] [PubMed] [Google Scholar]
  • 10.Hovhannisyan Z, Treatman J, Littman DR, Mayer L. Characterization of interleukin-17-producing regulatory T cells in inflamed intestinal mucosa from patients with inflammatory bowel diseases. Gastroenterology. 2011;140:957–65. doi: 10.1053/j.gastro.2010.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Voo KS, Wang YH, Santori FR, Boggiano C, Wang YH, Arima K, et al. Identification of IL-17-producing FOXP3+ regulatory T cells in humans. Proc Natl Acad Sci US A. 2009;106:4793–8. doi: 10.1073/pnas.0900408106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Whibley N, Maccallum DM, Vickers MA, Zafreen S, Waldmann H, Hori S, et al. Expansion of Foxp3(+) T-cell populations by Candida albicans enhances both Th17-cell responses and fungal dissemination after intravenous challenge. Eur J Immunol. 2014;44:1069–83. doi: 10.1002/eji.201343604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Barbi J, Pardoll D, Pan F. Treg functional stability and its responsiveness to the microenvironment. Immunol Rev. 2014;259:115–39. doi: 10.1111/imr.12172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Duarte JH, Zelenay S, Bergman ML, Martins AC, Demengeot J. Natural Treg cells spontaneously differentiate into pathogenic helper cells in lymphopenic conditions. Eur J Immunol. 2009;39:948–55. doi: 10.1002/eji.200839196. [DOI] [PubMed] [Google Scholar]
  • 15.Pesenacker AM, Broady R, Levings MK. Control of tissue-localized immune responses by human regulatory T cells. Eur J Immunol. 2014 doi: 10.1002/eji.201344205. [DOI] [PubMed] [Google Scholar]
  • 16.Takahashi R, Yoshimura A. SOCS1 and regulation of regulatory T cells plasticity. J Immunol Res. 2014;2014:943149. doi: 10.1155/2014/943149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Tsuji M, Komatsu N, Kawamoto S, Suzuki K, Kanagawa O, Honjo T, et al. Preferential generation of follicular B helper T cells from Foxp3+ T cells in gut Peyer’s patches. Science. 2009;323:1488–92. doi: 10.1126/science.1169152. [DOI] [PubMed] [Google Scholar]
  • 18.Zhou X, Bailey-Bucktrout S, Jeker LT, Bluestone JA. Plasticity of CD4(+) FoxP3(+) T cells. Curr Opin Immunol. 2009;21:281–5. doi: 10.1016/j.coi.2009.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Stock P, Akbari O, Berry G, Freeman GJ, Dekruyff RH, Umetsu DT. Induction of T helper type 1-like regulatory cells that express Foxp3 and protect against airway hyper-reactivity. Nat Immunol. 2004;5:1149–56. doi: 10.1038/ni1122. [DOI] [PubMed] [Google Scholar]
  • 20.Bovenschen HJ, van de Kerkhof PC, van Erp PE, Woestenenk R, Joosten I, Koenen HJ. Foxp3+ regulatory T cells of psoriasis patients easily differentiate into IL-17A-producing cells and are found in lesional skin. J Invest Dermatol. 2011;131:1853–60. doi: 10.1038/jid.2011.139. [DOI] [PubMed] [Google Scholar]
  • 21.Dominguez-Villar M, Baecher-Allan CM, Hafler DA. Identification of T helper type 1-like, Foxp3+ regulatory T cells in human autoimmune disease. Nat Med. 2011;17:673–5. doi: 10.1038/nm.2389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sanchez Rodriguez R, Pauli ML, Neuhaus IM, Yu SS, Arron ST, Harris HW, et al. Memory regulatory T cells reside in human skin. J Clin Invest. 2014;124:1027–36. doi: 10.1172/JCI72932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Sugiyama H, Gyulai R, Toichi E, Garaczi E, Shimada S, Stevens SR, et al. Dysfunctional blood and target tissue CD4+ CD25high regulatory T cells in psoriasis: mechanism underlying unrestrained pathogenic effector T cell proliferation. J Immunol. 2005;174:164–73. doi: 10.4049/jimmunol.174.1.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ayyoub M, Deknuydt F, Raimbaud I, Dousset C, Leveque L, Bioley G, et al. Human memory FOXP3+ Tregs secrete IL-17 ex vivo and constitutively express the T(H)17 lineage-specific transcription factor RORgamma t. Proc Natl Acad Sci USA. 2009;106:8635–40. doi: 10.1073/pnas.0900621106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hirahara K, Liu L, Clark RA, Yamanaka K, Fuhlbrigge RC, Kupper TS. The majority of human peripheral blood CD4+CD25highFoxp3+ regulatory T cells bear functional skin-homing receptors. J Immunol. 2006;177:4488–94. doi: 10.4049/jimmunol.177.7.4488. [DOI] [PubMed] [Google Scholar]
  • 26.Koenen HJ, Smeets RL, Vink PM, van Rijssen E, Boots AM, Joosten I. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17-producing cells. Blood. 2008;112:2340–52. doi: 10.1182/blood-2008-01-133967. [DOI] [PubMed] [Google Scholar]
  • 27.Colonna L, Florek M, Leveson-Gower DB, Sega EI, Baker J, Smith AT, et al. IL-17 gene ablation does not impact Treg-mediated suppression of graft-versus-host disease after bone marrow transplantation. Biol. Blood Marrow Transplant. J Am Soc Blood Marrow Transplant. 2013;19:1557–65. doi: 10.1016/j.bbmt.2013.07.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rudensky A. Foxp3 and dominant tolerance. Philos Trans R Soc Lond B Biol Sci. 2005;360:1645–6. doi: 10.1098/rstb.2005.1731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Himmel ME, MacDonald KG, Garcia RV, Steiner TS, Levings MK. Helios+ and Helios- cells coexist within the natural FOXP3+ T regulatory cell subset in humans. J Immunol. 2013;190:2001–8. doi: 10.4049/jimmunol.1201379. [DOI] [PubMed] [Google Scholar]
  • 30.Bensinger SJ, Walsh PT, Zhang J, Carroll M, Parsons R, Rathmell JC, et al. Distinct IL-2 receptor signaling pattern in CD4+CD25+ regulatory T cells. J Immunol. 2004;172:5287–96. doi: 10.4049/jimmunol.172.9.5287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pandiyan P, Lenardo MJ. The control of CD4+CD25+Foxp3+ regulatory T cell survival. Biol Direct. 2008;3:6. doi: 10.1186/1745-6150-3-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Wu YQ, Borde M, Heissmeyer V, Feuerer M, Lapan AD, Stroud JC, et al. FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell. 2006;126:375–87. doi: 10.1016/j.cell.2006.05.042. [DOI] [PubMed] [Google Scholar]
