Open access

Balancing pro- and anti-inflammatory CD4+ T helper cells in the intestine

Written By

Nicola Gagliani and Samuel Huber

Submitted: 11 November 2011 Published: 25 July 2012

DOI: 10.5772/48183

Chapter metrics overview

2,577 Chapter Downloads

View Full Metrics

1. Introduction

The intestinal mucosal surface represents a huge border where different pathogenic particles, such as bacteria, fungi, viruses or parasites can potentially invade and harm the body. One crucial task of the immune system in the intestine is to maintain this epithelial barrier, in order to prohibit or defeat a microbial invasion. Pro-inflammatory effector CD4+ T helper cells play a crucial role during this task. These effector T helper cells can be subdivided into different subsets (Figure 1), which are characterized by a master transcriptional regulator and a unique cytokine profile: Th17 cells express RORγt that in turn promotes the transcription of Il17a, Il17f, Il21 and Il22. Th1 cells express T-bet and produce IFN-γ, IL-2 and TNF-α. Th2 cells express GATA-3 and secrete IL-4, IL-5 and IL-13 [1-5]. The intestine also contains numerous non-pathogenic bacteria (commensal bacteria), which are beneficial to the host, as well as food antigens. This vast collection of non-self antigens can also promote the activation of T helper cells, and in turn cause immune-pathology. Therefore it is important for the immune system to control effector T helper cells. Indeed different types of regulatory T cells with anti-inflammatory properties team up in order to control effector T cells. The two most studied regulatory T cell subtypes are Foxp3+ regulatory T cells, which can be generated either in the thymus (nTreg) or induced in peripheral lymphoid organs (iTregs) and type 1 regulatory T cells (Tr1), which are induced in the periphery (Figure 1).

Advertisement

2. Differentiation of naïve CD4+ T cells into effector T helper cells

Naïve T cells, which are functionally immature, can be differentiated into different subsets of effector T cells upon activation. The fate of naïve T cell is directed by cytokines. These cytokines signal via different members of the STAT family, which induce master transcriptional regulators. Most of these transcriptional factors bind then to the effector cytokine gene thereby inducing gene activation, repression or epigenetic modification [6] (Figure 1). It should be noted that there is a certain amount of T helper cell heterogeneity and plasticity regarding cytokine production and expression of the master transcriptional factor of each T helper cell subset. This fact is currently also one of the most intriguing aspect of the ongoing research in Immunobiology. However, the model of different T helper cell lineages as first proposed by Mosmann and Coffman [7] is still the most useful one in order to understand the function and differentiation of T helper cells.

Figure 1.

Differentiation of naïve T cells into different effector and regulatory T cells. A specific combination of cytokine signals leads to the differentiation of naïve T cells into different T helper cell subsets. Each T helper cell subset is characterized by the production of a combination of cytokines and exerts specific functions.

2.1. Differentiation and function of Th1 cells

Th1 cells produce IFN-γ as their signature cytokine. Th1 cells secrete also IL-2 and/ or TNF-α. Naive T cells upon TCR stimulation in the presence of IL-12 differentiate in Th1 cells [8]. IL-12 signals via STAT4 promoting the expression of T-bet, which transcribes the Ifng gene [9, 10]. T-bet is the master transcriptional regulator of Th1 cells, which is essential for the IFN-γ production [5]. Accordingly T-bet deficient mice have a defective Th1 differentiation [11]. One other important function of T-bet is the inhibition of GATA-3 expression, the master transcriptional regulator of Th2 cells [9].

Th1 cells are particularly important for the defense against intra-cellular bacteria. Some microorganisms such as mycobacteria, like Mycobacterium tuberculosis or Mycobacterium lepromatosis, are examples for these intracellular pathogens. These bacteria grow primarily in phagolysosomes of macrophages. Because of this feature these microorganisms are protected from the effects of antibodies and cytotoxic T cells. These bacteria can inhibit the fusion of lysosomes to the phagosomes, in which they grow and prevent the activation of the lysosomal proteases. The defense against these microorganisms is the important task of Th1 effector cells because they can activate macrophages, which are then able to kill ingested pathogens. Accordingly, deficiency in Th1 cells increases the susceptibility to infections with intracellular pathogens in humans [12]. These patients suffer from infections with mycobacteria, particularly Mycobacterium tuberculosis, but also with Salmonella. Of note both of these bacteria strains can typically infect the gastrointestinal system.

2.2. Differentiation and function of Th2 cells

The signature cytokines of Th2 cells are IL-4, IL-5, and IL-13. Some Th2 cells also produce TNF-α and/ or IL-9. Additionally, some Th2 cells can secrete small amounts of IL-2. The cytokines leading to Th2 differentiation are IL-2 and IL-4. Therefore the signature cytokine of Th2 cells, IL-4 also promotes the differentiation of Th2 cells [13-15]. STAT6 is the major signaling pathway of IL-4 mediated Th2 differentiation, and induces GATA-3 expression [16-20]. GATA-3 is the master transcriptional regulator of Th2 cells [3, 21] and the differentiation of these cells is indeed dependent on the induction of this master transcriptional regulator [22]. GATA-3 binds to the promoters of Il5 and Il13, and the enhancer of Il4 thereby promoting their transcription [6]. Additionally STAT5, which can be activated by IL-2, is important for Th2 differentiation and for the maintenance of GATA-3 expression [23].

Th2 cells and their effector cytokines IL-4, IL-5, and IL-13 are essential to control helminth infections in the intestine. In line with this, mice deficient in IL-4 receptor a-chain (IL-4Rα), STAT6 or GATA-3 show highly compromised anti-helminth immunity [24]. One of the most unique tasks of Th2 cells is the induction of B-cell immunoglobulin class switching. Through CD40-CD40L interaction, Th2 cells promote B cells to secret IgG1, IgE and (in humans) IgG4 isotype antibodies. These antibodies are again important for mediating protection against helminth infections. The Th2-immune response involves also eosinophils, basophils and mast cells, which all together mount the immune response controlling helminth infection. The release of IL-4 and IL-13 is key for eliciting the alternative activation of macrophage, which is crucial in order to trap the intestinal parasite [25, 26]. Th2-cytokines, in particular IL-4 and IL-13, promote the goblet cell differentiation, the enhancement of mucus secretion and the production of resistin-like molecule-β (RELMβ), an innate protein with direct anti-helminth activity [27-29]. Moreover IL-4 stimulates intestinal muscle hyper-contractility and accelerates epithelial turnover to promote the ‘epithelial escalator’, which functions together with epithelial secretions to dislodge resident parasites [30, 31]. Another Th2 associated cytokine, namely IL-9, promotes the release of mast cell protease that can depredate tight junctions and in turn increase the fluid flow in the intestine. All together these mechanisms are part of the “weep and sweep” response, which is key for the control of a helminth invasion.

2.3. Differentiation and function of Th17 cells

The signature cytokines of Th17 cells are Il-17A and IL-17F. Th17 cells produce also, IL-22 and TNF-α. TGF-β, IL-1β, IL-6, and IL-23 are the cytokines, which are important for Th17 cell differentiation. IL-6 can activate STAT3, which induces IL-23R and RORγt [32-34], the master transcriptional regulator of Th17 cells. This master transcriptional regulator leads to the production of IL-17A and IL-17F [1, 4, 35, 36]. IL-6 also promotes the release of IL-21 [33], which synergizes with TGF-β, IL-6, and IL-1β, for the induction of IL-23 receptor expression [37]. In the presence of IL-23, CD4+ RORγt+ IL-17A+ T cells can expand and fully mature in Th17 cells [38, 39].

Human and mouse Th17 cells are rare in a non-pathological state [2, 40]. A specific member of commensal microbiota, known as segmented filamentous bacteria (SFB), attracts Th17 cells in the terminal ileum of mice [41]. Therefore in steady state condition most of the few Th17 cells accumulate mainly in the intestine. The commensal microbiota promotes the release of serum amyloid A [41] and adenosine 5′-triphosphate (ATP), which activates lamima propria mononuclear phagocytes. These phagocytes in turn promote Th17 cell differentiation [42]. Among all cytokines known to induce the differentiation of Th17 cells, the presence of IL-1β rather than IL-6 is essential in the intestine [43]. TGF- β 1 is also not essential for the differentiation of Th17 cells in the intestine, but may influence the phenotype of Th17 cells together with IL-1β [44]. Th17 cells, which have been differentiated in the presence of TGF-β1 are less pathogenic and produce more IL-10 compared to Th17 cell differentiated in the presence of IL-1β [45, 46].

Th17 cells also produce several other cytokines besides IL-17A and IL-17F. Cytokine production by Th17 cells is also modulated by environmental factors in the intestine. For example, the activation of the environmental chemical receptor and transcription factor aryl hydrocarbon receptor in Th17 cells is important for the production of IL-22 [47-49]. IL-22 is a critical cytokine for antimicrobial immunity exerted by Th17 cells [50]. On the other hand, the induction of c-maf upon stimulation with IL-27, promotes the release of IL-10 from Th17 cells [51], and these IL-10 producing Th17 cells are also particularly induced in the intestine [40]. Therefore Th17 cells can have different cytokine profiles depending on environment factors.