  • 33.Ono M, Yaguchi H, Ohkura N, Kitabayashi I, Nagamura Y, Nomura T, et al. Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1. Nature. 2007;446:685–9. doi: 10.1038/nature05673. [DOI] [PubMed] [Google Scholar]
  • 34.Pan F, Yu H, Dang EV, Barbi J, Pan X, Grosso JF, et al. Eos mediates Foxp3-dependent gene silencing in CD4+ regulatory T cells. Science. 2009;325:1142–6. doi: 10.1126/science.1176077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cretney E, Xin A, Shi W, Minnich M, Masson F, Miasari M, et al. The transcription factors Blimp-1 and IRF4 jointly control the differentiation and function of effector regulatory T cells. Nat Immunol. 2011;12:304–11. doi: 10.1038/ni.2006. [DOI] [PubMed] [Google Scholar]
  • 36.Li B, Greene MI. FOXP3 actively represses transcription by recruiting the HAT/ HDAC complex. Cell Cycle (Georgetown, Tex) 2007;6:1432–6. [PubMed] [Google Scholar]
  • 37.Ochs HD, Oukka M, Torgerson TR. TH17 cells and regulatory T cells in primary immunodeficiency diseases. J Allergy Clin Immunol. 2009;123:977–83. doi: 10.1016/j.jaci.2009.03.030. quiz 84-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–1. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
  • 39.Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20. doi: 10.1038/83707. [DOI] [PubMed] [Google Scholar]
  • 40.Gavin MA, Rasmussen JP, Fontenot JD, Vasta V, Manganiello VC, Beavo JA, et al. Foxp3-dependent programme of regulatory T-cell differentiation. Nature. 2007;445:771–5. doi: 10.1038/nature05543. [DOI] [PubMed] [Google Scholar]
  • 41.Hill JA, Benoist C, Mathis D. Treg cells: guardians for life. Nat Immunol. 2007;8:124–5. doi: 10.1038/ni0207-124. [DOI] [PubMed] [Google Scholar]
  • 42.Ohkura N, Hamaguchi M, Sakaguchi S. FOXP3+ regulatory T cells: control of FOXP3 expression by pharmacological agents. Trends Pharmacol Sci. 2011;32:158–66. doi: 10.1016/j.tips.2010.12.004. [DOI] [PubMed] [Google Scholar]
  • 43.Ouyang W, Liao W, Luo CT, Yin N, Huse M, Kim MV, et al. Novel Foxo1-dependent transcriptional programs control T(reg) cell function. Nature. 2012;491:554–9. doi: 10.1038/nature11581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Samstein RM, Arvey A, Josefowicz SZ, Peng X, Reynolds A, Sandstrom R, et al. Foxp3 exploits a pre-existent enhancer landscape for regulatory T cell lineage specification. Cell. 2012;151:153–66. doi: 10.1016/j.cell.2012.06.053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Baron U, Floess S, Wieczorek G, Baumann K, Grutzkau A, Dong J, et al. DNA demethylation in the human FOXP3 locus discriminates regulatory T cells from activated FOXP3(+) conventional T cells. Eur J Immunol. 2007;37:2378–89. doi: 10.1002/eji.200737594. [DOI] [PubMed] [Google Scholar]
  • 46.Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol. 2012;30:531–64. doi: 10.1146/annurev.immunol.25.022106.141623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Abbas AK, Benoist C, Bluestone JA, Campbell DJ, Ghosh S, Hori S, et al. Regulatory T cells: recommendations to simplify the nomenclature. Nat Immunol. 2013;14:307–8. doi: 10.1038/ni.2554. [DOI] [PubMed] [Google Scholar]
  • 48.Shevach EM. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity. 2009;30:636–45. doi: 10.1016/j.immuni.2009.04.010. [DOI] [PubMed] [Google Scholar]
  • 49.Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev. 2010;10:490–500. doi: 10.1038/nri2785. [DOI] [PubMed] [Google Scholar]
  • 50.Miyao T, Floess S, Setoguchi R, Luche H, Fehling HJ, Waldmann H, et al. Plasticity of Foxp3(+) T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells. Immunity. 2012;36:262–75. doi: 10.1016/j.immuni.2011.12.012. [DOI] [PubMed] [Google Scholar]
  • 51.Rubtsov YP, Niec RE, Josefowicz S, Li L, Darce J, Mathis D, et al. Stability of the regulatory T cell lineage in vivo. Science. 2010;329:1667–71. doi: 10.1126/science.1191996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hori S. Lineage stability and phenotypic plasticity of Foxp3(+) regulatory T cells. Immunol Rev. 2014;259:159–72. doi: 10.1111/imr.12175. [DOI] [PubMed] [Google Scholar]
  • 53.Vignali D. How many mechanisms do regulatory T cells need? Eur J Immunol. 2008;38:908–11. doi: 10.1002/eji.200738114. [DOI] [PubMed] [Google Scholar]
  • 54.Yamaguchi T, Wing JB, Sakaguchi S. Two modes of immune suppression by Foxp3(+) regulatory T cells under inflammatory or non-inflammatory conditions. Semin Immunol. 2011;23:424–30. doi: 10.1016/j.smim.2011.10.002. [DOI] [PubMed] [Google Scholar]
  • 55.Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJ. Cutting edge: contact-mediated suppression by CD4+ CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J Immunol. 2005;174:1783–6. doi: 10.4049/jimmunol.174.4.1783. [DOI] [PubMed] [Google Scholar]
  • 56.Klein L, Khazaie K, von Boehmer H. In vivo dynamics of antigen-specific regulatory T cells not predicted from behavior in vitro. Proc Natl Acad Sci US A. 2003;100:8886–91. doi: 10.1073/pnas.1533365100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pandiyan P, Lenardo M. Comment on “Cutting edge: regulatory T cells do not mediate suppression via programmed cell death pathways”. J Immunol. 2012;188:5203–4. doi: 10.4049/jimmunol.1290023. author reply 4–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Pandiyan P, Zheng L, Ishihara S, Reed J, Lenardo MJ. CD4(+)CD25(+)Foxp3(+) regulatory T cells induce cytokine deprivation-mediated apoptosis of effector CD4(+) T cells. Nat Immunol. 2007;8:1353–62. doi: 10.1038/ni1536. [DOI] [PubMed] [Google Scholar]
  • 59.Pillai MR, Collison LW, Wang X, Finkelstein D, Rehg JE, Boyd K, et al. The plasticity of regulatory T cell function. J Immunol. 2011;187:4987–97. doi: 10.4049/jimmunol.1102173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Fontenot JD, Rudensky AY. A well adapted regulatory contrivance: regulatory T cell development and the forkhead family transcription factor Foxp3. Nat Immunol. 2005;6:331–7. doi: 10.1038/ni1179. [DOI] [PubMed] [Google Scholar]
  • 61.Golden JB, McCormick TS, Ward NL. IL-17 in psoriasis: implications for therapy and cardiovascular co-morbidities. Cytokine. 2013;62:195–201. doi: 10.1016/j.cyto.2013.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zhang L, Yang XQ, Cheng J, Hui RS, Gao TW. Increased Th17 cells are accompanied by FoxP3(+) Treg cell accumulation and correlated with psoriasis disease severity. Clin Immunol. 2010;135:108–17. doi: 10.1016/j.clim.2009.11.008. [DOI] [PubMed] [Google Scholar]
  • 63.Imanguli MM, Cowen EW, Rose J, Dhamala S, Swaim W, Lafond S, et al. Comparative analysis of FoxP3(+) regulatory T cells in the target tissues and blood in chronic graft versus host disease. Leukemia. 2014;28:2016–27. doi: 10.1038/leu.2014.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Imanguli MM, Swaim WD, League SC, Gress RE, Pavletic SZ, Hakim FT. Increased T-bet+ cytotoxic effectors and type I interferon-mediated processes in chronic graft-versus-host disease of the oral mucosa. Blood. 2009;113:3620–30. doi: 10.1182/blood-2008-07-168351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Rieger K, Loddenkemper C, Maul J, Fietz T, Wolff D, Terpe H, et al. Mucosal FOXP3+ regulatory T cells are numerically deficient in acute and chronic GvHD. Blood. 2006;107:1717–23. doi: 10.1182/blood-2005-06-2529. [DOI] [PubMed] [Google Scholar]
  • 66.Fondi C, Nozzoli C, Benemei S, Baroni G, Saccardi R, Guidi S, et al. Increase in FOXP3+ regulatory T cells in GVHD skin biopsies is associated with lower disease severity and treatment response. Biol Blood Marrow Transplant: J Am Soc Blood Marrow Transplant. 2009;15:938–47. doi: 10.1016/j.bbmt.2009.04.009. [DOI] [PubMed] [Google Scholar]
  • 67.Choi J, Ritchey J, Prior JL, Holt M, Shannon WD, Deych E, et al. In vivo administration of hypomethylating agents mitigate graft-versus-host disease without sacrificing graft-versus-leukemia. Blood. 2010;116:129–39. doi: 10.1182/blood-2009-12-257253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Sanchez-Abarca LI, Gutierrez-Cosio S, Santamaria C, Caballero-Velazquez T, Blanco B, Herrero-Sanchez C, et al. Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting. Blood. 2010;115:107–21. doi: 10.1182/blood-2009-03-210393. [DOI] [PubMed] [Google Scholar]
  • 69.Trzonkowski P, Dukat-Mazurek A, Bieniaszewska M, Marek-Trzonkowska N, Dobyszuk A, Juscinska J, et al. Treatment of graft-versus-host disease with naturally occurring T regulatory cells. BioDrugs: Clin Immunotherapeut, Biopharmaceut Gene Ther. 2013;27:605–14. doi: 10.1007/s40259-013-0050-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Koch MA, Thomas KR, Perdue NR, Smigiel KS, Srivastava S, Campbell DJ. T-bet(+) Treg cells undergo abortive Th1 cell differentiation due to impaired expression of IL-12 receptor beta2. Immunity. 2012;37:501–10. doi: 10.1016/j.immuni.2012.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.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: 10.1038/ni.1731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Cong Y, Feng T, Fujihashi K, Schoeb TR, Elson CO. A dominant, coordinated T regulatory cell-IgA response to the intestinal microbiota. Proc Natl Acad Sci US A. 2009;106:19256–61. doi: 10.1073/pnas.0812681106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Cebula A, Seweryn M, Rempala GA, Pabla SS, McIndoe RA, Denning TL, et al. Thymus-derived regulatory T cells contribute to tolerance to commensal microbiota. Nature. 2013;497:258–62. doi: 10.1038/nature12079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451–5. doi: 10.1038/nature12726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Kryczek I, Wu K, Zhao E, Wei S, Vatan L, Szeliga W, et al. IL-17+ regulatory T cells in the microenvironments of chronic inflammation and cancer. J Immunol. 2011;186:4388–95. doi: 10.4049/jimmunol.1003251. [DOI] [PubMed] [Google Scholar]
  • 76.Li L, Boussiotis VA. The role of IL-17-producing Foxp3+ CD4+ T cells in inflammatory bowel disease and colon cancer. Clin Immunol. 2013;148:246–53. doi: 10.1016/j.clim.2013.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Ueno A, Jijon H, Chan R, Ford K, Hirota C, Kaplan GG, et al. Increased prevalence of circulating novel IL-17 secreting Foxp3 expressing CD4+ T cells and defective suppressive function of circulating Foxp3+ regulatory cells support plasticity between Th17 and regulatory T cells in inflammatory bowel disease patients. Inflamm Bowel Dis. 2013;19:2522–34. doi: 10.1097/MIB.0b013e3182a85709. [DOI] [PubMed] [Google Scholar]
  • 78.Bhaskaran N, Cohen S, Zhang Y, Weinberg A, Pandiyan P. TLR-2 signaling promotes IL-17A production in CD4+CD25+Foxp3+ regulatory cells during Oropharyngeal candidiasis. Pathogens. 2015;4:90–110. doi: 10.3390/pathogens4010090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Pandiyan P, Conti HR, Zheng L, Peterson AC, Mathern DR, Hernandez-Santos N, et al. CD4(+)CD25(+)Foxp3(+) regulatory T cells promote Th17 cells in vitro and enhance host resistance in mouse Candida albicans Th17 cell infection model. Immunity. 2011;34:422–34. doi: 10.1016/j.immuni.2011.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Feng T, Cao AT, Weaver CT, Elson CO, Cong Y. Interleukin-12 converts Foxp3+ regulatory T cells to interferon-gamma-producing Foxp3+ T cells that inhibit colitis. Gastroenterology. 2011;140:2031–43. doi: 10.1053/j.gastro.2011.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Okui T, Aoki Y, Ito H, Honda T, Yamazaki K. The presence of IL-17+/FOXP3+ double-positive cells in periodontitis. J Dent Res. 2012;91:574–9. doi: 10.1177/0022034512446341. [DOI] [PubMed] [Google Scholar]