In the absence of pathology, Th17 cells are very rare. However pathogenic infections, such as fungi infection with Candida albicans, or bacterial infection with gram positive or gram negative extracellular bacteria, such as Citrobacter rodentium or Klebsiella pneumoniae lead to a dramatic increase of the number of Th17 cells [52-56]. Viral infection also promotes a Th17-cell mediated immune response [40]. In line with this, Th17 cells and their effector cytokines IL-17A, IL-17F, and IL-22 are critical for proper host defense against various infections, especially against extracellular bacteria and fungi. The receptors for IL-17A, IL-17F and IL-22 are broadly expressed throughout the intestinal epithelial tissue. Therefore Th17 cells can provide crosstalk between immune system and tissues [2, 57].

IL-17A and IL-17F strongly induce the recruitment of neutrophils to the inflammatory site. The subsequent induction of the chemokine CCL20 attracts even more Th17 cells via CCR6, the chemokine receptor of CCL20, which is highly expressed by Th17 cells [58]. Additionally, both IL-17A and IL-17F promote β-defensin production [56, 59, 60]. β-defensins play an important role in the immune responses against bacterial infections. Interestingly, IL-17A and IL-17F can compensate each other during the host defense against S. aureus [56]. However during other infections, such as Citrobacter rodentium, IL-17A and IL-17F are both required in order to control the bacterial dissemination [56].

At the mucosal surface IL-22 has a crucial function for host defence and tissue homeostasis. IL-22 induces the expression of antimicrobial peptides from epithelial cells and limits bacterial replication and dissemination during Citrobacter rodentium infection [57, 61]. Furthermore IL-22 can promote epithelial cell proliferation, survival, and tissue repair in the intestine [62-64].

However it should be noted that several other immune cells besides Th17 cells can produce IL-17A and IL-22, thereby also contributing to the defense against pathogens (for review see [50]).

2.4. Differentiation and function of Foxp3+ Treg cells

In 1995 Sakaguchi et al. first described a subpopulation of regulatory T cells characterized by the constitutive expression of the IL-2 receptor α-chain (CD25). These regulatory T cells were called CD4+CD25+ Treg [65]. Foxp3 was identified later on as the master transcriptional regulator of CD4+CD25+ Treg cells, which have been called Foxp3+ Treg cells thereafter [66, 67]. Foxp3+ Treg cells can be generated within the thymus (tTreg) [65]. However, Foxp3+ Treg cell numbers are also regulated in peripheral lymphoid organs both by expansion of pre-existing Foxp3+ Treg and by de novo generation of induced regulatory T cells (iTreg). The combination of the cytokines IL-2 and TGF-β1 are key for the differentiation of naïve T cells into Foxp3+ iTreg cells [68-73]. Foxp3+ Treg cells are essential to control auto-reactive T cells, which can react to self-antigens and cause damage to the host. The key role of Foxp3+ Treg cells in the peripheral immune response is evident in murine models [74] and in humans [75]: scurfy mice [74] and IPEX (immunodysregulation polyendocrinopathy enteropathy X-linked syndrome) patients [75] lacking the master transcriptional factor of regulatory T cells - Foxp3 - consequently develop strong autoimmune disorders. Importantly, a severe form of autoimmune enteropathy is characteristic for scurfy mice and IPEX patients [75]. This underlines the importance of Foxp3+ Treg cells for controlling the immune response in the intestine. Foxp3+ Treg cells have different mechanism to suppress effector T cells. Some of these are mediated via soluble factors (i.e. IL-10, TGF-β1 [76, 77]) and others are cell contact dependent (i.e. CTLA-4, cAMP [78, 79]). Recent studies have demonstrated that Foxp3+ Treg cells can acquire some features of effector T helper cells in order to better control them (see paragraph 4.3).

2.5. Differentiation and function of Tr1 cells

In 1994, T regulatory type 1 (Tr1) cells were isolated from severe combined immuno deficiency (SCID) patients transplanted with allogeneic haematopoietic stem cells (HSCT). Subsequently it was possible to test the regulatory capacity of this new type of T cells directly in murine IBD models. To date Tr1 cells lack a defined cell surface signature, and their identification relays therefore on their unique cytokine profile. Tr1 cells secrete high levels of IL-10 as compared to IL-4 and IL-17A, the hallmark cytokines of Th2 and Th17 cells respectively. Depending on the milieu Tr1 cells can produce variable levels of IFN-γ, the key cytokine produced by Th1 cells [80]. However, Tr1 cells posses the capacity to suppress inflammatory T cell responses and, therefore are distinct from bona fide Th1, Th2 and Th17 cells that largely promote rather than suppress the inflammatory responses.

Tr1 cells are induced in the periphery, and they respond selectively to persistent foreign and self-antigens under steady-state conditions [81].

After the discovery of Foxp3 as the master transcriptional regulator of Foxp3+ Treg cells, it became a key point, if also Tr1 cells express a master regulator. The double reporter mouse model for IL-10 and Foxp3 was instrumental in order to demonstrate that these two types of regulatory T cells are distinct. Indeed, Tr1 cells do not constitutively express Foxp3 [82, 83] and can be induced from IPEX patients who lack Foxp3 [84]. However the master transcriptional regulator for Tr1 cells has not been identified so far.

IL-10 has been considered to be the driving force for Tr1-cell generation on the basis of experiments in which antigen-specific Tr1 cells are induced in vitro by repeated TCR stimulation in the presence of high doses of IL-10 [85, 86]. However, the frequency of Tr1 cells in IL-10 deficient mice is not altered. Several recent publications have demonstrated a key role of IL-27, which can even synergize with TGF-β, in the induction of Tr1 cells. During the induction of Tr1 cells by IL-27, the ligand-activated transcription factor hydrocarbon receptor (AhR) physically associates with c-avian musculoaponeurotic fibrosarcoma (c-Maf) and transactivates the Il10 and Il21 promoters. The secretion of IL-21 acts as an autocrine growth factor for Tr1 cells (Reviewed in [87]).

Tr1 cells can control Th1, Th2 and Th17 cell, and regulate immune responses mainly through the secretion of the immunosuppressive cytokines IL-10 and TGF-β1 [88]. The antigen-specific activation of Tr1 cells is important to potentiate their regulatory function [86]. IL-10 acts by limiting the magnitude of immune responses, as proved by mice that lack IL-10 and that exhibit spontaneous enterocolitis. IL-10 down-regulates the expression of co-stimulatory molecules, such as CD80, CD86, and MHC Class II, and pro-inflammatory cytokine production by APCs and inhibits the secretion of IL-2, TNF-α and IL-17 by effector T cells [89]. In particular, Tr1-cell supernatant diminishes the capacity of monocytes to stimulate Th1-cell responses and blocks the differentiation and maturation of DCs via IL-10 [90]. TGF-β down-regulates the functions of APCs [91] and inhibits the proliferation and cytokine production by T cells [92]. Therefore, the suppressive effects of Tr1 cells are reversed by the addition of anti-IL10 and anti-TGF-β neutralizing antibodies [85, 93, 94], but additional mechanisms may also contribute. Human Tr1 cells generated in vitro by crosslinking CD3 with CD46 can kill target cells through a granzyme B/ perforin dependent mechanism [95] [96]. Accordingly human Tr1 cells selectively kill myeloid cells (i.e., DC and monocytes) through granzyme B/ perforin [97]. This selective cell-killing is mediated by CD226, which is expressed on Tr1 cells. Only myeloid cells express the CD226-ligand (CD155). Thus, this type of regulatory mechanism by Tr1 cells requires a cell-cell contact with APCs.

Advertisement

3. The immune homeostasis in the intestine

The immune system has to respond selectively to harmful non-self pathogens and at the same time needs to minimize reactions against self and not-harmful antigens. This highly fine-tuned mechanism is possible due to a strict selection process, which happens in the thymus. Potentially auto-reactive CD4+ T cell progenitors, which recognize self-antigens with their T cell receptor (TCR), are either deleted or converted into thymic-derived CD4+ regulatory T cells (tTreg) with anti-inflammatory properties. This process, called central tolerance, is essential for the education of CD4+ T cells to respond selectively against foreign antigens. However this thymic control appears still to be insufficient. Therefore the immune system developed several other mechanisms to control potentially auto-reactive T cells, which take places in the periphery (peripheral tolerance). Among these mechanisms, the action of CD4+ regulatory T cells, which can be either selected in the thymus, (tTreg) or induced in the periphery (iTreg)[98], is one of the most studied. Treg cells are essential to control auto-reactive T cells, which can react to self-antigens and cause damage to the host.

The intestine is not only a source of self-antigens, but also contains a vast collection of non-self antigens, such as commensal bacteria. These antigens can promote the activation of naïve T cells causing immune-pathology such as inflammatory bowel disease (IBD). Therefore the immune system has established a second checkpoint in the intestine where naïve T cells, which are potentially able to respond to non self-antigens, are educated to be tolerant. There are important differences between thymus and intestine in tolerance induction. The driving force for the selection in the thymus is the affinity of TCRs to MHC, while the flora and cytokines are crucial to determine the fate of naïve T cells in the intestine. Accordingly different commensal bacteria can selectively drive a tolerogenic or pro-infammatory response. In line with this, the bacterial composition of the intestine has a substantial impact on the balance between pro-inflammatory and regulatory T cells in the intestine, and can also affect other organ specific diseases [41, 99-101]: For example mice lacking an innate sensor, which controls the intestinal micro-flora, are more susceptible to develop colitis [100]. Another interaction between the gut flora and an autoimmune disease was found in EAE (experimental autoimmune encephalomyelitis, a mouse model for multiple sclerosis). Multiple sclerosis is caused by an attack by auto-reactive T cells against brain white matter. Interestingly it was shown that the commensal gut flora can trigger these auto-reactive T cells, which then drive the disease. Finally it is known that the bacterial colonization between neonates born vaginally or by cesarean delivery differs, and interestingly these differences have been linked to an increased risk for atopic diseases such as allergic rhinitis and asthma in children born by cesarean delivery [102].

Considering the amount of self- and none self-antigens present in this organ and with it the potential to generate an unwanted immune response, different players are required to control the immune homeostasis in the intestine. The first one is a specialized subset of DC, which through the release of TGF-β and retinoic acid, is able to induce iTreg cells [103]. These iTreg cells represent then the second players. It is also known that naïve T cells migrate to the intestine in order to acquire an iTreg cell phenotype [104, 105]. Interestingly, these iTreg cells have a TCR repertoire, which is specific to an individuals microflora. Based on these results, one could hypothesize that iTreg cells have an advantage over tTreg cells (3th player), which are also present in the intestine, but are obviously non-bacteria specific. However, it was shown that specific commensal bacteria can directly activate tTreg cells bypassing the antigen specificity [106]. Both tTreg and iTreg are able to suppress effector T cells in the intestine, thereby curing or preventing colitis development [68, 98, 107-110]. It seems that tTregs and iTreg can also supplement the function of each other partially by expanding the TCR diversity [111]. Tr1 cells (4th players) are expanded in the absence of iTregs, and can at least partially compensate the absence of iTregs [112, 113]. Consistent with this, Tr1 cells and Treg cells can compensate each other to suppress effector T cells in the intestine [114].

In conclusion, commensal antigens in the intestine play an essential role in the regulation of the immune homeostasis. Naïve T cells, which could be potentially auto reactive, are converted into different types of regulatory T cells, which in turn control other effector T helper cells. The regulatory T cells originated in the thymus (nTregs) also participate in this regulatory environment by expanding the antigen specificity of the immune response.

Advertisement

4. Breakdown of the immune homeostasis in the intestine

Imbalance between pro- and anti-inflammatory T helper cells can cause intestinal pathology, such as IBD in humans. Crohn's disease (CD) and ulcerative colitis (UC) are the two main forms of IBD. CD can attack any part of the digestive tract. It typically manifests in the ileum, although it can also selectively affect the large intestine. Histological Crohn's disease shows a transmural inflammation. This inflammation is characterized by focal infiltration of neutrophils into the epithelium. These neutrophils, along with mononuclear cells, can infiltrate the crypts, leading to inflammation or abscess. Granulomas, aggregates of macrophage derivatives, known as giant cells, are found in CD and are specific for the disease. Ulceration can also be seen in highly active CD. On the other hand, UC is a disease mainly of the colon that includes ulcerations. UC normally begins in the rectum, and can continuously affect the whole colon and also the terminal ileum. The pathology in ulcerative colitis involves distortion of crypt architecture, inflammation of crypts and hemorrhage. The inflammation is more superficial compared to CD and affects the mucosa and submucosa.

The aetiology of IBD is still unknown, but it seems that genetic and environmental factors contribute to disease development. Initial studies suggested that CD and UC are mediated by Th1 and Th2 cells respectively. This was based on the cytokine profile seen in CD (IL-12 and IFN-γ) and UC (IL-5, IL-13) [115]. However more recent work has shown that Th17 cells also infiltrate the intestine in CD and UC patients as well [116-118]. Accordingly the signature cytokines of Th17 cells (IL-17A, IL-17F, IL-22) are produced in the intestine of CD and UC patients [116, 118-120]. Additionally, genome wide association studies have linked polymorphism in Th17-related genes, such as IL-23R and STAT3 with IBD [121-124]. In line with these associations murine studies have also shown that Th17 cells are involved in numerous autoimmune and chronic inflammatory diseases [2], and IBD is one of these diseases [125]: RORγt deficient mice, which lack Th17 cells, exhibit attenuated experimentally induced autoimmune disease [4]. Adoptive transfer of in vitro or in vivo differentiated Th17 cells into lymphopenic hosts leads to the development of colitis [114, 126-128]. IL-23, which is important for the maintenance, expansion and pathogenicity of Th17 cells [38, 39], is essential for the induction of colitis in mouse models. All together, these data argue for an important role of Th17 cells in IBD. However, Th17 cells produce several factors. And it is currently not completely understood, which of these is/are responsible for the pathogenicity of Th17 cells in the intestine [129-132].

One key feature of Th17 cells is their plasticity, which might also contribute to the pathogenic potential of Th17 cells. Epigenetic studies have shown that Th17 cells are more plastic compared to Th1 and Th2 cells [56, 133-135]. Th17 cells have bivalent domains of histone modifications in the Tbx21 locus, which encodes for T-bet, the key transcriptional factor for Th1 cells. On the contrary, Th1 cells have only repressive markers in both Rorc and Il17a loci. These differences might account for the higher plasticity of Th17 cells relative to Th1 cells [133]. In line with these data, CD4+ T cells, which express both the key transcriptional factors and cytokines of Th17 and Th1 cells, have been found in the colon of mouse colitis models and moreover in colon of human IBD patients. They are also suggested to play an important role for the development of chronic disease [39, 114, 118].

Human IBD is characterized by a mixture of effector T cells. Therefore it is difficult to assess the relative contribution of a specific T helper cell subset in patients. However there are mouse IBD models, which are dominated by one specific T helper cells subset: Selective deficiency in iTreg cells causes Th2 dominated intestinal pathology, which is characterized by gastritis and plasmacytic enteritis with increased frequencies of plasma cells in the intestinal lamina propria [113]. Another mouse model of colitis, which is induced by the transfer of naïve T cells into a lymphopenic host, is dominated by Th1 cells. This colitis model is characterized by IFN-γ dependent mucosal ulceration in the colon [136, 137]. Th17 cell-dominated intestinal pathology is characterized by mucosal hyperplasia but not ulceration [76, 136, 138]. IL-22, a signature cytokine of Th17 cells, can promote epithelial cell survival and proliferation. It is also important for the repair of the intestinal mucosa [63, 136, 139]. Accordingly, IL-22 induces the hyperplasia in the Th17 dominated colitis models [136]. On the contrary IL-22 is beneficial in Th1 dominated colitis models, which are characterized by ulceration [62]. Of note, the histomorphology in these Th1, Th2, or Th17 cell dominated mouse IBD models features only some characteristics of human IBD. But still these models are useful to evaluate the function of specific T helper cell subsets.

Advertisement

5. Control of pro-inflammatory T helper cells in the intestine

There are three ways to control effector T cells. First, inhibition of the differentiation of naïve T cells into effector T cells. Second, endogenous mechanism limiting the pathogenic potential of effector T cells. Third, control of effector T cells through regulatory T cells.

5.1. Inhibition of the de novo differentiation of effector T cells

One possibility is to inhibit the de novo differentiation of naïve T cells into effector T cells and generate regulatory T cells in stead. Such a reciprocal development pathway has been described for Th17 cells: High concentrations of TGF-β and/or retinoic acid up-regulate Foxp3 [140, 141], which in turn inhibits the induction of RORγt [37], thereby preventing the differentiation of Th17 cells. Moreover, IL-2 together with TGF-β1 promotes the induction of Foxp3+ regulatory T cells (iTregs) instead of Th17 cells [142]. Interestingly, IL-2 blocks Th17 cell differentiation by directly inhibiting Il17a transcription. This second mechanism is largely independent of Foxp3 or RORγt expression, but dependent on the induction of STAT5, which competes with STAT3 for the common sites across the locus encoding IL-17A [143]. Finally, IL-27 through the activation and interaction of AhR and c-maf promotes the induction of type 1 regulatory T cells (Tr1) and efficiently counteracts the effects of TGF-β and IL-6 on CD4+ T cells, resulting in the inhibition of Th17 development in a STAT1-dependent manner (Reviewed in [144]).

5.2. Endogenous control of effector T cells

All effector T cell subsets (Th1, Th2, Th17) have the ability to acquire IL-10 production, thereby limiting their own pathogenicity [145]. This mechanism of self control has been very well described for Th17 cells (Figure 2): During particular bacterial and viral infection, naïve T cells maturate in effector Th17 cells and contribute to the eradication of infections. However, if the Th17 response is too strong and potentially life threatening, Th17 cells are redirected to the small intestine in order to be controlled [40]. The reason why mature Th17 cells migrate mainly to the small intestine is because of the high expression of the chemokine receptor CCR6 [146]. The highest concentration of CCL20, the ligand of CCR6, is indeed in the small intestine [40]. Interestingly, IL-17A and IL-17F promote the release of CCL20 from epithelial cells in the duodenum. The recruited Th17 cells also produce CCL20, furthermore amplifying CCL20 production. This suggests that Th17 cells implement through a positive feedback loop the recruitment of other Th17 cells to the small intestine. Once effector Th17 cells migrated to the intestine, two complementary mechanisms occur in order to control them. First, effector Th17 cells are washed out and eliminated via the intestinal lumen due to the strong tissue destruction and diarrhoea. Secondly, Th17 cells are reprogrammed in regulatory Th17 (rTh17) cells. This last mechanism relays on the plasticity of these cells. In the intestine, effector Th17 cells acquire the capacity to produce IL-10 [40] and in parallel express IL-10Rα. If Th17 cells cannot respond to IL-10, they acquire a “promiscuous” phenotype co-expressing IFN-γ and promote the inflammation in the small intestine [114].