  • 82.Nistala K, Adams S, Cambrook H, Ursu S, Olivito B, de Jager W, et al. Th17 plasticity in human autoimmune arthritis is driven by the inflammatory environment. Proc Natl Acad Sci US A. 2010;107:14751–6. doi: 10.1073/pnas.1003852107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Nistala K, Moncrieffe H, Newton KR, Varsani H, Hunter P, Wedderburn LR. Interleukin-17-producing T cells are enriched in the joints of children with arthritis, but have a reciprocal relationship to regulatory T cell numbers. Arthritis Rheum. 2008;58:875–87. doi: 10.1002/art.23291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Jandus C, Bioley G, Rivals JP, Dudler J, Speiser D, Romero P. Increased numbers of circulating polyfunctional Th17 memory cells in patients with seronegative spondylarthritides. Arthritis Rheum. 2008;58:2307–17. doi: 10.1002/art.23655. [DOI] [PubMed] [Google Scholar]
  • 85.Yamada H, Nakashima Y, Okazaki K, Mawatari T, Fukushi JI, Kaibara N, et al. Th1 but not Th17 cells predominate in the joints of patients with rheumatoid arthritis. Ann Rheum Dis. 2008;67:1299–304. doi: 10.1136/ard.2007.080341. [DOI] [PubMed] [Google Scholar]
  • 86.Afzali B, Mitchell PJ, Edozie FC, Povoleri GA, Dowson SE, Demandt L, et al. CD161 expression characterizes a subpopulation of human regulatory T cells that produces IL-17 in a STAT3-dependent manner. Eur J Immunol. 2013;43:2043–54. doi: 10.1002/eji.201243296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Pesenacker AM, Bending D, Ursu S, Wu Q, Nistala K, Wedderburn LR. CD161 defines the subset of FoxP3+ T cells capable of producing proinflammatory cytokines. Blood. 2013;121:2647–58. doi: 10.1182/blood-2012-08-443473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Pesenacker AM, Wedderburn LR. T regulatory cells in childhood arthritis-novel insights. Expert Rev Mol Med. 2013;15:e13. doi: 10.1017/erm.2013.14. [DOI] [PubMed] [Google Scholar]
  • 89.Nylander A, Hafler DA. Multiple sclerosis. J Clin Invest. 2012;122:1180–8. doi: 10.1172/JCI58649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Cua DJ, Sherlock J, Chen Y, Murphy CA, Joyce B, Seymour B, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. 2003;421:744–8. doi: 10.1038/nature01355. [DOI] [PubMed] [Google Scholar]
  • 91.El-Behi M, Ciric B, Dai H, Yan Y, Cullimore M, Safavi F, et al. The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol. 2011;12:568–75. doi: 10.1038/ni.2031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kleinewietfeld M, Manzel A, Titze J, Kvakan H, Yosef N, Linker RA, et al. Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells. Nature. 2013;496:518–22. doi: 10.1038/nature11868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Lee YK, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci US A. 2011;108(Suppl 1):4615–22. doi: 10.1073/pnas.1000082107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.McPherson RC, Turner DG, Mair I, O’Connor RA, Anderton SM. T-bet expression by Foxp3(+) T regulatory cells is not essential for their suppressive function in CNS autoimmune disease or colitis. Front Immunol. 2015;6:69. doi: 10.3389/fimmu.2015.00069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.McClymont SA, Putnam AL, Lee MR, Esensten JH, Liu W, Hulme MA, et al. Plasticity of human regulatory T cells in healthy subjects and patients with type 1 diabetes. J Immunol. 2011;186:3918–26. doi: 10.4049/jimmunol.1003099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mao Y, Poschke I, Kiessling R. Tumour-induced immune suppression: role of inflammatory mediators released by myelomonocytic cells. J Intern Med. 2014;276:154–70. doi: 10.1111/joim.12229. [DOI] [PubMed] [Google Scholar]
  • 97.Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41:49–61. doi: 10.1016/j.immuni.2014.06.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Jandus C, Bioley G, Speiser DE, Romero P. Selective accumulation of differentiated FOXP3(+) CD4 (+) T cells in metastatic tumor lesions from melanoma patients compared to peripheral blood. Cancer Immunol Immunother. 2008;57:1795–805. doi: 10.1007/s00262-008-0507-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ye J, Su X, Hsueh EC, Zhang Y, Koenig JM, Hoft DF, et al. Human tumor-infiltrating Th17 cells have the capacity to differentiate into IFN-gamma+ and FOXP3+ T cells with potent suppressive function. Eur J Immunol. 2011;41:936–51. doi: 10.1002/eji.201040682. [DOI] [PubMed] [Google Scholar]
  • 100.Noval Rivas M, Burton OT, Wise P, Charbonnier LM, Georgiev P, Oettgen HC, et al. Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity. 2015;42:512–23. doi: 10.1016/j.immuni.2015.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Oldenhove G, Bouladoux N, Wohlfert EA, Hall JA, Chou D, Dos Santos L, et al. Decrease of Foxp3(+) Treg cell number and acquisition of effector cell phenotype during lethal infection. Immunity. 2009;31:772–86. doi: 10.1016/j.immuni.2009.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Whibley N, Gaffen SL. Brothers in arms: Th17 and Treg responses in Candida albicans immunity. PLoS Pathog. 2014;10:e1004456. doi: 10.1371/journal.ppat.1004456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pandiyan P, Bhaskaran N, Zhang Y, Weinberg A. Isolation of T cells from mouse oral tissues. Biol Proced Online. 2014;16:4. doi: 10.1186/1480-9222-16-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Bhaskaran N, Weinberg A, Pandiyan P. Th17 inflammation model of Oropharyngeal candidiasis in immunodeficient mice. J Visual Experim: JoVE. 2015 doi: 10.3791/52538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Allan SE, Crome SQ, Crellin NK, Passerini L, Steiner TS, Bacchetta R, et al. Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int Immunol. 2007;19:345–54. doi: 10.1093/intimm/dxm014. [DOI] [PubMed] [Google Scholar]