Figure 2.

Endogenous control of Th17 cells in the intestine. A strong Th17 response leads to the redirection of effector Th17 (eTh17) cells to the small intestine. Once eTh17 cells migrated to the intestine, two complementary mechanisms occur in order to control them. First, eTh17 cells are washed out and eliminated via the intestinal lumen due to the strong tissue destruction and diarrhoea. Secondly, Th17 cells are reprogrammed in regulatory Th17 (rTh17) cells.

5.3. Exogenous control of effector T cells via regulatory T cells

Importantly, other control mechanisms, which do not rely on the “sense of responsibility” of effector T helper cells, are also present (Figure 3). Regulatory T cells play an essential role for controlling T helper cells. The two most studied regulatory T cell subsets are Foxp3+ Treg and Tr1 cells. Foxp3+ Tregs can be induced in the periphery (Foxp3+ iTregs) or in the thymus (Foxp3+ tTregs). Interestingly, Foxp3+ iTregs are induced in the intestine by TCR recognition of commensal antigens [101, 104, 105, 147]. Foxp3+ tTregs cells are obviously non-bacteria specific, but nevertheless can be activated by some bacterial species in the intestine [106]. It is also important that Foxp3+ iTregs and Foxp3+ tTregs perform complementary functions, in part by expanding the TCR diversity [111].

Figure 3.

Control of effector T helper cells in the intestine. Different regulatory T cells can efficiently suppress specific T helper cell subsets in the intestine.

Although different types of regulatory T cells can partially compensate each other, it seems that regulatory T cells can also have a more specialized function, and suppress specific types of effector T cells more potent then others: mice with a selective deficiency in iTreg cells develop spontaneous intestinal inflammation, which is characterized by an expansion of Th2 cells [113], indicating that iTregs play an important role in controlling Th2 cells in the intestine. Moreover expression of GATA-3 and IRF-4, master regulators of Th2 cells, by Foxp3+ Treg cells is important for the control of Th2 cells [148, 149]. Additionally, some Foxp3+ Treg cells can express T-bet the master transcriptional regulator of Th1 cells. These T-bet+Foxp3+ Tregs express CXCR3, which is also highly expressed by Th1 cells. Thanks to the expression of the same chemokine receptor T-bet+Foxp3+ Treg cells can better “follow” and in turn suppress Th1 cells [150]. Finally, it was shown that IL-10 can induce IL-10 production by Foxp3+ Treg via STAT3 activation, and Foxp3+IL-10+ Tregs are particularly important to control Th17 cells [76, 138, 151]. In addition to Foxp3+ Treg the immune system uses an alternative type of regulatory T cell, which can compensate a possible paucity of Foxp3+ Treg in order to avoid immune pathology in the intestine [112]. These cells, Tr1 cells, which are characterized by an abundant production of IL-10 and by the absence of Foxp3 expression, exert an efficient regulation of Th17 cells in the intestine [114]. Interestingly, IL-10 seems to play a non-redundant role in controlling Th17 cells: acting on both Th17 cells and regulatory T cells. Th17 cells are suppressed directly via IL-10, which is produced by Tr1 and Foxp3+Treg cells [114, 136]. Additionally, IL-10 acts on Foxp3+Treg. It activates STAT3 in Foxp3+Tregs, which is crucial to enable them to suppress Th17 cells [138, 151]. Moreover IL-10 signalling in Foxp3+Treg cells is required to promote IL-10 production [138] (Figure 3).

In conclusion Foxp3+ Treg cells can have different phenotypes. This feature allows Foxp3+ Tregs to suppress specific effector T cells more efficiently. Additionally Foxp3+ Tregs can team up with Tr1 cells to maintain the immune homeostasis in the intestine.

However, regulatory T cells do not only suppress effector T cells but can also promote effector T cell function in some settings [152, 153], indicating that the immune system aims to maintain a proper balance between regulatory and effector T cells rather then uncontrolled suppression of effector T cells.

Advertisement

6. Conclusions

CD4+ T helper cells have important physiological functions at the large intestinal mucosal surface: they secrete cytokines thereby attracting other immune cells, inducing anti-microbial peptides, and promoting tissue repair. Therefore effector CD4+ T helper cells play an important ‘border patrol’ function, and protect the body against infections. Thymic derived naïve CD4+ T cells express bacterial antigen specific TCRs. Encounter with these bacterial derived foreign antigens in the colon can drive the differentiation of regulatory T cells or pro-inflammatory effector T cells dependent on the bacteria and the environmental milieu. If effector CD4+ T helper cells are uncontrolled, they can elicit tissue damage and induce disease such as IBD. Therefore the immune system has established several mechanisms in order to control pro-inflammatory T helper cells. These mechanisms are primarily important to avoid immune pathology and in turn to maintain tolerance in the intestine. However a growing body of evidence suggests that these mechanisms can also be used to suppress other organ specific diseases. One example for this interaction between the intestine and another organ is that the commensal gut flora can trigger a T-cell mediated immune response, which leads to autoimmune disease in the brain [99]. Therefore one possible strategy for the treatment of autoimmune diseases in future would be to specifically target the gut flora. Although the mechanisms controlling effector T cells in the intestine work well in most humans, the frequency of autoimmune and chronic inflammatory disease is steadily increasing. Unfortunately, there are currently no curative treatments for these diseases available. Therefore the patients suffer from the side effects of the drugs and from the relapses of their disease. The the challenge will be to better understand the mechanisms controlling effector T cells in order to establish new and potentially curative treatments for autoimmune and chronic inflammatory diseases. The intestine has the potential to serve as the key target organ of these therapies.