  • 106.McMurchy AN, Levings MK. In vitro generation of human T regulatory cells: generation, culture, and analysis of FOXP3-transduced T cells. Methods Mol Biol. 2013;946:115–32. doi: 10.1007/978-1-62703-128-8_8. [DOI] [PubMed] [Google Scholar]
  • 107.Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30:899–911. doi: 10.1016/j.immuni.2009.03.019. [DOI] [PubMed] [Google Scholar]
  • 108.Cosmi L, De Palma R, Santarlasci V, Maggi L, Capone M, Frosali F, et al. Human interleukin 17-producing cells originate from a CD161+CD4+ T cell precursor. J Exp Med. 2008;205:1903–16. doi: 10.1084/jem.20080397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kleinewietfeld M, Hafler DA. The plasticity of human Treg and Th17 cells and its role in autoimmunity. Semin Immunol. 2013;25:305–12. doi: 10.1016/j.smim.2013.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Maggi L, Santarlasci V, Capone M, Peired A, Frosali F, Crome SQ, et al. CD161 is a marker of all human IL-17-producing T-cell subsets and is induced by RORC. Eur J Immunol. 2010;40:2174–81. doi: 10.1002/eji.200940257. [DOI] [PubMed] [Google Scholar]
  • 111.Leveque L, Deknuydt F, Bioley G, Old LJ, Matsuzaki J, Odunsi K, et al. Interleukin 2-mediated conversion of ovarian cancer-associated CD4+ regulatory T cells into proinflammatory interleukin 17-producing helper T cells. J Immunother. 2009;32:101–8. doi: 10.1097/CJI.0b013e318195b59e. [DOI] [PubMed] [Google Scholar]
  • 112.Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu BP, et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity. 2008;29:44–56. doi: 10.1016/j.immuni.2008.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Li L, Kim J, Boussiotis VA. IL-1beta-mediated signals preferentially drive conversion of regulatory T cells but not conventional T cells into IL-17-producing cells. J Immunol. 2010;185:4148–53. doi: 10.4049/jimmunol.1001536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Xu L, Kitani A, Fuss I, Strober W. Cutting edge: regulatory T cells induce CD4+CD25-Foxp3-T cells or are self-induced to become Th17 cells in the absence of exogenous TGF-beta. J Immunol. 2007;178:6725–9. doi: 10.4049/jimmunol.178.11.6725. [DOI] [PubMed] [Google Scholar]
  • 115.Osorio F, LeibundGut-Landmann S, Lochner M, Lahl K, Sparwasser T, Eberl G, et al. DC activated via dectin-1 convert Treg into IL-17 producers. Eur J Immunol. 2008;38:3274–81. doi: 10.1002/eji.200838950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Li J, Shi J, Ren W, Wu W, Chen Z. Regulatory role of CD4(+)CD25 (+)Foxp3 (+) regulatory T cells on IL-17-secreting T cells in chronic hepatitis B patients. Dig Dis Sci. 2014;59:1475–83. doi: 10.1007/s10620-013-3022-1. [DOI] [PubMed] [Google Scholar]
  • 117.Pandiyan P, Yang XP, Saravanamuthu SS, Zheng L, Ishihara S, O’Shea JJ, et al. The role of IL-15 in activating STAT5 and fine-tuning IL-17A production in CD4 T lymphocytes. J Immunol. 2012;189:4237–46. doi: 10.4049/jimmunol.1201476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, et al. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity. 2007;26:371–81. doi: 10.1016/j.immuni.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 119.Baban B, Chandler PR, Sharma MD, Pihkala J, Koni PA, Munn DH, et al. IDO activates regulatory T cells and blocks their conversion into Th17-like T cells. J Immunol. 2009;183:2475–83. doi: 10.4049/jimmunol.0900986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Sharma MD, Hou DY, Liu Y, Koni PA, Metz R, Chandler P, et al. Indoleamine 2,3-dioxygenase controls conversion of Foxp3+ Tregs to TH17-like cells in tumor-draining lymph nodes. Blood. 2009;113:6102–11. doi: 10.1182/blood-2008-12-195354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Sharma MD, Huang L, Choi JH, Lee EJ, Wilson JM, Lemos H, et al. An inherently bifunctional subset of Foxp3+ T helper cells is controlled by the transcription factor eos. Immunity. 2013;38:998–1012. doi: 10.1016/j.immuni.2013.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Nyirenda MH, Sanvito L, Darlington PJ, O’Brien K, Zhang GX, Constantinescu CS, et al. TLR2 stimulation drives human naive and effector regulatory T cells into a Th17-like phenotype with reduced suppressive function. J Immunol. 2011;187:2278–90. doi: 10.4049/jimmunol.1003715. [DOI] [PubMed] [Google Scholar]
  • 123.Ishii KJ, Koyama S, Nakagawa A, Coban C, Akira S. Host innate immune receptors and beyond: making sense of microbial infections. Cell Host Microbe. 2008;3:352–63. doi: 10.1016/j.chom.2008.05.003. [DOI] [PubMed] [Google Scholar]
  • 124.Uematsu S, Akira S. Toll-Like receptors (TLRs) and their ligands. Handb Exp Pharmacol. 2008:1–20. doi: 10.1007/978-3-540-72167-3_1. [DOI] [PubMed] [Google Scholar]
  • 125.Caramalho I, Lopes-Carvalho T, Ostler D, Zelenay S, Haury M, Demengeot J. Regulatory T cells selectively express toll-like receptors and are activated by lipopolysaccharide. J Exp Med. 2003;197:403–11. doi: 10.1084/jem.20021633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Gelman AE, Zhang J, Choi Y, Turka LA. Toll-like receptor ligands directly promote activated CD4+ T cell survival. J Immunol. 2004;172:6065–73. doi: 10.4049/jimmunol.172.10.6065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Komai-Koma M, Jones L, Ogg GS, Xu D, Liew FY. TLR2 is expressed on activated T cells as a costimulatory receptor. Proc Natl Acad Sci US A. 2004;101:3029–34. doi: 10.1073/pnas.0400171101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Liu H, Komai-Koma M, Xu D, Liew FY. Toll-like receptor 2 signaling modulates the functions of CD4+ CD25+ regulatory T cells. Proc Natl Acad Sci US A. 2006;103:7048–53. doi: 10.1073/pnas.0601554103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.van Maren WW, Nierkens S, Toonen LW, Bolscher JM, Sutmuller RP, Adema GJ. Multifaceted effects of synthetic TLR2 ligand and Legionella pneumophilia on Treg-mediated suppression of T cell activation. BMC Immunol. 2011;12:23. doi: 10.1186/1471-2172-12-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Oberg HH, Juricke M, Kabelitz D, Wesch D. Regulation of T cell activation by TLR ligands. Eur J Cell Biol. 2011;90:582–92. doi: 10.1016/j.ejcb.2010.11.012. [DOI] [PubMed] [Google Scholar]
  • 131.Jin B, Sun T, Yu XH, Yang YX, Yeo AE. The effects of TLR activation on T-cell development and differentiation. Clin Dev Immunol. 2012;2012:836485. doi: 10.1155/2012/836485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Hoffmann P, Boeld TJ, Eder R, Huehn J, Floess S, Wieczorek G, et al. Loss of FOXP3 expression in natural human CD4+CD25+ regulatory T cells upon repetitive in vitro stimulation. Eur J Immunol. 2009;39:1088–97. doi: 10.1002/eji.200838904. [DOI] [PubMed] [Google Scholar]
  • 133.LaRosa DF, Gelman AE, Rahman AH, Zhang J, Turka LA, Walsh PT. CpG DNA inhibits CD4+CD25+ Treg suppression through direct MyD88-dependent costimulation of effector CD4+ T cells. Immunol Lett. 2007;108:183–8. doi: 10.1016/j.imlet.2006.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Peng G, Guo Z, Kiniwa Y, Voo KS, Peng W, Fu T, et al. Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science. 2005;309:1380–4. doi: 10.1126/science.1113401. [DOI] [PubMed] [Google Scholar]
  • 135.Sutmuller R, Garritsen A, Adema GJ. Regulatory T cells and toll-like receptors: regulating the regulators. Ann Rheum Dis. 2007;66(Suppl 3):iii91–5. doi: 10.1136/ard.2007.078535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Xu D, Liu H, Komai-Koma M, Campbell C, McSharry C, Alexander J, et al. CD4+CD25+ regulatory T cells suppress differentiation and functions of Th1 and Th2 cells, Leishmania major infection, and colitis in mice. J Immunol. 2003;170:394–9. doi: 10.4049/jimmunol.170.1.394. [DOI] [PubMed] [Google Scholar]
  • 137.Netea MG, Sutmuller R, Hermann C, Van der Graaf CA, Van der Meer JW, van Krieken JH, et al. Toll-like receptor 2 suppresses immunity against Candida albicans through induction of IL-10 and regulatory T cells. J Immunol. 2004;172:3712–8. doi: 10.4049/jimmunol.172.6.3712. [DOI] [PubMed] [Google Scholar]
  • 138.Pasare C, Medzhitov R. Toll pathway-dependent blockade of CD4+CD25+ T cell-mediated suppression by dendritic cells. Science. 2003;299:1033–6. doi: 10.1126/science.1078231. [DOI] [PubMed] [Google Scholar]
  • 139.Komatsu N, Mariotti-Ferrandiz ME, Wang Y, Malissen B, Waldmann H, Hori S. Heterogeneity of natural Foxp3+ T cells: a committed regulatory T-cell lineage and an uncommitted minor population retaining plasticity. Proc Natl Acad Sci US A. 2009;106:1903–8. doi: 10.1073/pnas.0811556106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Sutmuller RP, den Brok MH, Kramer M, Bennink EJ, Toonen LW, Kullberg BJ, et al. Toll-like receptor 2 controls expansion and function of regulatory T cells. J Clin Invest. 2006;116:485–94. doi: 10.1172/JCI25439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Gratz IK, Rosenblum MD, Abbas AK. The life of regulatory T cells. Ann NY Acad Sci. 2013;1283:8–12. doi: 10.1111/nyas.12011. [DOI] [PubMed] [Google Scholar]
  • 142.Turka LA, Walsh PT. IL-2 signaling and CD4+ CD25+ Foxp3+ regulatory T cells. Front Biosci. 2008;13:1440–6. doi: 10.2741/2773. [DOI] [PubMed] [Google Scholar]
  • 143.Chen Q, Davidson TS, Huter EN, Shevach EM. Engagement of TLR2 does not reverse the suppressor function of mouse regulatory T cells, but promotes their survival. J Immunol. 2009;183:4458–66. doi: 10.4049/jimmunol.0901465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Crellin NK, Garcia RV, Hadisfar O, Allan SE, Steiner TS, Levings MK. Human CD4+ T cells express TLR5 and its ligand flagellin enhances the suppressive capacity and expression of FOXP3 in CD4+CD25+ T regulatory cells. J Immunol. 2005;175:8051–9. doi: 10.4049/jimmunol.175.12.8051. [DOI] [PubMed] [Google Scholar]
  • 145.Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, et al. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science. 2011;332:974–7. doi: 10.1126/science.1206095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Hall JA, Bouladoux N, Sun CM, Wohlfert EA, Blank RB, Zhu Q, et al. Commensal DNA limits regulatory T cell conversion and is a natural adjuvant of intestinal immune responses. Immunity. 2008;29:637–49. doi: 10.1016/j.immuni.2008.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Liu J, Radler D, Illi S, Klucker E, Turan E, von Mutius E, et al. TLR2 polymorphisms influence neonatal regulatory T cells depending on maternal atopy. Allergy. 2011;66:1020–9. doi: 10.1111/j.1398-9995.2011.02573.x. [DOI] [PubMed] [Google Scholar]
  • 148.Takeuchi O, Akira S. MyD88 as a bottle neck in Toll/IL-1 signaling. Curr Top Microbiol Immunol. 2002;270:155–67. doi: 10.1007/978-3-642-59430-4_10. [DOI] [PubMed] [Google Scholar]
  • 149.Reynolds JM, Pappu BP, Peng J, Martinez GJ, Zhang Y, Chung Y, et al. Toll-like receptor 2 signaling in CD4(+) T lymphocytes promotes T helper 17 responses and regulates the pathogenesis of autoimmune disease. Immunity. 2010;32:692–702. doi: 10.1016/j.immuni.2010.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Gil ML, Gozalbo D. Role of Toll-like receptors in systemic Candida albicans infections. Front Biosci. 2009;14:570–82. doi: 10.2741/3263. [DOI] [PubMed] [Google Scholar]