References

  1. 1. BettelliE.CarrierY.GaoW.KornT.StromT. B.OukkaM.WeinerH. L.KuchrooV. K.2006Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 70902358
  2. 2. KornT.BettelliE.OukkaM.KuchrooV. K.2009IL-17 and Th17 Cells. Annu Rev Immunol. 485517
  3. 3. ZhengW.FlavellR. A.1997The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells. Cell. 458796
  4. 4. IvanovI. I.BSMc KenzieZhou. L.CETadokoroLepelley. A.LafailleJ. J.CuaD. J.LittmanD. R.2006The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 6112133
  5. 5. SzaboS. J.KimS. T.CostaG. L.ZhangX.FathmanC. G.GlimcherL. H.2000A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell. 665569
  6. 6. ZhuJ.YamaneH.PaulW. E.2010Differentiation of effector CD4 T cell populations (*). Annu Rev Immunol. 44589
  7. 7. MosmannT. R.CherwinskiH.BondM. W.MAGiedlinCoffman. R. L.1986Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol. 7234857
  8. 8. HsiehC. S.MacatoniaS. E.TrippC. S.WolfS. F.O’GarraA.MurphyK. M.1993Development of TH1 CD4+ T cells through IL-12 produced by Listeria-induced macrophages. Science. 51075479
  9. 9. UsuiT.PreissJ. C.KannoY.YaoZ. J.BreamJ. H.O’SheaJ. J.StroberW.2006T-bet regulates Th1 responses through essential effects on GATA-3 function rather than on IFNG gene acetylation and transcription. J Exp Med. 375566
  10. 10. UsuiT.NishikomoriR.KitaniA.StroberW.2003GATA-3 suppresses Th1 development by downregulation of Stat4 and not through effects on IL-12Rbeta2 chain or T-bet. Immunity. 341528
  11. 11. SzaboS. J.BMSullivanStemmann. C.SatoskarA. R.SleckmanB. P.GlimcherL. H.2002Distinct effects of T-bet in TH1 lineage commitment and IFN-gamma production in CD4 and CD8 T cells. Science. 555333842
  12. 12. Filipe-SantosO.BustamanteJ.ChapgierA.VogtG.de BeaucoudreyL.FeinbergJ.JouanguyE.Boisson-DupuisS.FieschiC.PicardC.CasanovaJ. L.2006Inborn errors of IL-12/23- and IFN-gamma-mediated immunity: molecular, cellular, and clinical features. Semin Immunol. 634761
  13. 13. Le GrosG.Ben-SassonS. Z.SederR.FinkelmanF. D.PaulW. E.1990Generation of interleukin 4 (IL-4)-producing cells in vivo and in vitro: IL-2 and IL-4 are required for in vitro generation of IL-4-producing cells. J Exp Med. 39219
  14. 14. SederR. A.BoulayJ. L.FinkelmanF.BarbierS.Ben-SassonS. Z.Le GrosG.PaulW. E.1992CD8+ T cells can be primed in vitro to produce IL-4. J Immunol. 616526
  15. 15. SwainS. L.WeinbergA. D.EnglishM.HustonG.1990IL-4 directs the development of Th2-like helper effectors. J Immunol. 113796806
  16. 16. KaplanM. H.SchindlerU.SmileyS. T.MJGrusby1996Stat6 is required for mediating responses to IL-4 and for development of Th2 cells. Immunity. 33139
  17. 17. ShimodaK.van DeursenJ.SangsterM. Y.SarawarS. R.CarsonR. T.TrippR. A.ChuC.QuelleF. W.NosakaT.VignaliD. A.DohertyP. C.GrosveldG.PaulW. E.IhleJ. N.1996Lack of IL-4-induced Th2 response and IgE class switching in mice with disrupted Stat6 gene. Nature. 65756303
  18. 18. TakedaK.TanakaT.ShiW.MatsumotoM.MinamiM.KashiwamuraS.NakanishiK.YoshidaN.KishimotoT.AkiraS.1996Essential role of Stat6 in IL-4 signalling. Nature. 657562730
  19. 19. KurataH.LeeH. J.O’GarraA.AraiN.1999Ectopic expression of activated Stat6 induces the expression of Th2-specific cytokines and transcription factors in developing Th1 cells. Immunity. 667788
  20. 20. ZhuJ.GuoL.WatsonC. J.Hu-LiJ.PaulW. E.2001Stat6 is necessary and sufficient for IL-4’s role in Th2 differentiation and cell expansion. J Immunol. 12727681
  21. 21. ZhangD. H.CohnL.RayP.BottomlyK.RayA.1997Transcription factor GATA-3 is differentially expressed in murine Th1 and Th2 cells and controls Th2-specific expression of the interleukin-5 gene. J Biol Chem. 3421597603
  22. 22. ZhuJ.MinB.Hu-LiJ.WatsonC. J.GrinbergA.WangQ.KilleenN.UrbanJ. F.Jr GuoL.PaulW. E.2004Conditional deletion of Gata3 shows its essential function in T(H)1-T(H)2 responses. Nat Immunol. 11115765
  23. 23. ZhuJ.Cote-SierraJ.GuoL.PaulW. E.2003Stat5 activation plays a critical role in Th2 differentiation. Immunity. 573948
  24. 24. UrbanJ. F.Jr Noben-TrauthN.DonaldsonD. D.MaddenK. B.MorrisS. C.CollinsM.FinkelmanF. D.1998IL-13, IL-4Ralpha, and Stat6 are required for the expulsion of the gastrointestinal nematode parasite Nippostrongylus brasiliensis. Immunity. 225564
  25. 25. Anthony RM, Rutitzky LI, Urban JF, Jr., Stadecker MJ, Gause WC2007Protective immune mechanisms in helminth infection. Nat Rev Immunol. 1297587
  26. 26. AnthonyR. M.UrbanJ. F.Jr AlemF.HamedH. A.RozoC. T.BoucherJ. L.Van RooijenN.GauseW. C.2006Memory T(H)2 cells induce alternatively activated macrophages to mediate protection against nematode parasites. Nat Med. 895560
  27. 27. HasnainS. Z.EvansC. M.RoyM.GallagherA. L.KindrachukK. N.BarronL.DickeyB. F.MSWilsonWynn. T. A.GrencisR. K.ThorntonD. J.2011Muc5ac: a critical component mediating the rejection of enteric nematodes. J Exp Med. 5893900
  28. 28. HerbertD. R.YangJ. Q.HoganS. P.GroschwitzK.KhodounM.MunitzA.OrekovT.PerkinsC.WangQ.BrombacherF.UrbanJ. F.Jr MERothenbergFinkelman. F. D.2009Intestinal epithelial cell secretion of RELM-beta protects against gastrointestinal worm infection. J Exp Med. 13294757
  29. 29. ArtisD.WangM. L.KeilbaughS. A.HeW.BrenesM.SwainG. P.KnightP. A.DonaldsonD. D.MALazarMiller. H. R.SchadG. A.ScottP.WuG. D.2004RELMbeta/FIZZ2 is a goblet cell-specific immune-effector molecule in the gastrointestinal tract. Proc Natl Acad Sci U S A. 3713596600
  30. 30. AkihoH.BlennerhassettP.DengY.CollinsS. M.2002Role of IL-4, IL-13, and STAT6 in inflammation-induced hypercontractility of murine smooth muscle cells. Am J Physiol Gastrointest Liver Physiol. 2: G22632
  31. 31. CliffeL. J.HumphreysN. E.LaneT. E.PottenC. S.BoothC.GrencisR. K.2005Accelerated intestinal epithelial cell turnover: a new mechanism of parasite expulsion. Science. 572714635
  32. 32. ZhouL.IvanovI. I.SpolskiR.MinR.ShenderovK.EgawaT.LevyD. E.LeonardW. J.LittmanD. R.2007IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol. 996774
  33. 33. NurievaR.YangX. O.MartinezG.ZhangY.PanopoulosA. D.MaSchlunsL.TianK.WatowichQ.JettenS. S.DongA. M.C.2007Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature. 71524803
  34. 34. YangX. O.PanopoulosA. D.NurievaR.ChangS. H.WangD.WatowichS. S.DongC.2007STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem. 13935863
  35. 35. VeldhoenM.HockingR. J.AtkinsC. J.LocksleyR. M.StockingerB.2006TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 217989
  36. 36. Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR, Weaver CT2006Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. 70902314
  37. 37. ZhouL.LopesJ. E.MMChongIvanov. I. I.MinR.VictoraG. D.ShenY.DuJ.RubtsovY. P.RudenskyA. Y.ZieglerS. F.LittmanD. R.2008TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature. 719223640
  38. 38. MJMc GeachyChen. Y.TatoC. M.LaurenceA.Joyce-ShaikhB.BlumenscheinW. M.Mc ClanahanT. K.O’SheaJ. J.CuaD. J.2009The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat Immunol. 331424
  39. 39. AhernP. P.SchieringC.BuonocoreS.MJMc GeachyCua. D. J.MaloyK. J.PowrieF.2010Interleukin-23 drives intestinal inflammation through direct activity on T cells. Immunity. 227988
  40. 40. EspluguesE.HuberS.GaglianiN.HauserA. E.TownT.WanY. Y.O’ConnorW.Jr RongvauxA.Van RooijenN.HabermanA. M.IwakuraY.KuchrooV. K.KollsJ. K.BluestoneJ. A.HeroldK. C.FlavellR. A.2011Control of TH17 cells occurs in the small intestine. Nature. 73575148
  41. 41. IvanovI. I.AtarashiK.ManelN.BrodieE. L.ShimaT.KaraozU.WeiD.GoldfarbK. C.CASanteeLynch. S. V.TanoueT.ImaokaA.ItohK.TakedaK.UmesakiY.HondaK.LittmanD. R.2009Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 348598
  42. 42. AtarashiK.NishimuraJ.ShimaT.UmesakiY.YamamotoM.OnoueM.YagitaH.IshiiN.EvansR.HondaK.TakedaK.2008ATP drives lamina propria T(H)17 cell differentiation. Nature. 721480812
  43. 43. ShawM. H.KamadaN.KimY. G.NunezG.2012Microbiota-induced IL-1beta, but not IL-6, is critical for the development of steady-state TH17 cells in the intestine. J Exp Med. 22518
  44. 44. CEZielinskiMele. F.AschenbrennerD.JarrossayD.RonchiF.GattornoM.MonticelliS.LanzavecchiaA.SallustoF.2012Pathogen-induced human T(H)17 cells produce IFN-gamma or IL-10 and are regulated by IL-1beta. Nature.
  45. 45. GhoreschiK.LaurenceA.YangX. P.TatoC. M.MJMc GeachyKonkel. J. E.RamosH. L.WeiL.DavidsonT. S.BouladouxN.GraingerJ. R.ChenQ.KannoY.WatfordW. T.SunH. W.EberlG.ShevachE. M.BelkaidY.CuaD. J.ChenW.O’SheaJ. J.2010Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling. Nature. 731896771
  46. 46. MJMc Geachy-JensenBak.ChenK. S.TatoY.BlumenscheinC. M.Mc ClanahanW.CuaT.D. J.2007TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol. 1213907
  47. 47. VeldhoenM.HirotaK.ChristensenJ.O’GarraA.StockingerB.2009Natural agonists for aryl hydrocarbon receptor in culture medium are essential for optimal differentiation of Th17 T cells. J Exp Med. 1439
  48. 48. VeldhoenM.HirotaK.WestendorfA. M.BuerJ.DumoutierL.RenauldJ. C.StockingerB.2008The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature. 71911069
  49. 49. QuintanaF. J.ASBassoIglesias. A. H.KornT.FarezM. F.BettelliE.CaccamoM.OukkaM.WeinerH. L.2008Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature. 71916571
  50. 50. O’ConnorW.Jr ZenewiczL. A.FlavellR. A.2010The dual nature of T(H)17 cells: shifting the focus to function. Nat Immunol. 64716
  51. 51. ApetohL.QuintanaF. J.PotC.JollerN.XiaoS.KumarD.BurnsE. J.SherrD. H.WeinerH. L.KuchrooV. K.2010The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat Immunol. 985461
  52. 52. ContiH. R.ShenF.NayyarN.StocumE.SunJ. N.MJLindemannHo. A. W.HaiJ. H.YuJ. J.JungJ. W.FillerS. G.Masso-WelchP.EdgertonM.GaffenS. L.2009Th17 cells and IL-17 receptor signaling are essential for mucosal host defense against oral candidiasis. J Exp Med. 2299311
  53. 53. HuangW.NaL.FidelP. L.SchwarzenbergerP.2004Requirement of interleukin-17A for systemic anti-Candida albicans host defense in mice. J Infect Dis. 362431
  54. 54. YeP.GarveyP. B.ZhangP.NelsonS.BagbyG.SummerW. R.SchwarzenbergerP.ShellitoJ. E.KollsJ. K.2001Interleukin-17 and lung host defense against Klebsiella pneumoniae infection. Am J Respir Cell Mol Biol. 333540
  55. 55. IwakuraY.NakaeS.SaijoS.IshigameH.2008The roles of IL-17A in inflammatory immune responses and host defense against pathogens. Immunol Rev. 5779
  56. 56. IshigameH.KakutaS.NagaiT.KadokiM.NambuA.KomiyamaY.FujikadoN.TanahashiY.AkitsuA.KotakiH.SudoK.NakaeS.SasakawaC.IwakuraY.2009Differential roles of interleukin-17A and-17F in host defense against mucoepithelial bacterial infection and allergic responses. Immunity. 110819
  57. 57. OuyangW.KollsJ. K.ZhengY.2008The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity. 445467
  58. 58. PelletierM.MaggiL.MichelettiA.LazzeriE.TamassiaN.CostantiniC.CosmiL.LunardiC.AnnunziatoF.RomagnaniS.MACassatella2010Evidence for a cross-talk between human neutrophils and Th17 cells. Blood. 233543
  59. 59. KaoC. Y.ChenY.ThaiP.WachiS.HuangF.KimC.HarperR. W.WuR.2004IL-17 markedly up-regulates beta-defensin-2 expression in human airway epithelium via JAK and NF-kappaB signaling pathways. J Immunol. 5348291
  60. 60. LiangS. C.TanX. Y.LuxenbergD. P.KarimR.Dunussi-JoannopoulosK.CollinsM.FouserL. A.2006Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med. 1022719
  61. 61. ZhengY.ValdezP. A.DanilenkoD. M.HuY.SaS. M.GongQ.AbbasA. R.ModrusanZ.GhilardiN.de SauvageF. J.OuyangW.2008Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nat Med. 32829
  62. 62. ZenewiczL. A.YancopoulosG. D.ValenzuelaD. M.MurphyA. J.StevensS.FlavellR. A.2008Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease. Immunity. 694757
  63. 63. PickertG.NeufertC.LeppkesM.ZhengY.WittkopfN.WarntjenM.LehrH. A.HirthS.WeigmannB.WirtzS.OuyangW.NeurathM. F.BeckerC.2009STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. J Exp Med. 7146572
  64. 64. SugimotoK.OgawaA.MizoguchiE.ShimomuraY.AndohA.BhanA. K.BlumbergR. S.XavierR. J.MizoguchiA.2008IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Invest. 253444
  65. 65. SakaguchiS.SakaguchiN.AsanoM.ItohM.TodaM.1995Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 3115164
  66. 66. Fontenot JD, Gavin MA, Rudensky AY2003Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol. 43306
  67. 67. HoriS.NomuraT.SakaguchiS.2003Control of regulatory T cell development by the transcription factor Foxp3. Science. 5609105761
  68. 68. HuberS.SchrammC.LehrH. A.MannA.SchmittS.BeckerC.ProtschkaM.GalleP. R.NeurathM. F.BlessingM.2004Cutting edge: TGF-beta signaling is required for the in vivo expansion and immunosuppressive capacity of regulatory CD4+CD25+ T cells. J Immunol. 11652631
  69. 69. SchrammC.HuberS.ProtschkaM.CzochraP.BurgJ.SchmittE.LohseA. W.GalleP. R.BlessingM.2004TGFbeta regulates the CD4+CD25+ T-cell pool and the expression of Foxp3 in vivo. Int Immunol. 912419
  70. 70. FantiniM. C.BeckerC.MonteleoneG.PalloneF.GalleP. R.NeurathM. F.2004Cutting edge: TGF-beta induces a regulatory phenotype in CD4+CD25- T cells through Foxp3 induction and down-regulation of Smad7. J Immunol. 9514953
  71. 71. ChenW.JinW.HardegenN.LeiK. J.LiL.MarinosN.Mc GradyG.WahlS. M.2003Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 12187586
  72. 72. KretschmerK.ApostolouI.HawigerD.KhazaieK.NussenzweigM. C.vonBoehmer. H.2005Inducing and expanding regulatory T cell populations by foreign antigen. Nat Immunol. 12121927
  73. 73. MarieJ. C.LetterioJ. J.GavinM.RudenskyA. Y.2005TGF-beta1 maintains suppressor function and Foxp3 expression in CD4+CD25+ regulatory T cells. J Exp Med. 710617
  74. 74. BennettC. L.ChristieJ.RamsdellF.MEBrunkowFerguson. P. J.WhitesellL.KellyT. E.SaulsburyF. T.ChanceP. F.OchsH. D.2001The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 1201
  75. 75. MEBrunkowJeffery. E. W.HjerrildK. A.PaeperB.ClarkL. B.YasaykoS. A.WilkinsonJ. E.GalasD.ZieglerS. F.RamsdellF.2001Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. 16873
  76. 76. HuberS.GaglianiN.EspluguesE.O’ConnorW.Jr HuberF. J.ChaudhryA.KamanakaM.KobayashiY.BoothC. J.RudenskyA. Y.RoncaroloM. G.BattagliaM.FlavellR. A.2011Th17 cells express interleukin-10 receptor and are controlled by Foxp3- and Foxp3+ regulatory CD4+ T cells in an interleukin-10-dependent manner. Immunity. 455465
  77. 77. FahlenL.ReadS.GorelikL.HurstS. D.CoffmanR. L.FlavellR. A.PowrieF.2005T cells that cannot respond to TGF-{beta} escape control by CD4+CD25+ regulatory T cells. J Exp Med. 573746
  78. 78. AnnunziatoF.CosmiL.LiottaF.LazzeriE.ManettiR.VaniniV.RomagnaniP.MaggiE.RomagnaniS.2002Phenotype, localization, and mechanism of suppression of CD4(+)CD25(+) human thymocytes. J Exp Med. 337987
  79. 79. BoppT.BeckerC.KleinM.Klein-HesslingS.PalmetshoferA.SerflingE.HeibV.BeckerM.KubachJ.SchmittS.StollS.SchildH.MSStaegeStassen. M.JonuleitH.SchmittE.2007Cyclic adenosine monophosphate is a key component of regulatory T cell-mediated suppression. J Exp Med. 6130310
  80. 80. RoncaroloM. G.GregoriS.BattagliaM.BacchettaR.FleischhauerK.LevingsM. K.2006Interleukin-10secreting type 1 regulatory T cells in rodents and humans. Immunol Rev. 28-50.
  81. 81. HaringerB.LozzaL.SteckelB.GeginatJ.2009Identification and characterization of IL-10/IFN-gamma-producing effector-like T cells with regulatory function in human blood. J Exp Med. 5100917
  82. 82. VieiraP. L.ChristensenJ. R.MinaeeS.O’NeillE. J.BarratF. J.BoonstraA.BarthlottT.StockingerB.WraithD. C.O’GarraA.2004IL-10-secreting regulatory T cells do not express Foxp3 but have comparable regulatory function to naturally occurring CD4+CD25+ regulatory T cells. J Immunol. 10598693
  83. 83. Maynard CL, Harrington LE, Janowski KM, Oliver JR, Zindl CL, Rudensky AY, Weaver CT2007Regulatory T cells expressing interleukin 10 develop from Foxp3+ and Foxp3- precursor cells in the absence of interleukin 10. Nat Immunol. 993141
  84. 84. PasseriniL.Di NunzioS.GregoriS.GambineriE.CecconiM.SeidelM. G.CazzolaG.PerroniL.TommasiniA.VignolaS.GuidiL.RoncaroloM. G.BacchettaR.2011Functional type 1 regulatory T cells develop regardless of FOXP3 mutations in patients with IPEX syndrome. Eur J Immunol. 4112031
  85. 85. GrouxH.O’GarraA.BiglerM.RouleauM.AntonenkoS.de VriesJ. E.RoncaroloM. G.1997A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature. 665273742
  86. 86. GaglianiN.JofraT.StabiliniA.ValleA.AtkinsonM.RoncaroloM. G.BattagliaM.2010Antigen-specific dependence of Tr1-cell therapy in preclinical models of islet transplant. Diabetes. 24339
  87. 87. PotC.ApetohL.AwasthiA.KuchrooV. K.2010Molecular pathways in the induction of interleukin-27-driven regulatory type 1 cells. J Interferon Cytokine Res. 63818
  88. 88. CobboldS. P.NolanK. F.GracaL.CastejonR.Le MoineA.FrewinM.HummS.AdamsE.ThompsonS.ZelenikaD.PatersonA.YatesS.FairchildP. J.WaldmannH.2003Regulatory T cells and dendritic cells in transplantation tolerance: molecular markers and mechanisms. Immunol Rev. 10924
  89. 89. PestkaS.KrauseC. D.SarkarD.WalterM. R.ShiY.FisherP. B.2004Interleukin-10 and related cytokines and receptors. Annu Rev Immunol. 92979
  90. 90. CavaniA.NasorriF.PrezziC.SebastianiS.AlbanesiC.GirolomoniG.2000Human CD4+ T lymphocytes with remarkable regulatory functions on dendritic cells and nickel-specific Th1 immune responses. J Invest Dermatol. 2295302
  91. 91. StroblH.EmshoffR.RothlerG.1999Conservative treatment of unilateral condylar fractures in children: a long-term clinical and radiologic follow-up of 55 patients. Int J Oral Maxillofac Surg. 2958
  92. 92. CerwenkaA.SwainS. L.1999TGF-beta1: immunosuppressant and viability factor for T lymphocytes. Microbes Infect. 1512916
  93. 93. LevingsM. K.GregoriS.TresoldiE.CazzanigaS.BoniniC.RoncaroloM. G.2005Differentiation of Tr1 cells by immature dendritic cells requires IL-10 but not CD25+CD4+ Tr cells. Blood. 311629
  94. 94. GregoriS.TomasoniD.PaccianiV.ScirpoliM.BattagliaM.MagnaniC. F.HaubenE.RoncaroloM. G.2010Differentiation of type 1 T regulatory cells (Tr1) by tolerogenic DC-10 requires the IL-10-dependent ILT4/HLA-G pathway. Blood. 693544
  95. 95. GrossmanW. J.VerbskyJ. W.TollefsenB. L.KemperC.AtkinsonJ. P.LeyT. J.2004Differential expression of granzymes A and B in human cytotoxic lymphocyte subsets and T regulatory cells. Blood. 928408
  96. 96. GrossmanW. J.VerbskyJ. W.BarchetW.ColonnaM.AtkinsonJ. P.LeyT. J.2004Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity. 4589601
  97. 97. MagnaniC. F.AlberigoG.BacchettaR.SerafiniG.AndreaniM.RoncaroloM. G.GregoriS.2011Killing of myeloid APCs via HLA class I, CD2 and CD226 defines a novel mechanism of suppression by human Tr1 cells. Eur J Immunol. 6165262
  98. 98. HuberS.SchrammC.2006TGF-beta and CD4+CD25+ regulatory T cells. Front Biosci. 101423
  99. 99. BererK.MuesM.KoutrolosM.RasbiZ. A.BozikiM.JohnerC.WekerleH.KrishnamoorthyG.2011Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature. 737453841
  100. 100. ElinavE.StrowigT.KauA. L.Henao-MejiaJ.CAThaissBooth. C. J.PeaperD. R.BertinJ.EisenbarthS. C.GordonJ. I.FlavellR. A.2011NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 574557
  101. 101. LathropS. K.BloomS. M.RaoS. M.NutschK.LioC. W.SantacruzN.PetersonD. A.StappenbeckT. S.HsiehC. S.2011Peripheral education of the immune system by colonic commensal microbiota. Nature. 73682504
  102. 102. DeckerE.HornefM.StockingerS.2011Cesarean delivery is associated with celiac disease but not inflammatory bowel disease in children. Gut Microbes. 2918
  103. 103. MJBenson-LagosPino.RosemblattK.NoelleM.R. J.2007All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation. J Exp Med. 8176574
  104. 104. HadisU.WahlB.SchulzO.Hardtke-WolenskiM.SchippersA.WagnerN.MullerW.SparwasserT.ForsterR.PabstO.2011Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity. 223746
  105. 105. CassaniB.VillablancaE. J.QuintanaF. J.LoveP. E.Lacy-HulbertA.BlanerW. S.SparwasserT.SnapperS. B.WeinerH. L.MoraJ. R.2011Gut-Tropic T Cells That Express Integrin alpha4beta7 and CCR9 Are Required for Induction of Oral Immune Tolerance in Mice. Gastroenterology.
  106. 106. GeukingM. B.CahenzliJ.MALawsonNg. D. C.SlackE.HapfelmeierS.Mc CoyK. D.MacphersonA. J.2011Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity. 5794806
  107. 107. HuberS.SchrammC.2011Role of activin A in the induction of Foxp3+ and Foxp3- CD4+ regulatory T cells. Crit Rev Immunol. 15360
  108. 108. HuberS.StahlF. R.SchraderJ.LuthS.PresserK.CarambiaA.FlavellR. A.WernerS.BlessingM.HerkelJ.SchrammC.2009Activin a promotes the TGF-beta-induced conversion of CD4+CD25- T cells into Foxp3+ induced regulatory T cells. J Immunol. 8463340
  109. 109. FantiniM. C.BeckerC.TubbeI.NikolaevA.LehrH. A.GalleP.NeurathM. F.2006Transforming growth factor beta induced FoxP3+ regulatory T cells suppress Th1 mediated experimental colitis. Gut. 567180
  110. 110. MottetC.UhligH. H.PowrieF.2003Cutting edge: cure of colitis by CD4+CD25+ regulatory T cells. J Immunol. 8393943
  111. 111. HaribhaiD.WilliamsJ. B.JiaS.NickersonD.SchmittE. G.EdwardsB.ZiegelbauerJ.YassaiM.LiS. H.RellandL. M.WiseP. M.ChenA.ZhengY. Q.SimpsonP. M.GorskiJ.SalzmanN. H.MJHessnerChatila. T. A.WilliamsC. B.2011A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity. Immunity. 110922
  112. 112. ZhengY.JosefowiczS.ChaudhryA.PengX. P.ForbushK.RudenskyA. Y.2010Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature. 728280812
  113. 113. JosefowiczS. Z.NiecR. E.KimH. Y.TreutingP.ChinenT.ZhengY.UmetsuD. T.RudenskyA. Y.2012Extrathymically generated regulatory T cells control mucosal TH2 inflammation. Nature. 73853959
  114. 114. HuberS.GaglianiN.EspluguesE.O’ConnorW.Jr HuberF. J.ChaudhryA.KamanakaM.KobayashiY.BoothC. J.RudenskyA. Y.RoncaroloM. G.BattagliaM.FlavellR. A.2011Th17 cells express interleukin-10 receptor and are controlled by Foxp3 and Foxp3+ regulatory CD4+ T cells in an interleukin-10-dependent manner. Immunity. 455465
  115. 115. FussI. J.NeurathM.BoirivantM.KleinJ. S.de la MotteC.StrongS. A.FiocchiC.StroberW.1996Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn’s disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J Immunol. 3126170
  116. 116. FujinoS.AndohA.BambaS.OgawaA.HataK.ArakiY.BambaT.FujiyamaY.2003Increased expression of interleukin 17 in inflammatory bowel disease. Gut. 16570
  117. 117. RovedattiL.KudoT.BiancheriP.SarraM.KnowlesC. H.DSRamptonCorazza. G. R.MonteleoneG.Di SabatinoA.MacdonaldT. T.2009Differential regulation of interleukin 17 and interferon gamma production in inflammatory bowel disease. Gut. 12162936
  118. 118. AnnunziatoF.CosmiL.SantarlasciV.MaggiL.LiottaF.MazzinghiB.ParenteE.FiliL.FerriS.FrosaliF.GiudiciF.RomagnaniP.ParronchiP.TonelliF.MaggiE.RomagnaniS.2007Phenotypic and functional features of human Th17 cells. J Exp Med. 8184961
  119. 119. BrandS.BeigelF.OlszakT.ZitzmannK.EichhorstS. T.OtteJ. M.DiepolderH.MarquardtA.JaglaW.PoppA.LeclairS.HerrmannK.SeidererJ.OchsenkuhnT.GokeB.AuernhammerC. J.DambacherJ.2006IL-22 is increased in active Crohn’s disease and promotes proinflammatory gene expression and intestinal epithelial cell migration. Am J Physiol Gastrointest Liver Physiol. 4: G82738
  120. 120. SeidererJ.ElbenI.DiegelmannJ.GlasJ.StallhoferJ.TillackC.PfennigS.JurgensM.SchmechelS.KonradA.GokeB.OchsenkuhnT.Muller-MyhsokB.LohseP.BrandS.2008Role of the novel Th17 cytokine IL-17F in inflammatory bowel disease (IBD): upregulated colonic IL-17F expression in active Crohn’s disease and analysis of the IL17F His161Argpolymorphism in IBD. Inflamm Bowel Dis. 4: 437-45.
  121. 121. DuerrR. H.TaylorK. D.BrantS. R.JDRiouxSilverberg.MJDalySteinhart. A. H.AbrahamC.RegueiroM.GriffithsA.DassopoulosT.BittonA.YangH.TarganS.DattaL. W.KistnerE. O.SchummL. P.LeeA. T.GregersenP. K.MMBarmadaRotter. J. I.NicolaeD. L.ChoJ. H.2006A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 580414613
  122. 122. Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, Ahmad T, Lees CW, Balschun T, Lee J, Roberts R, Anderson CA, Bis JC, Bumpstead S, Ellinghaus D, Festen EM, Georges M, Green T, Haritunians T, Jostins L, Latiano A, Mathew CG, Montgomery GW, Prescott NJ, Raychaudhuri S, Rotter JI, Schumm P, Sharma Y, Simms LA, Taylor KD, Whiteman D, Wijmenga C, Baldassano RN, Barclay M, Bayless TM, Brand S, Buning C, Cohen A, Colombel JF, Cottone M, Stronati L, Denson T, De Vos M, D’Inca R, Dubinsky M, Edwards C, Florin T, Franchimont D, Gearry R, Glas J, Van Gossum A, Guthery SL, Halfvarson J, Verspaget HW, Hugot JP, Karban A, Laukens D, Lawrance I, Lemann M, Levine A, Libioulle C, Louis E, Mowat C, Newman W, Panes J, Phillips A, Proctor DD, Regueiro M, Russell R, Rutgeerts P, Sanderson J, Sans M, Seibold F, Steinhart AH, Stokkers PC, Torkvist L, Kullak-Ublick G, Wilson D, Walters T, Targan SR, Brant SR, Rioux JD, D’Amato M, Weersma RK, Kugathasan S, Griffiths AM, Mansfield JC, Vermeire S, Duerr RH, Silverberg MS, Satsangi J, Schreiber S, Cho JH, Annese V, Hakonarson H, Daly MJ, Parkes M (2010) Genome-wide meta-analysis increases to 71 the number of confirmed Crohn’s disease susceptibility loci. Nat Genet. 12: 1118-25.
  123. 123. Anderson CA, Boucher G, Lees CW, Franke A, D’Amato M, Taylor KD, Lee JC, Goyette P, Imielinski M, Latiano A, Lagace C, Scott R, Amininejad L, Bumpstead S, Baidoo L, Baldassano RN, Barclay M, Bayless TM, Brand S, Buning C, Colombel JF, Denson LA, De Vos M, Dubinsky M, Edwards C, Ellinghaus D, Fehrmann RS, Floyd JA, Florin T, Franchimont D, Franke L, Georges M, Glas J, Glazer NL, Guthery SL, Haritunians T, Hayward NK, Hugot JP, Jobin G, Laukens D, Lawrance I, Lemann M, Levine A, Libioulle C, Louis E, McGovern DP, Milla M, Montgomery GW, Morley KI, Mowat C, Ng A, Newman W, Ophoff RA, Papi L, Palmieri O, Peyrin-Biroulet L, Panes J, Phillips A, Prescott NJ, Proctor DD, Roberts R, Russell R, Rutgeerts P, Sanderson J, Sans M, Schumm P, Seibold F, Sharma Y, Simms LA, Seielstad M, Steinhart AH, Targan SR, van den Berg LH, Vatn M, Verspaget H, Walters T, Wijmenga C, Wilson DC, Westra HJ, Xavier RJ, Zhao ZZ, Ponsioen CY, Andersen V, Torkvist L, Gazouli M, Anagnou NP, Karlsen TH, Kupcinskas L, Sventoraityte J, Mansfield JC, Kugathasan S, Silverberg MS, Halfvarson J, Rotter JI, Mathew CG, Griffiths AM, Gearry R, Ahmad T, Brant SR, Chamaillard M, Satsangi J, Cho JH, Schreiber S, Daly MJ, Barrett JC, Parkes M, Annese V, Hakonarson H, Radford-Smith G, Duerr RH, Vermeire S, Weersma RK, Rioux JD (2011) Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47. Nat Genet. 3: 246-52.
  124. 124. GlasJ.StallhoferJ.RipkeS.WetzkeM.PfennigS.KleinW.EpplenJ. T.GrigaT.SchiemannU.LacherM.KoletzkoS.FolwacznyM.LohseP.GokeB.OchsenkuhnT.Muller-MyhsokB.BrandS.2009Novel genetic risk markers for ulcerative colitis in the IL2/IL21 region are in epistasis with IL23R and suggest a common genetic background for ulcerative colitis and celiac disease. Am J Gastroenterol. 7173744
  125. 125. MiossecP.KornT.KuchrooV. K.2009Interleukin-17 and type 17 helper T cells. N Engl J Med. 988898
  126. 126. LeeY. K.TurnerH.MaynardC. L.OliverJ. R.ChenD.ElsonC. O.WeaverC. T.2009Late developmental plasticity in the T helper 17 lineage. Immunity. 192107
  127. 127. WangC.KangS. G.LeeJ.SunZ.KimC. H.2009The roles of CCR6 in migration of Th17 cells and regulation of effector T-cell balance in the gut. Mucosal Immunol. 217383
  128. 128. ElsonC. O.CongY.WeaverC. T.SchoebT. R.Mc ClanahanT. K.FickR. B.KasteleinR. A.2007Monoclonal anti-interleukin 23 reverses active colitis in a T cell-mediated model in mice. Gastroenterology. 7235970
  129. 129. ZhangZ.Zheng (2006) Critical role of IL-17 receptor signaling in acute TNBS-induced colitis. Inflamm Bowel Dis. 5: 382-8.
  130. 130. OgawaA.AndohA.ArakiY.BambaT.FujiyamaY.2004Neutralization of interleukin-17 aggravates dextran sulfate sodium-induced colitis in mice. Clin Immunol. 15562
  131. 131. YangX. O.ChangS. H.ParkH.NurievaR.ShahB.AceroL.WangY. H.SchlunsK. S.BroaddusR. R.ZhuZ.DongC.2008Regulation of inflammatory responses by IL-17F. J Exp Med. 5106375
  132. 132. O’ConnorW.Jr KamanakaM.BoothC. J.TownT.NakaeS.IwakuraY.KollsJ. K.FlavellR. A.2009A protective function for interleukin 17A in T cell-mediated intestinal inflammation. Nat Immunol. 66039
  133. 133. WeiG.WeiL.ZhuJ.ZangC.Hu-LiJ.YaoZ.CuiK.KannoY.RohT. Y.WatfordW. T.SchonesD. E.PengW.SunH. W.PaulW. E.O’SheaJ. J.ZhaoK.2009Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity. 115567
  134. 134. BernsteinB. E.MikkelsenT. S.XieX.KamalM.HuebertD. J.CuffJ.FryB.MeissnerA.WernigM.PlathK.JaenischR.WagschalA.FeilR.SchreiberS. L.LanderE. S.2006A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell. 231526
  135. 135. AzuaraV.PerryP.SauerS.SpivakovM.JorgensenH. F.JohnR. M.GoutiM.CasanovaM.WarnesG.MerkenschlagerM.FisherA. G.2006Chromatin signatures of pluripotent cell lines. Nat Cell Biol. 55328
  136. 136. KamanakaM.HuberS.ZenewiczL. A.GaglianiN.RathinamC.O’ConnorW.Jr WanY. Y.NakaeS.IwakuraY.HaoL.FlavellR. A.2011Memory/effector (CD45RB(lo)) CD4 T cells are controlled directly by IL-10 and cause IL-22-dependent intestinal pathology. J Exp Med. 5102740
  137. 137. PowrieF.LeachM. W.MauzeS.MenonS.CaddleL. B.CoffmanR. L.1994Inhibition of Th1 responses prevents inflammatory bowel disease in scid mice reconstituted with CD45RBhi CD4+ T cells. Immunity. 755362
  138. 138. ChaudhryA.SamsteinR. M.TreutingP.LiangY.PilsM. C.HeinrichJ. M.JackR. S.WunderlichF. T.BruningJ. C.MullerW.RudenskyA. Y.2011Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity. 456678
  139. 139. WitteE.WitteK.WarszawskaK.SabatR.WolkK.2010Interleukin-22: a cytokine produced by T, NK and NKT cell subsets, with importance in the innate immune defense and tissue protection. Cytokine Growth Factor Rev. 536579
  140. 140. CoombesJ. L.SiddiquiK. R.Arancibia-CarcamoC. V.HallJ.SunC. M.BelkaidY.PowrieF.2007A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med. 8175764
  141. 141. EliasK. M.LaurenceA.DavidsonT. S.StephensG.KannoY.ShevachE. M.O’SheaJ. J.2008Retinoic acid inhibits Th17 polarization and enhances FoxP3 expression through a Stat-3/Stat-5 independent signaling pathway. Blood. 3101320
  142. 142. LaurenceA.TatoC. M.DavidsonT. S.KannoY.ChenZ.YaoZ.BlankR. B.MeylanF.SiegelR.HennighausenL.ShevachE. M.O’SheaJ. J.2007Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity. 337181
  143. 143. YangX. P.GhoreschiK.Steward-TharpS. M.Rodriguez-CanalesJ.ZhuJ.GraingerJ. R.HiraharaK.SunH. W.WeiL.VahediG.KannoY.O’SheaJ. J.LaurenceA.2011Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5. Nat Immunol. 324754
  144. 144. PotC.ApetohL.AwasthiA.KuchrooV. K.2011Induction of regulatory Tr1 cells and inhibition of T(H)17 cells by IL-27. Semin Immunol. 643845
  145. 145. SaraivaM.O’GarraA.2010The regulation of IL-10 production by immune cells. Nat Rev Immunol. 317081
  146. 146. ReboldiA.CoisneC.BaumjohannD.BenvenutoF.BottinelliD.LiraS.UccelliA.LanzavecchiaA.EngelhardtB.SallustoF.2009C-C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat Immunol. 551423
  147. 147. GaglianiN.HuberS.FlavellR. A.2012The Intestine: where amazing things happen. Cell Res. 22779
  148. 148. WangY.MASuWan. Y. Y.2011An essential role of the transcription factor GATA-3 for the function of regulatory T cells. Immunity. 333748
  149. 149. ZhengY.ChaudhryA.KasA.de RoosP.KimJ. M.ChuT. T.CorcoranL.TreutingP.KleinU.RudenskyA. Y.2009Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature. 72363516
  150. 150. MAKoch-HeardTucker.PerdueG.KillebrewN. R.UrdahlJ. R.CampbellK. B.D. J.2009The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat Immunol. 6595602
  151. 151. ChaudhryA.RudraD.TreutingP.SamsteinR. M.LiangY.KasA.RudenskyA. Y.2009CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science. 595598691
  152. 152. ChenY.HainesC. J.GutcherI.HochwellerK.BlumenscheinW. M.Mc ClanahanT.HammerlingG.LiM. O.CuaD. J.MJMc Geachy2011Foxp3(+) regulatory T cells promote T helper 17 cell development in vivo through regulation of interleukin-2. Immunity. 340921
  153. 153. GutcherI.DonkorM. K.MaRudenskyQ.FlavellA. Y.LiR. A.M. O.2011Autocrine transforming growth factor-beta1 promotes in vivo Th17 cell differentiation. Immunity. 3396408

Written By

Nicola Gagliani and Samuel Huber

Submitted: 11 November 2011 Published: 25 July 2012