  • 151.Cho SN, Song CH, Jin J, Kim SH, Rha KS, Kim YM. Role of staphylococcal enterotoxin B on the differentiation of regulatory T cells in nasal polyposis. Am J Rhinol Allergy. 2014;28:e17–24. doi: 10.2500/ajra.2014.28.3995. [DOI] [PubMed] [Google Scholar]
  • 152.O’Shea JJ, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science. 2010;327:1098–102. doi: 10.1126/science.1178334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Zhu J, Paul WE. Peripheral CD4+ T-cell differentiation regulated by networks of cytokines and transcription factors. Immunol Rev. 2010;238:247–62. doi: 10.1111/j.1600-065X.2010.00951.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Ciofani M, Madar A, Galan C, Sellars M, Mace K, Pauli F, et al. A validated regulatory network for Th17 cell specification. Cell. 2012;151:289–303. doi: 10.1016/j.cell.2012.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Chen Q, Yang W, Gupta S, Biswas P, Smith P, Bhagat G, et al. IRF-4-binding protein inhibits interleukin-17 and interleukin-21 production by controlling the activity of IRF-4 transcription factor. Immunity. 2008;29:899–911. doi: 10.1016/j.immuni.2008.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Reinhardt RL, Liang HE, Locksley RM. Cytokine-secreting follicular T cells shape the antibody repertoire. Nat Immunol. 2009;10:385–93. doi: 10.1038/ni.1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Crotty S. Follicular helper CD4 T cells (TFH) Annu Rev Immunol. 2011;29:621–63. doi: 10.1146/annurev-immunol-031210-101400. [DOI] [PubMed] [Google Scholar]
  • 158.Qi H, Cannons JL, Klauschen F, Schwartzberg PL, Germain RN. SAP-controlled T-B cell interactions underlie germinal centre formation. Nature. 2008;455:764–9. doi: 10.1038/nature07345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Chung Y, Tanaka S, Chu F, Nurieva RI, Martinez GJ, Rawal S, et al. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat Med. 2011;17:983–8. doi: 10.1038/nm.2426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Linterman MA, Pierson W, Lee SK, Kallies A, Kawamoto S, Rayner TF, et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat Med. 2011;17:975–82. doi: 10.1038/nm.2425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Wollenberg I, Agua-Doce A, Hernandez A, Almeida C, Oliveira VG, Faro J, et al. Regulation of the germinal center reaction by Foxp3+ follicular regulatory T cells. J Immunol. 2011;187:4553–60. doi: 10.4049/jimmunol.1101328. [DOI] [PubMed] [Google Scholar]
  • 162.Lim HW, Hillsamer P, Banham AH, Kim CH. Cutting edge: direct suppression of B cells by CD4+ CD25+ regulatory T cells. J Immunol. 2005;175:4180–3. doi: 10.4049/jimmunol.175.7.4180. [DOI] [PubMed] [Google Scholar]
  • 163.Lim HW, Hillsamer P, Kim CH. Regulatory T cells can migrate to follicles upon T cell activation and suppress GC-Th cells and GC-Th cell-driven B cell responses. J Clin Invest. 2004;114:1640–9. doi: 10.1172/JCI22325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Wing JB, Sakaguchi S. Foxp3(+) T(reg) cells in humoral immunity. Int Immunol. 2014;26:61–9. doi: 10.1093/intimm/dxt060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Li L, Patsoukis N, Petkova V, Boussiotis VA. Runx1 and Runx3 are involved in the generation and function of highly suppressive IL-17-producing T regulatory cells. PLoS ONE. 2012;7:e45115. doi: 10.1371/journal.pone.0045115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Rudra D, deRoos P, Chaudhry A, Niec RE, Arvey A, Samstein RM, et al. Transcription factor Foxp3 and its protein partners form a complex regulatory network. Nat Immunol. 2012;13:1010–9. doi: 10.1038/ni.2402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos 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;198:1875–86. doi: 10.1084/jem.20030152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 cells. Annu Rev Immunol. 2009;27:485–517. doi: 10.1146/annurev.immunol.021908.132710. [DOI] [PubMed] [Google Scholar]
  • 169.McGeachy MJ, Cua DJ. T cells doing it for themselves: TGF-beta regulation of Th1 and Th17 cells. Immunity. 2007;26:547–9. doi: 10.1016/j.immuni.2007.05.003. [DOI] [PubMed] [Google Scholar]
  • 170.Zhou L, Lopes JE, Chong MM, Ivanov II, Min R, Victora GD, et al. TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature. 2008;453:236–40. doi: 10.1038/nature06878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Komatsu N, Okamoto K, Sawa S, Nakashima T, Oh-hora M, Kodama T, et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat Med. 2014;20:62–8. doi: 10.1038/nm.3432. [DOI] [PubMed] [Google Scholar]
  • 172.Chaudhry A, Rudensky AY. Control of inflammation by integration of environmental cues by regulatory T cells. J Clin Invest. 2013;123:939–44. doi: 10.1172/JCI57175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Chaudhry A, Rudra D, Treuting P, Samstein RM, Liang Y, Kas A, et al. CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science. 2009;326:986–91. doi: 10.1126/science.1172702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.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: 10.1038/ni.3053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Oldenhove G, Bouladoux N, Wohlfert EA, Hall JA, Chou D, Dos Santos L, et al. Decrease of Foxp3+ Treg cell number and acquisition of effector cell phenotype during lethal infection. Immunity. 2009;31:772–86. doi: 10.1016/j.immuni.2009.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Wohlfert EA, Grainger JR, Bouladoux N, Konkel JE, Oldenhove G, Ribeiro CH, et al. GATA3 controls Foxp3(+) regulatory T cell fate during inflammation in mice. J Clin Invest. 2011;121:4503–15. doi: 10.1172/JCI57456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Wang Y, Su MA, Wan YY. An essential role of the transcription factor GATA-3 for the function of regulatory T cells. Immunity. 2011;35:337–48. doi: 10.1016/j.immuni.2011.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Zhu J, Min B, Hu-Li J, Watson CJ, Grinberg A, Wang Q, et al. Conditional deletion of Gata3 shows its essential function in T(H)1-T(H)2 responses. Nat Immunol. 2004;5:1157–65. doi: 10.1038/ni1128. [DOI] [PubMed] [Google Scholar]
  • 179.Wan YSY, Flavell RA. Regulatory T-cell functions are subverted and converted owing to attenuated Foxp3 expression. Nature. 2007;445:766–70. doi: 10.1038/nature05479. [DOI] [PubMed] [Google Scholar]
  • 180.Wei G, Abraham BJ, Yagi R, Jothi R, Cui K, Sharma S, et al. Genome-wide analyses of transcription factor GATA3-mediated gene regulation in distinct T cell types. Immunity. 2011;35:299–311. doi: 10.1016/j.immuni.2011.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Zhu J, Paul WE. Heterogeneity and plasticity of T helper cells. Cell Res. 2010;20:4–12. doi: 10.1038/cr.2009.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Hegazy AN, Peine M, Helmstetter C, Panse I, Frohlich A, Bergthaler A, 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;32:116–28. doi: 10.1016/j.immuni.2009.12.004. [DOI] [PubMed] [Google Scholar]
  • 183.Yagi R, Junttila IS, Wei G, Urban JF, Jr, Zhao K, Paul WE, et al. The transcription factor GATA3 actively represses RUNX3 protein-regulated production of interferon-gamma. Immunity. 2010;32:507–17. doi: 10.1016/j.immuni.2010.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Mosmann TR, Coffman RL. Heterogeneity of cytokine secretion patterns and functions of helper T cells. Adv Immunol. 1989;46:111–47. doi: 10.1016/s0065-2776(08)60652-5. [DOI] [PubMed] [Google Scholar]
  • 185.Mosmann TR, Coffman RL. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol. 1989;7:145–73. doi: 10.1146/annurev.iy.07.040189.001045. [DOI] [PubMed] [Google Scholar]
  • 186.Paul WE, Seder RA. Lymphocyte responses and cytokines. Cell. 1994;76:241–51. doi: 10.1016/0092-8674(94)90332-8. [DOI] [PubMed] [Google Scholar]
  • 187.Seder RA, Paul WE. Acquisition of lymphokine-producing phenotype by CD4+ T cells. Annu Rev Immunol. 1994;12:635–73. doi: 10.1146/annurev.iy.12.040194.003223. [DOI] [PubMed] [Google Scholar]
  • 188.Murphy TL, Tussiwand R, Murphy KM. Specificity through cooperation: BATF–IRF interactions control immune-regulatory networks. Nat Rev. 2013;13:499–509. doi: 10.1038/nri3470. [DOI] [PubMed] [Google Scholar]
  • 189.Thornton AM, Shevach EM. CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med. 1998;188:287–96. doi: 10.1084/jem.188.2.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Hu G, Tang Q, Sharma S, Yu F, Escobar TM, Muljo SA, et al. Expression and regulation of intergenic long noncoding RNAs during T cell development and differentiation. Nat Immunol. 2013;14:1190–8. doi: 10.1038/ni.2712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Levine AG, Arvey A, Jin W, Rudensky AY. Continuous requirement for the TCR in regulatory T cell function. Nat Immunol. 2014;15:1070–8. doi: 10.1038/ni.3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Vahl JC, Drees C, Heger K, Heink S, Fischer JC, Nedjic J, et al. Continuous T cell receptor signals maintain a functional regulatory T cell pool. Immunity. 2014;41:722–36. doi: 10.1016/j.immuni.2014.10.012. [DOI] [PubMed] [Google Scholar]
  • 193.Zheng Y, Chaudhry A, Kas A, deRoos P, Kim JM, Chu TT, et al. Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature. 2009;458:351–6. doi: 10.1038/nature07674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Zhou X, Bailey-Bucktrout SL, Jeker LT, Penaranda C, Martinez-Llordella M, Ashby M, et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol. 2009;10:1000–7. doi: 10.1038/ni.1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Korn T, Anderson AC, Bettelli E, Oukka M. The dynamics of effector T cells and Foxp3+ regulatory T cells in the promotion and regulation of autoimmune encephalomyelitis. J Neuroimmunol. 2007;191:51–60. doi: 10.1016/j.jneuroim.2007.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Lanteri MC, O’Brien KM, Purtha WE, Cameron MJ, Lund JM, Owen RE, et al. Tregs control the development of symptomatic West Nile virus infection in humans and mice. J Clin Invest. 2009;119:3266–77. doi: 10.1172/JCI39387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Chen Y, Haines CJ, Gutcher I, Hochweller K, Blumenschein WM, McClanahan T, et al. Foxp3(+) regulatory T cells promote T helper 17 cell development in vivo through regulation of interleukin-2. Immunity. 2011;34:409–21. doi: 10.1016/j.immuni.2011.02.011. [DOI] [PubMed] [Google Scholar]
  • 198.Pandiyan P, Gartner D, Soezeri O, Radbruch A, Schulze-Osthoff K, Brunner-Weinzierl MC. CD152 (CTLA-4) determines the unequal resistance of Th1 and Th2 cells against activation-induced cell death by a mechanism requiring PI3 kinase function. J Exp Med. 2004;199:831–42. doi: 10.1084/jem.20031058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Zhou Y. Regulatory T cells and viral infections. Front Biosci. 2008;13:1152–70. doi: 10.2741/2752. [DOI] [PubMed] [Google Scholar]
  • 200.Hernandez-Santos N, Huppler AR, Peterson AC, Khader SA, McKenna KC, Gaffen SL. Th17 cells confer long-term adaptive immunity to oral mucosal Candida albicans infections. Mucosal Immunol. 2013;6:900–10. doi: 10.1038/mi.2012.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Chaudhry A, Rudra D, Treuting P, Samstein RM, Liang Y, Kas A, et al. CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science (New York, NY) 2009 doi: 10.1126/science.1172702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Rosenblum MD, Gratz IK, Paw JS, Lee K, Marshak-Rothstein A, Abbas AK. Response to self antigen imprints regulatory memory in tissues. Nature. 2011;480:538–42. doi: 10.1038/nature10664. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES