Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Sep 11.
Published in final edited form as: Immunol Res. 2014 May;58(0):358–368. doi: 10.1007/s12026-014-8512-5

Regulatory T cells and their roles in immune dysregulation and allergy

Laurence Pellerin 1, Jennifer A Jenks 1, Philippe Bégin 1, Rosa Bacchetta 1, Kari C Nadeau 1,
PMCID: PMC4161462  NIHMSID: NIHMS606190  PMID: 24781194

Abstract

The main function of the immune system is to fight off potential infections, but also to maintain its activity below a level that would trigger self-reactivity. Regulatory T cells (Tregs) such as forkhead box P3+ (FOXP3) Tregs and type 1 regulatory T cells (Tr1) play an essential role in this active process, using several distinct suppressive mechanisms. A wide range of pathologies have been associated with altered Treg cell function. This is best exemplified by the impact of mutations of genes essential for Treg function and the associated autoimmune syndromes. This review summarizes the main features of different subtypes of Tregs and focuses on the clinical implications of their altered function in human studies. More specifically, we discuss abnormalities affecting FOXP3+ Tregs and Tr1 cells that will lead to autoimmune manifestations and/or allergic reactions, and the potential therapeutic use of Tregs.

Keywords: Regulatory T cells, Type 1 regulatory T cells, FOXP3, Allergy, Immune dysregulation

Introduction

The ability of the immune system to control the inflammatory response and prevent reactions against self-antigens (Ags) or harmless environmental molecules is mainly acquired during fetal life and early after birth. Selection and acquisition of both effector and regulatory functions occur in the thymus for T cells and in the bone marrow for B cells and ensure that the naïve or Ag inexperienced newborn repertoire is devoid of the large majority of dangerous auto-reactive T and B cells that could cause harm. Throughout life, peripheral mechanisms of regulation control auto-reactive cells that escaped initial checkpoints and shut down undesired immune responses toward commensal and environmental Ags [1]. Once thought merely as a lack of response, it is now common knowledge that peripheral tolerance consists of active mechanisms in which specialized regulatory T cells (Treg) play a major role. In the last decade, a large body of evidence has unraveled novel features of Treg cells, and it has become possible to isolate them ex vivo or manipulate them in vitro for clinical purposes. This said, several questions remain to be addressed, especially in relation to their role in disease and their precise monitoring in vivo in humans. This review focuses on CD4+ Treg cells and summarizes their main characteristics, highlighting important issues that are critical to study their role in pathologies associated with loss of tolerance, such as in immune dysregulation of genetic origin leading to autoimmunity and in allergies.

Different types of human regulatory T cells

FOXP3+ Tregs

A consensus nomenclature has been recently introduced by Abbas and collaborators in order to facilitate understanding [2]. According to this work, Forkhead box P3 (FOXP3)+ Treg cells can be defined as follows: (1) “thymus-derived Treg cells”(tTregs) of thymic origin also known as thymic-derived/’ “natural” Tregs; (2) “peripherally derived Treg cells” (pTregs) which differentiate in the periphery, (3) “FOXP3+ Treg cell” should be used when the origin of the subset is unclear; and (4) “in vitro-induced Treg cells” should be used for all Treg cell populations generated ex vivo.

Thymic-derived Tregs constitute the best studied population of CD4+ Treg cells. Their existence was proven over a decade ago by Sakaguchi and collaborators who showed that tTregs have suppressive properties toward the development of autoimmune manifestations in mice [3]. They can be identified as expressing high levels of the interleukin 2 (IL-2) receptor alpha-chain (CD25), the co-inhibitory receptor cytotoxic T lymphocyte Ag 4 (CTLA4) and the transcription factor FOXP3 [36] which is essential for their function (Fig. 1a). In addition, they express low levels of the IL-7 receptor, CD127 [7], and mainly have a naïve CD45RA phenotype, although Ag-experienced memory tTregs are also detectable [8].

Fig. 1.

Fig. 1

FOXP3+ cells in healthy and pathologic conditions. a Healthy FOXP3+ regulatory T cells (Tregs) express the IL-2 receptor CD25, CTLA4 and the transcription factors STAT5 and FOXP3. Genetic abnormalities of these molecules are associated with Treg dysfunction: b FOXP3 mutations are causative for the immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome. c, d Mutations of CD25 and STAT5 are responsible for the development of immunodeficiency with autoimmunity related to IPEX. e Polymorphisms of CTLA4 have been associated with polyautoimmune pathologies. f Polymorphisms of the FOXP3 gene have been observed in allergic patients, suggesting a causative link

tTregs exert their function in different tissues, at sites of inflammation and in close contact with T effector (Teff) cells. Tregs use the same homing molecules used by naïve and Teff cells and thus home to sites of Teff generation and function [9]. More specifically, CCR7 has been shown to be important for Treg cell homing and function to the lymphoid compartment during the initiation of immune responses [10], while CCR4, CCR5, and CXCR3 are more relevant for Treg cell recruitment and function to peripheral sites of inflammation [11].

The generation of tTregs appears to be dependent on the strength of the binding between TCR and MHC class II peptides in mice [12]. In their work, Hsieh and colleagues cloned Tregs’ TCR into RAG deficient T cells, and subsequently demonstrated that the TCR-peptide avidity required for the generation of tTregs was higher than for the generation of Teff cells. This finding was confirmed in a more recent study [13]. Reducing the strength of the TCR–MHC interaction in mice leads to a decreased negative selection in the thymus, but also to an increase in the numbers of tTregs. This result suggests that the strength of interaction which induces Tregs would fall between that which induces clonal deletion and that which induces a conventional response [14].

FOXP3+ Tregs proliferate very poorly in vitro and do not produce cytokines, with the exception of low transforming growth factor-β (TGF-β) and IL-35 [15]. They are, however, very responsive to IL-2, which acts through its receptor and activation of STAT5. Indeed, IL-2 is the main factor for Treg survival and maintenance in vivo, and it is required at high doses for their in vitro expansion [16]. Other factors influence tTregs, such as TGF-β [17], thymic stromal lymphopoietin (TSLP) [18], and costimulatory molecules such as CD28 [19].

FOXP3 is a potent repressor of IL-2 production, but upregulates the expression of its receptor (CD25) and of the Treg marker CTLA4 [20]. Interestingly, FOXP3 also induces the expression of anti-inflammatory cytokine IL-10, as described in human tumor-associated Tregs, via a mechanism occurring in cooperation with the transcription factor STAT3 [21]. These data suggest that FOXP3 supports the maintenance of an immunosuppressive milieu. For a long time, FOXP3 was considered to be a master regulator for the development and function of Tregs because its absence in specific KO mice or in the natural mouse mutant, the scurfy mouse, is responsible for massive lymphoproliferation [22] and for a severe autoimmune syndrome. Mutations of FOXP3 lead to a similar phenotype in men [23] (Fig. 1b and see below, “tTregs in immune dysregulation” section). However, the establishment of a mice strain carrying a defective FoxP3 allele (knocked down by the insertion of a GFP cassette) showed that this gene is essential for the function but not for the development of tTregs. Rather, FOXP3 would potentiate pre-established Treg features, such as responsiveness to IL-2 [24]. Ectopic stable overexpression of FOXP3 by a lentivirus-based method in CD4+ T cells allows the generation of a stable population of human Treg-like cells, starting with na໱ve and memory CD4+ T cells, which are potent suppressor cells [26, 27]. In humans, Treg differentiation is characterized by specific demethylation of over a hundred of loci which become accessible for FOXP3 binding once it is expressed [25]. Importantly, the CpG methylation status of DNA regulates FoxP3 expression and accesses to its targets [28]. The regulation of FOXP3 expression occurs at both the transcriptional and translational levels. Three conserved non-coding sequences (CNS) have been identified at the FOXP3 locus [29, 30]. Their epigenetic state of DNA demethylation controls FOXP3 expression. Specifically, demethylation of the CNS2 (which is also called Treg-specific demethylated region—TSDR-) region is essential for inducing and stabilizing FOXP3 expression as it allows for FOXP3 to access its own locus and create a feedback loop that will ensure its stable expression [3032]. Activated Teff cells can also express FOXP3 but this expression is only transient and their TSDR remains methylated. TSDR demethylation is a very specific marker for tTreg, and its quantification is possible from blood and tissue samples [33, 34].

A study performed in 2012 in both mice and humans demonstrated that other genes show characteristic DNA demethylation specific for Treg phenotype. These genes include CTLA4, CD25, glucocorticoid-induced tumor necrosis factor-related receptor (GITR), and Eos and can be used to define the Treg population [28]. Previously, the lack of appropriate markers to differentiate tTregs from activated FOXP3+ Teff leads to many controversial results, especially in human studies of different pathologies such as in type 1 diabetes, multiple sclerosis, or rheumatoid arthritis. Depending on which markers were used to detect Treg, the authors would come to different conclusion. The combined use of these epigenetic markers in different pathologic conditions could provide important information on the state of the disease, response to therapy and still unravel new features on the role of Treg cells.

A new line of evidences indicate that tTregs present context-dependent functional diversity. Specific tTreg populations have been linked to the regulation of different T helper (Th) subclasses. As a whole, FOXP3 appears to cooperate with lineage-specific transcription factors in order to skew Treg abilities toward the regulation of diverse types of immune responses [35]. Several studies also indicate that specific subsets of human Treg cells could lose regulatory activity and become memory Teff cells producing IL-17, under specific conditions [36], or could convert to a prevalent Th2 phenotype [37]. In addition, ablation of FoxP3 in mature Tregs enables them to produce Th1 effector cytokines [38]. These observations are of particular importance in understanding the role of Treg cells in disease inflammatory context and the complex interactions with different Th subsets in allergy and autoimmunity.

FOXP3+ Tregs use various distinct mechanisms to exert their suppressive action [39]. They can use perforin and granzyme B to induce cytolysis of CTLs, B, and NK cells [4042]. However, these studies were performed in mice, and these properties need further investigations in human cells. Mouse and human Tregs can also use the tumor necrosis factor-related apoptosis inducing ligand (TRAIL)/death receptor 5 (DR5) pathway to induce cytolysis of T cells [43]. They can produce inhibitory cytokine such as IL-10, IL-35, and TGF-β that can further mediate their suppressive function (reviewed in [39, 44]) and modulate the adenosine nucleosides–adenosine receptor pathway to suppress Teff function in mice and human. The latter mechanism occurs by gap junction transfer of cyclic adenosine monophosphate, a potent inhibitor of proliferation and IL-2, to the target effector cells [4548]. Finally, in vivo visualization experiments have shown that the absence of Tregs increases the duration of the Teff-dendritic cell (DC) contacts in mice [49, 50]. This process is supposed to be dependent on Treg expression of CTLA4 [51].

Type 1 regulatory cells

Type 1 regulatory T (Tr1) cells are Ag-specific adaptive Tregs that are induced in the periphery upon chronic exposure to Ag in the presence of IL-10. They are defined by their cytokine response, namely their ability to produce high levels of IL-10 and TGF-β, low amounts of interferon γ (IFN-γ) and IL-2, and detectable levels of IL-5, in the absence of IL-4 [5254]. Unlike nTreg cells, Tr1 cells do not rely on FOXP3 expression for their function [5557]. Similar to other Treg cells, Tr1 cells are anergic, but their proliferation can be partially restored by IL-2 and IL-15, independently from TCR-mediated activation [52].

To specifically identify Tr1 cells, previous researchers have used a variety of transcriptional and membrane-bound markers. A comparative study of mouse models between Tr1 and tTregs cells showed that Tr1 cells selectively express repressor of GATA-3 (ROG) [58]. In humans, IL-10-producing T cells, which resemble Tr1 cells, selectively express the co-inhibitory molecule ICOS (inducible T cell co-stimulator) [59] or adhesion molecule CD18 [60]. Most recently, the markers LAG3 (lymphocyte-activation gene 3) and CD49b have been identified as cell-specific markers for murine and human Tr1 cells [61]. However, the specific transcription factor for this lineage specification is still unclear.

Tr1 cells are induced in an Ag-specific manner. Once activated through their specific TCR, Tr1 cells secrete IL-10 and TGF-β. These cytokines directly inhibit Teff proliferation, but also the expression of MHC class II and co-stimulatory molecules on Ag-presenting cells (APCs), thus indirectly suppressing Teff activation. In particular, IL-10 up-regulates the expression of the tolerogenic molecules immunoglobulin-like transcript-3 (ILT3), ILT4 [62, 63], and HLA-G [63, 64] on APCs, thus initiating a regulatory loop. IL-10-producing Tr1 cells have also been shown to directly suppress T cell responses via perforin and granzyme B production [63], cell-contact-dependent mechanisms [65], metabolic disruption [66], and mediate T cell suppression by selectively lysing myeloid cells [67]. Although Tr1 and FOXP3+ Tregs can act using similar mechanisms (e.g., production of TGF-β, perforin, granzyme B,…), the secretion of IL-10 is a hallmark of Tr1 suppressive activity [39, 65].

In addition to FOXP3+ Treg and Tr1, other Treg cells include Th3 cells, γδT cells, and CD8+ Treg cells [68]. The contribution of these cells to clinical tolerance is less well defined.

Regulatory T cells in pathologic conditions

Regulatory T cells and immune dysregulation of genetic origin

Genetic diseases of the immune system have been useful models to study immune functions. Primary immune deficiencies due to mutations of genes encoding for key molecules in lymphocyte functions have greatly improved our understanding of how immune responses are articulated. Similarly mutations in specific genes can also impair regulatory mechanisms [69].

A prototype of genetic disease with lack of peripheral tolerance is the immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome. Mutations of FOXP3 lead to the development of dysfunctional tTreg cells resulting in severe autoimmunity with very early onset (Fig. 1b) [70]. The main target organs are skin, intestine, and pancreas, manifesting with severe eczema, intractable diarrhea, and type I diabetes in the first months of life. IPEX syndrome is a life-threatening disease if not quickly recognized. The study of patients with IPEX syndrome helped recognize in humans that FOXP3 mutations do not impair the thymic development of Treg cells but rather their function [71]. The “programming” of tTreg is preserved as evidenced by complete demethylation at the TSDR and detectable FOXP3 expression, except in rare cases in which the mutation per se prevents the protein codification [72].

FOXP3-mutated tTregs have a growing disadvantage in a chimeric situation in which they co-exist with wild-type Treg, as it occurs in carrier mother or after bone marrow transplantations in IPEX patients [73, 74]. In addition, FOXP3-mutated tTreg have an unstable phenotype. Usually, tTregs are very poor cytokine producers because FOXP3 is a potent cytokine repressor. FOXP3-mutated cells loose this intrinsic regulation and produce effector cytokines, including IL-17, which is known to contribute to autoimmunity and inflammation [75, 76]. FOXP3-deficient tTreg cells are also inefficient at controlling B cell function, another relevant step in the pathogenesis of the disease [77]. Altered FOXP3 leads to ineffective control of Teff cell proliferation thus to lymphocytosis. Lack of this peripheral arm of the immune system is definitively detrimental and affects multiple cells and functions.

Interestingly, there are other molecules that when hit by genetic alterations can lead to Treg dysfunction. One example is CD25 deficiency (Fig. 1c), which is characterized by clinical manifestations similar to IPEX. Treg cells express high levels of CD25, and they are dependent on exogenous IL-2 for their peripheral survival [78]. In this IPEX-like disease, the impaired Treg function leads to autoimmunity that is associated with a profound immunodeficiency because of the inability of Teff cells to expand properly in response to pathogens. CD25-deficient patients are affected with severe CMV infections that can also induce severe enteropathy and autoimmune manifestations such as alopecia, thyroiditis, and cytopenia. tTreg cells of these patients normally express FOXP3 but they are less responsive to IL-2 and therefore less functional. Importantly, this disease is autosomal recessive and can also manifest in female, unlike IPEX which only affects males. Mutations in STAT5, the signaling molecule downstream CD25, can also impinge in the ability of Treg cells to survive in the periphery, to promptly respond to CD25 and TCR activation, and to upregulate FOXP3 expression (Fig. 1d) [79]. However, these patients usually have other symptoms such as growth defect and milder autoimmunity as compared to patients affected with IPEX, suggesting that other regulatory mechanisms could take place or that other pathways can partially replace the STAT5 intracellular defect.

Other gene mutations can alter tTreg mechanism of action. Those include adenosine deaminase mutations which are responsible for ADA-severe combined immunodeficiencies (ADA-SCID) impairing an important enzymatic pathway for the function of Treg [47] and Wiskott-Aldrich syndrome in which the mutated WAS protein, an essential component of the T cell synapse, alters a series of events following TCR-mediated signaling in multiple lineages including tTreg cells [80]. Although not described as a monogenic disease, several polymorphisms affecting CTLA4 have been associated with polyautoimmune symptoms which could be related to impaired function of Tregs (Fig. 1e) [81]. All these examples comprise rare diseases; however, as a group, early immunodysregulatory disorders involve a large number of patients with similar clinical manifestations and pathogenesis even though in many cases the underlying defect is still unrecognized. A quantitative defect of tTreg cells has been found in patients with IPEX-like symptoms of unknown origin with undetectable FOXP3 or CD25 mutations [72]. Interestingly, a proportion of them had reduced numbers of tTreg cells as detected on whole blood by measuring TSDR demethylation of FOXP3. The few Tregs present exerted normal in vitro suppressive function.

Considering other types of Treg cells, IL-10 and IL-10 receptor (IL-10R) gene mutations have been identified in patients with severe early onset ulcerative colitis [82]. Although Tr1 cells have not been investigated in these subjects, it is likely that a profound defect is occurring in this induced Treg subset given the complete dependence (demonstrated both in vitro and in vivo) of Tr1 cell generation from IL-10.

An emerging line of investigation aimed at understanding these disorders comprises a more systematic and efficient genome-wide analysis focused not only on identifying new causative genes but also hypomorphic mutations of already known genes that could determine a wide range of symptoms. In addition, it will be interesting to understand the interplay between the different types of Treg cells and how the genetic impairment of one can be replaced by expansion of another or they have mutual effects. Moreover, novel insights into the pathogenesis of immune dysregulation will be provided by studying Treg cells and their interaction with innate immunity cells such as innate lymphoid cells (ILCs) in different inflammatory environment.

Regulatory T cells in allergic disease

In addition to self-Ag, immune tolerance must be maintained throughout life against innocuous environmental peptides. Failure to do so may result in allergic reactions, the most important and best described type of which are IgE-mediated allergic diseases. This type of response is initiated when a naïve T cell recognizes a foreign peptide and is activated in the presence of IL-4 and other co-factors which trigger a Th2-type differentiation. Th2 cells release IL-4, IL-5, and IL-13 that stimulate B cells to produce allergen-specific IgE responsible for the acute symptoms. Th2 cytokines can also stimulate eosinophil development and recruitment, mucus production and smooth muscle contraction, and tissue homing of Th2 cells leading to chronic allergic inflammation in the skin and mucosal surfaces [8385]. In healthy responses, Tregs appear to be essential for maintenance of immune tolerance by preventing the Th2 induction and Th2 cytokine release in response to allergens [54].

A deregulation of FOXP3+ Tregs seems to play an important role in allergic diseases as well. Some evidence for example indicates that FOXP3 polymorphisms and impaired tTregs function have been associated with the development of allergy in different populations (Fig. 1f) [8688]. In addition, monozygotic twin studies in which one of the twins had allergic asthma showed the FOXP3 locus to be differentially methylated between the twin pairs [89]. In vitro function of FOXP3+ Tregs has been shown to be reduced in subjects with allergic rhinitis and refractive asthma, and Tregs from asthmatic subjects showed deregulation in chemokine signaling pathway [9097]. Most of the aforementioned studies looked at Tregs in blood. However, some allergic asthma studies looked at tTregs from bronchoalveolar fluid (BALF), an effective way to study tissue tTreg function in allergy. Consistent with previous studies on peripheral blood, asthmatic subjects were found to have lower FOXP3+ Tregs in BALF and their function was impaired [98, 99]. These findings were reversed with corticosteroid treatment and increased with allergen provocation [11, 98100] showing that migration of functional FOXP3+ Tregs is crucial to the prevention of allergic airway disease. The fact homing of Treg cells toward airway epithelial cells was shown to be impaired in chronic rhinosinusitis with nasal polyposis also supports this concept [101].

To study human FOXP3+ Treg in a humanized mouse model, Martin et al. [102] injected PBMCs from allergic or healthy donors into NOD/SCID mice, which were then challenged with allergens. After activation of Treg by treatment with the CD4-binding, lck-activating recombinant HIV-1 surface protein gp120, they found that PBMCs from healthy but not allergic donors prevented the development of allergen-induced airway inflammation and airway hyper-responsiveness and that treatment with gp120 attenuated both responses. These results further support the concept of a functional tTreg defect in allergic individuals.

Tr1 cells similarly suppress allergen-driven Th2 cytokine production from IL-4-producing Teff cells by releasing IL-10, TGF-β and expressing CTLA4 and PD-1. Their number has also been found to be reduced in peripheral blood from atopic subjects [103]. Furthermore, Meiler et al. [104] showed in a series of experiments that rapid switch and expansion of IL-10-producing Tr1 cells and the use of multiple suppressive factors represent essential mechanisms in immune tolerance to a high dose of allergens in non-allergic individuals. These studies suggest Tr1 cells also play a role in preventing allergic diseases but more research is needed on the role of this Treg subset.

Because the gut is a preferential tissue for homing of different kind of Treg cells, these cells are considered very important in the pathogenesis of food allergy and in defining new therapeutic interventions to control food allergy. Indeed, innovative therapies suggestive of restoration of tolerance are associated with increased proportion of either FOXP3+ Treg or IL-10-producing cells [105109]. In addition, oral immunotherapy against peanut allergen associated with an absence of response to the antigen after 3 months of therapy withdrawal induced a modification of FOXP3 methylation pattern and of tTreg activation [110].

Possible therapeutic interventions

Tregs are a powerful biological tool for immunomodulation. Much effort by several groups has been devoted to translate Treg therapy into the clinic. Two main approaches can be envisaged: (1) the use of pharmacological compounds that can boost Treg induction or expansion in vivo or (2) the in vitro expansion or in vitro induction, upon good manufacturing practices (GMP) conditions. Limitations have been encountered in both types of approach but the field is rapidly growing [111, 112]. As for in vivo induction, the problem of producing a selective effect on the target Treg cells has been the major challenge. The use of rapamycin has been found to selectively spare Treg cells and suppress Teff [113], and its use in different indications than organ transplantation should be assessed. Recently, novel modified small compounds of the IL-2/IL-2R complex which can partially mimic the effect of IL-2 but being selective for Treg cells are under investigation in preclinical studies and could lead to promising results [114, 115]. The in vivo expansion could represent an advantage over the in vitro expansion; however, both approaches would not allow selective action on Ag-specific Treg cells.

In allergic patients, immunotherapy has been proven to be beneficial and could be a very promising treatment for allergic diseases. It consists in the administration (subcutaneous, epicutaneous, sublingual, oral) of increasing doses of the pathogenic allergen, during a long period of time. The rational of this chronic Ag exposure is the induction of specific T cells which are not pathogenic but rather tolerogenic. After 1 year of allergen-specific immunotherapy, tTregs from these patients were increased numerically and functionally and showed specific demethylation of the FOXP3 locus [107]. However, the number of patients who remain tolerant to the Ag once the immunotherapy has stopped is still limited. Several complementary approaches are ongoing in order to render this strategy more efficacious, such as the parallel administration of anti-IgE Ab, allowing a faster Ag dose escalation [116].

The possibility of “producing” Treg from conventional T cells by in vitro induction has been the ambitious aim of many laboratories. From their discovery, Tr1 cells were known for being easily generated in vitro in an Ag-specific manner. This has been widely demonstrated in an allogenic system [117]. Up to now, two major proof-of-concept (POC) clinical trials have been performed with Ag-specific Tr1 cells and both considered the contribution of Ag-specific stimulation once the cells were re-injected in vivo. In a first trial, Crohn’s disease patients in which ovalbumin (OVA)-specific Tr1 cells were established in vitro and then infused in the patients who were fed with OVA in order to favor the gut homing of the infused Ag-specific cells and possibly their maintenance [118]. In a different POC trial, donor-derived Tr1 cells specific for the HLA Ag of the host were infused in patients affected with hematologic malignancies who were transplanted with haploidentical hematopoietic stem cells [119]. Safety was established and this Tr1 cell therapy improved the outcome of the transplant providing fast immune reconstitution and inducing tolerance across the allogeneic barrier. In both trials, the number of cells injected was a critical issue, and larger clinical trials in more homogenous cohort of patients are required to better assess the right cell dose to be administered.

A manufacturing protocol for establishing Ag-specific FOXP3+ Treg has been recently published [120] and could be applied in the future. However, polyclonal FOXP3+ Treg cells have been successfully used by different groups, as cell therapy in the context of allogeneic transplantation in order to prevent GVHD [121, 122]. In these protocols, Treg isolation was achieved by purification with beads-conjugated antibodies and cells were either infused fresh or upon a short period of expansion in vitro. In both cases, the purity of the cell population and therefore the stability and potency of suppression were critical. Encouraging positive results were obtained, although there have been concerns on the possible general suppression generated by Treg cells infusion. This issue could be avoided by administration of Ag-specific cells. Beside the allo-Ag system that has already been explored, differentiation of Treg cells toward a variety of different Ag could be now envisaged to cure patients with autoimmunity such as rheumatoid arthritis or patients affected with specific allergies.

An additional in vitro strategy for either Tr1 or FOXP3+ Treg generation consists in IL-10 or FOXP3 gene transfer into conventional T cells, respectively [27, 123]. Results obtained in vitro and in vivo in murine models of xeno GVHD show the feasibility of both approaches. While such a strategy would be mandatory in case of genetic mutations (for example, in IPEX patients), a large application in different pathologies remain to be assessed.

In conclusion, the therapeutic use of Treg cells could be achieved in several different ways, and there are good premises supporting the fact that implementation of Treg cell therapy will be of great benefit, alone or in combination with pharmacological compounds, in order to boost immune regulation, similarly to what has been achieved boosting immune responses with vaccinations.

Biography

graphic file with name nihms606190b1.gif

Kari C. Nadeau

References

  • 1.Xing Y, Hogquist KA. T-cell tolerance: central and peripheral. Cold Spring Harb Perspect Biol. 2012:4. doi: 10.1101/cshperspect.a006957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Abbas AK, Benoist C, Bluestone JA, Campbell DJ, Ghosh S, Hori S, Jiang S, Kuchroo VK, Mathis D, Roncarolo MG, et al. Regulatory T cells: recommendations to simplify the nomenclature. Nat Immunol. 2013;14:307–308. doi: 10.1038/ni.2554. [DOI] [PubMed] [Google Scholar]
  • 3.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155:1151–1164. [PubMed] [Google Scholar]
  • 4.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+ CD25+ regulatory T cells. Nat Immunol. 2003;4:330–336. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 5.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299:1057–1061. [PubMed] [Google Scholar]
  • 6.Sansom DM, Walker LS. The role of CD28 and cytotoxic T-lymphocyte antigen-4 (CTLA-4) in regulatory T-cell biology. Immunol Rev. 2006;212:131–148. doi: 10.1111/j.0105-2896.2006.00419.x. [DOI] [PubMed] [Google Scholar]
  • 7.Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P, Gingeras TR, de St Fazekas GB, et al. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. 2006;203:1701–1711. doi: 10.1084/jem.20060772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, Parizot C, Taflin C, Heike T, Valeyre D, 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]
  • 9.Bono MR, Elgueta R, Sauma D, Pino K, Osorio F, Michea P, Fierro A, Rosemblatt M. The essential role of chemokines in the selective regulation of lymphocyte homing. Cytokine Growth Factor Rev. 2007;18:33–43. doi: 10.1016/j.cytogfr.2007.01.004. [DOI] [PubMed] [Google Scholar]
  • 10.Forster R, Davalos-Misslitz AC, Rot A. CCR7 and its ligands: balancing immunity and tolerance. Nat Rev Immunol. 2008;8:362–371. doi: 10.1038/nri2297. [DOI] [PubMed] [Google Scholar]
  • 11.Afshar R, Strassner JP, Seung E, Causton B, Cho JL, Harris RS, Hamilos DL, Medoff BD, Luster AD. Compartmentalized che-mokine-dependent regulatory T-cell inhibition of allergic pulmonary inflammation. J Allergy Clin Immunol. 2013;131:1644–1652. doi: 10.1016/j.jaci.2013.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hsieh CS, Liang Y, Tyznik AJ, Self SG, Liggitt D, Rudensky AY. Recognition of the peripheral self by naturally arising CD25+ CD4+ T cell receptors. Immunity. 2004;21:267–277. doi: 10.1016/j.immuni.2004.07.009. [DOI] [PubMed] [Google Scholar]
  • 13.Moran AE, Holzapfel KL, Xing Y, Cunningham NR, Maltzman JS, Punt J, Hogquist KA. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J Exp Med. 2011;208:1279–1289. doi: 10.1084/jem.20110308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Hwang S, Song KD, Lesourne R, Lee J, Pinkhasov J, Li L, El-Khoury D, Love PE. Reduced TCR signaling potential impairs negative selection but does not result in autoimmune disease. J Exp Med. 2012;209:1781–1795. doi: 10.1084/jem.20120058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Levings MK, Roncarolo MG. Phenotypic and functional differences between human CD4+ CD25+ and type 1 regulatory T cells. Curr Top Microbiol Immunol. 2005;293:303–326. doi: 10.1007/3-540-27702-1_14. [DOI] [PubMed] [Google Scholar]
  • 16.Mahmud SA, Manlove LS, Farrar MA. Interleukin-2 and STAT5 in regulatory T cell development and function. JAK-STAT. 2013;2:e23154. doi: 10.4161/jkst.23154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fantini MC, Becker C, Monteleone G, Pallone F, Galle PR, Neurath MF. Cutting edge: TGF-beta induces a regulatory phenotype in CD4+ CD25- T cells through Foxp3 induction and down-regulation of Smad7. J Immunol. 2004;172:5149–5153. doi: 10.4049/jimmunol.172.9.5149. [DOI] [PubMed] [Google Scholar]
  • 18.Hanabuchi S, Ito T, Park WR, Watanabe N, Shaw JL, Roman E, Arima K, Wang YH, Voo KS, Cao W, et al. Thymic stromal lymphopoietin-activated plasmacytoid dendritic cells induce the generation of FOXP3+ regulatory T cells in human thymus. J Immunol. 2010;184:2999–3007. doi: 10.4049/jimmunol.0804106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Tai X, Cowan M, Feigenbaum L, Singer A. CD28 costimulation of developing thymocytes induces Foxp3 expression and regulatory T cell differentiation independently of interleukin 2. Nat Immunol. 2005;6:152–162. doi: 10.1038/ni1160. [DOI] [PubMed] [Google Scholar]
  • 20.Wu Y, Borde M, Heissmeyer V, Feuerer M, Lapan AD, Stroud JC, Bates DL, Guo L, Han A, Ziegler SF, et al. FOXP3 controls regulatory T cell function through cooperation with NFAT. Cell. 2006;126:375–387. doi: 10.1016/j.cell.2006.05.042. [DOI] [PubMed] [Google Scholar]
  • 21.Hossain DM, Panda AK, Manna A, Mohanty S, Bhattacharjee P, Bhattacharyya S, Saha T, Chakraborty S, Kar RK, Das T, et al. FoxP3 acts as a cotranscription factor with STAT3 in tumorinduced regulatory T cells. Immunity. 2013;39:1057–1069. doi: 10.1016/j.immuni.2013.11.005. [DOI] [PubMed] [Google Scholar]
  • 22.Komatsu N, Hori S. Full restoration of peripheral Foxp3+ regulatory T cell pool by radioresistant host cells in scurfy bone marrow chimeras. Proc Natl Acad Sci USA. 2007;104:8959–8964. doi: 10.1073/pnas.0702004104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, Kelly TE, Saulsbury FT, Chance PF, Ochs HD. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–21. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
  • 24.Gavin MA, Rasmussen JP, Fontenot JD, Vasta V, Manganiello VC, Beavo JA, Rudensky AY. Foxp3-dependent programme of regulatory T-cell differentiation. Nature. 2007;445:771–775. doi: 10.1038/nature05543. [DOI] [PubMed] [Google Scholar]
  • 25.Zhang Y, Maksimovic J, Naselli G, Qian J, Chopin M, Blewitt ME, Oshlack A, Harrison LC. Genome-wide DNA methylation analysis identifies hypomethylated genes regulated by FOXP3 in human regulatory T cells. Blood. 2013;122:2823–2836. doi: 10.1182/blood-2013-02-481788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Allan SE, Alstad AN, Merindol N, Crellin NK, Amendola M, Bacchetta R, Naldini L, Roncarolo MG, Soudeyns H, Levings MK. Generation of potent and stable human CD4+ T regulatory cells by activation-independent expression of FOXP3. Mol Ther. 2008;16:194–202. doi: 10.1038/sj.mt.6300341. [DOI] [PubMed] [Google Scholar]
  • 27.Passerini L, Mel ER, Sartirana C, Fousteri G, Bondanza A, Naldini L, Roncarolo MG, Bacchetta R. CD4+ T cells from IPEX patients convert into functional and stable regulatory T cells by FOXP3 gene transfer. Sci Transl Med. 2013;5 doi: 10.1126/scitranslmed.3007320. 215ra174. [DOI] [PubMed] [Google Scholar]
  • 28.Ohkura N, Hamaguchi M, Morikawa H, Sugimura K, Tanaka A, Ito Y, Osaki M, Tanaka Y, Yamashita R, Nakano N, et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity. 2012;37:785–799. doi: 10.1016/j.immuni.2012.09.010. [DOI] [PubMed] [Google Scholar]
  • 29.Passerini L, Santoni de Sio FR, Roncarolo MG, Bacchetta R. Forkhead box P3: the peacekeeper of the immune system. Int Rev Immunol. 2014;33(2):129–145. doi: 10.3109/08830185.2013.863303. [DOI] [PubMed] [Google Scholar]
  • 30.Zheng Y, Josefowicz S, Chaudhry A, Peng XP, Forbush K, Rudensky AY. Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature. 2010;463:808–812. doi: 10.1038/nature08750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kim HP, Leonard WJ. CREB/ATF-dependent T cell receptorinduced FoxP3 gene expression: a role for DNA methylation. J Exp Med. 2007;204:1543–1551. doi: 10.1084/jem.20070109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Polansky JK, Kretschmer K, Freyer J, Floess S, Garbe A, Baron U, Olek S, Hamann A, von Boehmer H, Huehn J. DNA methylation controls Foxp3 gene expression. Eur J Immunol. 2008;38:1654–1663. doi: 10.1002/eji.200838105. [DOI] [PubMed] [Google Scholar]
  • 33.Wieczorek G, Asemissen A, Model F, Turbachova I, Floess S, Liebenberg V, Baron U, Stauch D, Kotsch K, Pratschke J, et al. Quantitative DNA methylation analysis of FOXP3 as a new method for counting regulatory T cells in peripheral blood and solid tissue. Cancer Res. 2009;69:599–608. doi: 10.1158/0008-5472.CAN-08-2361. [DOI] [PubMed] [Google Scholar]
  • 34.Baron U, Floess S, Wieczorek G, Baumann K, Grutzkau A, Dong J, Thiel A, Boeld TJ, Hoffmann P, Edinger M, et al. DNA demethylation in the human FOXP3 locus discriminates regulatory T cells from activated FOXP3(+) conventional T cells. Eur J Immunol. 2007;37:2378–2389. doi: 10.1002/eji.200737594. [DOI] [PubMed] [Google Scholar]
  • 35.Oestreich KJ, Weinmann AS. Encoding stability versus flexibility: lessons learned from examining epigenetics in T helper cell differentiation. Curr Top Microbiol Immunol. 2012;356:145–164. doi: 10.1007/82_2011_141. [DOI] [PubMed] [Google Scholar]
  • 36.Zhou X, Bailey-Bucktrout S, Jeker LT, Bluestone JA. Plasticity of CD4(+) FoxP3(+) T cells. Curr Opin Immunol. 2009;21:281–285. doi: 10.1016/j.coi.2009.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhou X, Jeker LT, Fife BT, Zhu S, Anderson MS, McManus MT, Bluestone JA. Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity. J Exp Med. 2008;205:1983–1991. doi: 10.1084/jem.20080707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Williams LM, Rudensky AY. Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires continued expression of Foxp3. Nat Immunol. 2007;8:277–284. doi: 10.1038/ni1437. [DOI] [PubMed] [Google Scholar]
  • 39.Vignali DA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. 2008;8:523–532. doi: 10.1038/nri2343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zhao DM, Thornton AM, DiPaolo RJ, Shevach EM. Activated CD4+ CD25+ T cells selectively kill B lymphocytes. Blood. 2006;107:3925–3932. doi: 10.1182/blood-2005-11-4502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cao X, Cai SF, Fehniger TA, Song J, Collins LI, Piwnica-Worms DR, Ley TJ. Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity. 2007;27:635–646. doi: 10.1016/j.immuni.2007.08.014. [DOI] [PubMed] [Google Scholar]
  • 42.Loebbermann J, Thornton H, Durant L, Sparwasser T, Webster KE, Sprent J, Culley FJ, Johansson C, Openshaw PJ. Regulatory T cells expressing granzyme B play a critical role in controlling lung inflammation during acute viral infection. Mucosal Immunol. 2012;5:161–172. doi: 10.1038/mi.2011.62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ren X, Ye F, Jiang Z, Chu Y, Xiong S, Wang Y. Involvement of cellular death in TRAIL/DR5-dependent suppression induced by CD4(+) CD25(+) regulatory T cells. Cell Death Differ. 2007;14:2076–2084. doi: 10.1038/sj.cdd.4402220. [DOI] [PubMed] [Google Scholar]
  • 44.Collison LW, Pillai MR, Chaturvedi V, Vignali DA. Regulatory T cell suppression is potentiated by target T cells in a cell contact, IL-35- and IL-10-dependent manner. J Immunol. 2009;182:6121–6128. doi: 10.4049/jimmunol.0803646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204:1257–1265. doi: 10.1084/jem.20062512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hausler SF, Montalban del Barrio I, Strohschein J, Anoop Chandran P, Engel JB, Honig A, Ossadnik M, Horn E, Fischer B, Krockenberger M, et al. Ectonucleotidases CD39 and CD73 on OvCA cells are potent adenosine-generating enzymes responsible for adenosine receptor 2A-dependent suppression of T cell function and NK cell cytotoxicity. Cancer Immunol Im-munother. 2011;60:1405–1418. doi: 10.1007/s00262-011-1040-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sauer AV, Brigida I, Carriglio N, Aiuti A. Autoimmune dysregulation and purine metabolism in adenosine deaminase deficiency. Front Immunol. 2012;3:265. doi: 10.3389/fimmu.2012.00265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bopp T, Becker C, Klein M, Klein-Hessling S, Palmetshofer A, Serfling E, Heib V, Becker M, Kubach J, Schmitt S, et al. Cyclic adenosine monophosphate is a key component of regulatory T cell-mediated suppression. J Exp Med. 2007;204:1303–1310. doi: 10.1084/jem.20062129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tadokoro CE, Shakhar G, Shen S, Ding Y, Lino AC, Maraver A, Lafaille JJ, Dustin ML. Regulatory T cells inhibit stable contacts between CD4+ T cells and dendritic cells in vivo. J Exp Med. 2006;203:505–511. doi: 10.1084/jem.20050783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tang Q, Adams JY, Tooley AJ, Bi M, Fife BT, Serra P, Santamaria P, Locksley RM, Krummel MF, Bluestone JA. Visualizing regulatory T cell control of autoimmune responses in nonobese diabetic mice. Nat Immunol. 2006;7:83–92. doi: 10.1038/ni1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Oderup C, Cederbom L, Makowska A, Cilio CM, Ivars F. Cytotoxic T lymphocyte antigen-4-dependent down-modulation of costimulatory molecules on dendritic cells in CD4+ CD25+ regulatory T-cell-mediated suppression. Immunology. 2006;118:240–9. doi: 10.1111/j.1365-2567.2006.02362.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bacchetta R, Sartirana C, Levings MK, Bordignon C, Narula S, Roncarolo MG. Growth and expansion of human T regulatory type 1 cells are independent from TCR activation but require exogenous cytokines. Eur J Immunol. 2002;32:2237–2245. doi: 10.1002/1521-4141(200208)32:8<2237::AID-IMMU2237>3.0.CO;2-2. [DOI] [PubMed] [Google Scholar]
  • 53.Groux H, O’Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, Roncarolo MG. A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature. 1997;389:737–742. doi: 10.1038/39614. [DOI] [PubMed] [Google Scholar]
  • 54.Cottrez F, Hurst SD, Coffman RL, Groux H. T regulatory cells 1 inhibit a Th2-specific response in vivo. J Immunol. 2000;165:4848–4853. doi: 10.4049/jimmunol.165.9.4848. [DOI] [PubMed] [Google Scholar]
  • 55.Vieira PL, Christensen JR, Minaee S, O’Neill EJ, Barrat FJ, Boonstra A, Barthlott T, Stockinger B, Wraith DC, O’Garra A. IL-10-secreting regulatory T cells do not express Foxp3 but have comparable regulatory function to naturally occurring CD4+ CD25+ regulatory T cells. J Immunol. 2004;172:5986–93. doi: 10.4049/jimmunol.172.10.5986. [DOI] [PubMed] [Google Scholar]
  • 56.Levings MK, Gregori S, Tresoldi E, Cazzaniga S, Bonini C, Roncarolo MG. Differentiation of Tr1 cells by immature dendritic cells requires IL-10 but not CD25+ CD4+ Tr cells. Blood. 2005;105:1162–9. doi: 10.1182/blood-2004-03-1211. [DOI] [PubMed] [Google Scholar]
  • 57.Passerini L, Di Nunzio S, Gregori S, Gambineri E, Cecconi M, Seidel MG, Cazzola G, Perroni L, Tommasini A, Vignola S, et al. Functional type 1 regulatory T cells develop regardless of FOXP3 mutations in patients with IPEX syndrome. Eur J Immunol. 2011;41:1120–1131. doi: 10.1002/eji.201040909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Cobbold SP, Nolan KF, Graca L, Castejon R, Le Moine A, Frewin M, Humm S, Adams E, Thompson S, Zelenika D, et al. Regulatory T cells and dendritic cells in transplantation tolerance: molecular markers and mechanisms. Immunol Rev. 2003;196:109–124. doi: 10.1046/j.1600-065x.2003.00078.x. [DOI] [PubMed] [Google Scholar]
  • 59.Kohyama M, Sugahara D, Sugiyama S, Yagita H, Okumura K, Hozumi N. Inducible costimulator-dependent IL-10 production by regulatory T cells specific for self-antigen. Proc Natl Acad Sci USA. 2004;101:4192–4197. doi: 10.1073/pnas.0400214101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Rahmoun M, Foussat A, Groux H, Pene J, Yssel H, Chanez P. Enhanced frequency of CD18- and CD49b-expressing T cells in peripheral blood of asthmatic patients correlates with disease severity. Int Arch Allergy Immunol. 2006;140:139–149. doi: 10.1159/000092533. [DOI] [PubMed] [Google Scholar]
  • 61.Gagliani N, Magnani CF, Huber S, Gianolini ME, Pala M, Licona-Limon P, Guo B, Herbert DR, Bulfone A, Trentini F, et al. Coexpression of CD49b and LAG-3 identifies human and mouse T regulatory type 1 cells. Nat Med. 2013;19:739–746. doi: 10.1038/nm.3179. [DOI] [PubMed] [Google Scholar]
  • 62.Manavalan JS, Rossi PC, Vlad G, Piazza F, Yarilina A, Cortesini R, Mancini D, Suciu-Foca N. High expression of ILT3 and ILT4 is a general feature of tolerogenic dendritic cells. Transpl Immunol. 2003;11:245–258. doi: 10.1016/S0966-3274(03)00058-3. [DOI] [PubMed] [Google Scholar]
  • 63.Gregori S, Tomasoni D, Pacciani V, Scirpoli M, Battaglia M, Magnani CF, Hauben E, Roncarolo MG. Differentiation of type 1 T regulatory cells (Tr1) by tolerogenic DC-10 requires the IL-10-dependent ILT4/HLA-G pathway. Blood. 2010;116:935–944. doi: 10.1182/blood-2009-07-234872. [DOI] [PubMed] [Google Scholar]
  • 64.Moreau P, Adrian-Cabestre F, Menier C, Guiard V, Gourand L, Dausset J, Carosella ED, Paul P. IL-10 selectively induces HLA-G expression in human trophoblasts and monocytes. Int Immunol. 1999;11:803–811. doi: 10.1093/intimm/11.5.803. [DOI] [PubMed] [Google Scholar]
  • 65.Gregori S, Goudy KS, Roncarolo MG. The cellular and molecular mechanisms of immuno-suppression by human type 1 regulatory T cells. Front Immunol. 2012;3:30. doi: 10.3389/fimmu.2012.00030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mandapathil M, Szczepanski MJ, Szajnik M, Ren J, Jackson EK, Johnson JT, Gorelik E, Lang S, Whiteside TL. Adenosine and prostaglandin E2 cooperate in the suppression of immune responses mediated by adaptive regulatory T cells. J Biol Chem. 2010;285:27571–27580. doi: 10.1074/jbc.M110.127100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Magnani CF, Alberigo G, Bacchetta R, Serafini G, Andreani M, Roncarolo MG, Gregori S. Killing of myeloid APCs via HLA class I, CD2 and CD226 defines a novel mechanism of suppression by human Tr1 cells. Eur J Immunol. 2011;41:1652–1662. doi: 10.1002/eji.201041120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Shalev I, Schmelzle M, Robson SC, Levy G. Making sense of regulatory T cell suppressive function. Semin Immunol. 2011;23:282–292. doi: 10.1016/j.smim.2011.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Bacchetta R, Notarangelo LD. Immunodeficiency with autoimmunity: beyond the paradox. Front Immunol. 2013;4:77. doi: 10.3389/fimmu.2013.00077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Barzaghi F, Passerini L, Bacchetta R. Immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome: a paradigm of immunodeficiency with autoimmunity. Front Immunol. 2012;3:211. doi: 10.3389/fimmu.2012.00211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bacchetta R, Passerini L, Gambineri E, Dai M, Allan SE, Perroni L, Dagna-Bricarelli F, Sartirana C, Matthes-Martin S, Lawitschka A, et al. Defective regulatory and effector T cell functions in patients with FOXP3 mutations. J Clin Invest. 2006;116:1713–1722. doi: 10.1172/JCI25112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Barzaghi F, Passerini L, Gambineri E, Ciullini Mannurita S, Cornu T, Kang ES, Choe YH, Cancrini C, Corrente S, Ciccocioppo R, et al. Demethylation analysis of the FOXP3 locus shows quantitative defects of regulatory T cells in IPEX-like syndrome. J Autoimmun. 2012;38:49–58. doi: 10.1016/j.jaut.2011.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Di Nunzio S, Cecconi M, Passerini L, McMurchy AN, Baron U, Turbachova I, Vignola S, Valencic E, Tommasini A, Junker A, et al. Wild-type FOXP3 is selectively active in CD4+ CD25(hi) regulatory T cells of healthy female carriers of different FOXP3 mutations. Blood. 2009;114:4138–41. doi: 10.1182/blood-2009-04-214593. [DOI] [PubMed] [Google Scholar]
  • 74.Seidel MG, Fritsch G, Lion T, Jurgens B, Heitger A, Bacchetta R, Lawitschka A, Peters C, Gadner H, Matthes-Martin S. Selective engraftment of donor CD4+ 25high FOXP3-positive T cells in IPEX syndrome after nonmyeloablative hematopoietic stem cell transplantation. Blood. 2009;113:5689–5691. doi: 10.1182/blood-2009-02-206359. [DOI] [PubMed] [Google Scholar]
  • 75.Passerini L, Olek S, Di Nunzio S, Barzaghi F, Hambleton S, Abinun M, Tommasini A, Vignola S, Cipolli M, Amendola M, et al. Forkhead box protein 3 (FOXP3) mutations lead to increased TH17 cell numbers and regulatory T-cell instability. J Allergy Clin Immunol. 2011;128(1376–1379):e1371. doi: 10.1016/j.jaci.2011.09.010. [DOI] [PubMed] [Google Scholar]
  • 76.Marwaha AK, Leung NJ, McMurchy AN, Levings MK. TH17 Cells in autoimmunity and immunodeficiency: protective or pathogenic+ Front Immunol. 2012;3:129. doi: 10.3389/fimmu.2012.00129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kinnunen T, Chamberlain N, Morbach H, Choi J, Kim S, Craft J, Mayer L, Cancrini C, Passerini L, Bacchetta R, et al. Accumulation of peripheral autoreactive B cells in the absence of functional human regulatory T cells. Blood. 2013;121:1595–603. doi: 10.1182/blood-2012-09-457465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Goudy K, Aydin D, Barzaghi F, Gambineri E, Vignoli M, Ciullini Mannurita S, Doglioni C, Ponzoni M, Cicalese MP, Assanelli A, et al. Human IL2RA null mutation mediates immunodeficiency with lymphoproliferation and autoimmunity. Clin Immunol. 2013;146:248–261. doi: 10.1016/j.clim.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Jenks JA, Seki S, Kanai T, Huang J, Morgan AA, Scalco RC, Nath R, Bucayu R, Wit JM, Al-Herz W, et al. Differentiating the roles of STAT5B and STAT5A in human CD4+ T cells. Clin Immunol. 2013;148:227–236. doi: 10.1016/j.clim.2013.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Catucci M, Castiello MC, Pala F, Bosticardo M, Villa A. Autoimmunity in wiskott-Aldrich syndrome: an unsolved enigma. Front Immunol. 2012;3:209. doi: 10.3389/fimmu.2012.00209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol Rev. 2008;224:166–182. doi: 10.1111/j.1600-065X.2008.00662.x. [DOI] [PubMed] [Google Scholar]
  • 82.Glocker EO, Kotlarz D, Boztug K, Gertz EM, Schaffer AA, Noyan F, Perro M, Diestelhorst J, Allroth A, Murugan D, et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 2009;361:2033–2045. doi: 10.1056/NEJMoa0907206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Mosmann TR, Sad S. The expanding universe of T-cell subsets: Th1, Th2 and more. Immunol Today. 1996;17:138–146. doi: 10.1016/0167-5699(96)80606-2. [DOI] [PubMed] [Google Scholar]
  • 84.Akdis CA, Blaser K, Akdis M. Apoptosis in tissue inflammation and allergic disease. Curr Opin Immunol. 2004;16:717–723. doi: 10.1016/j.coi.2004.09.004. [DOI] [PubMed] [Google Scholar]
  • 85.Ying S, Meng Q, Barata LT, Robinson DS, Durham SR, Kay AB. Associations between IL-13 and IL-4 (mRNA and protein), vascular cell adhesion molecule-1 expression, and the infiltration of eosinophils, macrophages, and T cells in allergeninduced late-phase cutaneous reactions in atopic subjects. J Immunol. 1997;158:5050–5057. [PubMed] [Google Scholar]
  • 86.Zhang L, Zhang Y, Desrosiers M, Wang C, Zhao Y, Han D. Genetic association study of FOXP3 polymorphisms in allergic rhinitis in a Chinese population. Hum Immunol. 2009;70:930–934. doi: 10.1016/j.humimm.2009.08.001. [DOI] [PubMed] [Google Scholar]
  • 87.Bottema RW, Kerkhof M, Reijmerink NE, Koppelman GH, Thijs C, Stelma FF, Smit HA, Brunekreef B, van Schayck CP, Postma DS. X-chromosome Forkhead box P3 polymorphisms associate with atopy in girls in three Dutch birth cohorts. Allergy. 2010;65:865–874. doi: 10.1111/j.1398-9995.2009.02291.x. [DOI] [PubMed] [Google Scholar]
  • 88.Fodor E, Garaczi E, Polyanka H, Koreck A, Kemeny L, Szell M. The rs3761548 polymorphism of FOXP3 is a protective genetic factor against allergic rhinitis in the Hungarian female population. Hum Immunol. 2011;72:926–929. doi: 10.1016/j.humimm.2011.06.011. [DOI] [PubMed] [Google Scholar]
  • 89.Runyon RS, Cachola LM, Rajeshuni N, Hunter T, Garcia M, Ahn R, Lurmann F, Krasnow R, Jack LM, Miller RL, et al. Asthma discordance in twins is linked to epigenetic modifications of T cells. PLoS One. 2012;7:e48796. doi: 10.1371/journal.pone.0048796. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Grindebacke H, Wing K, Andersson AC, Suri-Payer E, Rak S, Rudin A. Defective suppression of Th2 cytokines by CD4CD25 regulatory T cells in birch allergics during birch pollen season. Clin Exp Allergy. 2004;34:1364–1372. doi: 10.1111/j.1365-2222.2004.02067.x. [DOI] [PubMed] [Google Scholar]
  • 91.Ling EM, Smith T, Nguyen XD, Pridgeon C, Dallman M, Arbery J, Carr VA, Robinson DS. Relation of CD4+ CD25+ regulatory T-cell suppression of allergen-driven T-cell activation to atopic status and expression of allergic disease. Lancet. 2004;363:608–615. doi: 10.1016/S0140-6736(04)15592-X. [DOI] [PubMed] [Google Scholar]
  • 92.Kinoshita T, Baatjes A, Smith SG, Dua B, Watson R, Kawayama T, Larche M, Gauvreau GM, O’Byrne PM. Natural regulatory T cells in isolated early responders compared with dual responders with allergic asthma. J Allergy Clin Immunol. 2014;133(3):696–703. doi: 10.1016/j.jaci.2013.08.025. [DOI] [PubMed] [Google Scholar]
  • 93.Lee JH, Yu HH, Wang LC, Yang YH, Lin YT, Chiang BL. The levels of CD4+ CD25+ regulatory T cells in paediatric patients with allergic rhinitis and bronchial asthma. Clin Exp Immunol. 2007;148:53–63. doi: 10.1111/j.1365-2249.2007.03329.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lin YL, Shieh CC, Wang JY. The functional insufficiency of human CD4+ CD25 high T-regulatory cells in allergic asthma is subjected to TNF-alpha modulation. Allergy. 2008;63:67–74. doi: 10.1111/j.1398-9995.2007.01526.x. [DOI] [PubMed] [Google Scholar]
  • 95.Moniuszko M, Kowal K, Zukowski S, Dabrowska M, Bodzenta-Lukaszyk A. Frequencies of circulating CD4+ CD25+ CD127low cells in atopics are altered by bronchial allergen challenge. Eur J Clin Invest. 2008;38:201–204. doi: 10.1111/j.1365-2362.2007.01920.x. [DOI] [PubMed] [Google Scholar]
  • 96.Nguyen KD, Fohner A, Booker JD, Dong C, Krensky AM, Nadeau KC. XCL1 enhances regulatory activities of CD4+ CD25(high) CD127(low/−) T cells in human allergic asthma. J Immunol. 2008;181:5386–5395. doi: 10.4049/jimmunol.181.8.5386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Nguyen KD, Vanichsarn C, Fohner A, Nadeau KC. Selective deregulation in chemokine signaling pathways of CD4+ CD25(hi)CD127(lo)/(−) regulatory T cells in human allergic asthma. J Allergy Clin Immunol. 2009;123(933–939):e910. doi: 10.1016/j.jaci.2008.11.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Hartl D, Koller B, Mehlhorn AT, Reinhardt D, Nicolai T, Schendel DJ, Griese M, Krauss-Etschmann S. Quantitative and functional impairment of pulmonary CD4+ CD25hi regulatory T cells in pediatric asthma. J Allergy Clin Immunol. 2007;119:1258–1266. doi: 10.1016/j.jaci.2007.02.023. [DOI] [PubMed] [Google Scholar]
  • 99.Thunberg S, Gafvelin G, Nord M, Gronneberg R, Grunewald J, Eklund A, van Hage M. Allergen provocation increases TH2-cytokines and FOXP3 expression in the asthmatic lung. Allergy. 2010;65:311–318. doi: 10.1111/j.1398-9995.2009.02218.x. [DOI] [PubMed] [Google Scholar]
  • 100.Smyth LJ, Eustace A, Kolsum U, Blaikely J, Singh D. Increased airway T regulatory cells in asthmatic subjects. Chest. 2010;138:905–912. doi: 10.1378/chest.09-3079. [DOI] [PubMed] [Google Scholar]
  • 101.Kim YM, Munoz A, Hwang PH, Nadeau KC. Migration of regulatory T cells toward airway epithelial cells is impaired in chronic rhinosinusitis with nasal polyposis. Clin Immunol. 2010;137:111–121. doi: 10.1016/j.clim.2010.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Martin H, Reuter S, Dehzad N, Heinz A, Bellinghausen I, Saloga J, Haasler I, Korn S, Jonuleit H, Buhl R, et al. CD4-mediated regulatory T-cell activation inhibits the development of disease in a humanized mouse model of allergic airway disease. J Allergy Clin Immunol. 2012;129:521–528. doi: 10.1016/j.jaci.2011.09.038. [DOI] [PubMed] [Google Scholar]
  • 103.Akdis M, Verhagen J, Taylor A, Karamloo F, Karagiannidis C, Crameri R, Thunberg S, Deniz G, Valenta R, Fiebig H, et al. Immune responses in healthy and allergic individuals are characterized by a fine balance between allergen-specific T regulatory 1 and T helper 2 cells. J Exp Med. 2004;199:1567–1575. doi: 10.1084/jem.20032058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Meiler F, Zumkehr J, Klunker S, Ruckert B, Akdis CA, Akdis M. In vivo switch to IL-10-secreting T regulatory cells in high dose allergen exposure. J Exp Med. 2008;205:2887–2898. doi: 10.1084/jem.20080193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Jones SM, Pons L, Roberts JL, Scurlock AM, Perry TT, Kulis M, Shreffler WG, Steele P, Henry KA, Adair M, et al. Clinical efficacy and immune regulation with peanut oral immunotherapy. J Allergy Clin Immunol. 2009;124:292–300. doi: 10.1016/j.jaci.2009.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Scadding GW, Shamji MH, Jacobson MR, Lee DI, Wilson D, Lima MT, Pitkin L, Pilette C, Nouri-Aria K, Durham SR. Sublingual grass pollen immunotherapy is associated with increases in sublingual Foxp3-expressing cells and elevated allergen-specific immunoglobulin G4, immunoglobulin A and serum inhibitory activity for immunoglobulin E-facilitated allergen binding to B cells. Clin Exp Allergy. 2010;40:598–606. doi: 10.1111/j.1365-2222.2010.03462.x. [DOI] [PubMed] [Google Scholar]
  • 107.Swamy RS, Reshamwala N, Hunter T, Vissamsetti S, Santos CB, Baroody FM, Hwang PH, Hoyte EG, Garcia MA, Nadeau KC. Epigenetic modifications and improved regulatory T-cell function in subjects undergoing dual sublingual immunotherapy. J Allergy Clin Immunol. 2012;130(215–224):e217. doi: 10.1016/j.jaci.2012.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Jutel M, Akdis M, Blaser K, Akdis CA. Are regulatory T cells the target of venom immunotherapy+ Curr Opin Allergy Clin Immunol. 2005;5:365–369. doi: 10.1097/01.all.0000173784.81024.7a. [DOI] [PubMed] [Google Scholar]
  • 109.Syed A, Kohli A, Nadeau KC. Food allergy diagnosis and therapy: where are we now+ Immunotherapy. 2013;5:931–944. doi: 10.2217/imt.13.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Syed A, Garcia MA, Lyu SC, Bucayu R, Kohli A, Ishida S, Berglund JP, Tsai M, Maecker H, O’Riordan G, et al. Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3) J Allergy Clin Immunol. 2014;133(500–510):e511. doi: 10.1016/j.jaci.2013.12.1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Fischbach MA, Bluestone JA, Lim WA. Cell-based therapeutics: the next pillar of medicine. Sci Transl Med. 2013;5 doi: 10.1126/scitranslmed.3005568. 179ps177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Schneidawind D, Pierini A, Negrin RS. Regulatory T cells and natural killer T cells for modulation of GVHD following allogeneic hematopoietic cell transplantation. Blood. 2013;122:3116–3121. doi: 10.1182/blood-2013-08-453126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Battaglia M, Stabilini A, Migliavacca B, Horejs-Hoeck J, Kaupper T, Roncarolo MG. Rapamycin promotes expansion of functional CD4+ CD25+ FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients. J Immunol. 2006;177:8338–8347. doi: 10.4049/jimmunol.177.12.8338. [DOI] [PubMed] [Google Scholar]
  • 114.Webster KE, Walters S, Kohler RE, Mrkvan T, Boyman O, Surh CD, Grey ST, Sprent J. In vivo expansion of T reg cells with IL-2-mAb complexes: induction of resistance to EAE and long-term acceptance of islet allografts without immunosuppression. J Exp Med. 2009;206:751–760. doi: 10.1084/jem.20082824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Shatry A, Levy RB. In situ activation and expansion of host tregs: a new approach to enhance donor chimerism and stable engraftment in major histocompatibility complex-matched allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2009;15:785–794. doi: 10.1016/j.bbmt.2009.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Nadeau KC, Kohli A, Iyengar S, DeKruyff RH, Umetsu DT. Oral immunotherapy and anti-IgE antibody-adjunctive treatment for food allergy. Immunol Allergy Clin North Am. 2012;32:111–133. doi: 10.1016/j.iac.2011.11.004. [DOI] [PubMed] [Google Scholar]
  • 117.Bacchetta R, Gregori S, Serafini G, Sartirana C, Schulz U, Zino E, Tomiuk S, Jansen U, Ponzoni M, Paties CT, et al. Molecular and functional characterization of allogantigen-specific anergic T cells suitable for cell therapy. Haematologica. 2010;95:2134–2143. doi: 10.3324/haematol.2010.025825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Desreumaux P, Foussat A, Allez M, Beaugerie L, Hebuterne X, Bouhnik Y, Nachury M, Brun V, Bastian H, Belmonte N, et al. Safety and efficacy of antigen-specific regulatory T-cell therapy for patients with refractory Crohn’s disease. Gastroenterology. 2012;143(1207–1217):e1201–e1202. doi: 10.1053/j.gastro.2012.07.116. [DOI] [PubMed] [Google Scholar]
  • 119.Bacchetta R, Lucarrelli B, Sartirana C, Gregori S, Stanghellini L, Terasa M, Miqueu P, Tomiuk S, Hernandez-Fuentes M, Gianolini ME, Greco R, Bernardi M, Zappone E, Rossini S, Janssen U, Ambrosi A, Salomoni M, Peccatori J, Ciceri F, Roncarolo MG. Immunological outcome in haploidentical-HSC transplanted patients treated with IL-10-anergized donor T cells. Front Immunol. 2014;5:16. doi: 10.3389/fimmu.2014.00016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Putnam AL, Safinia N, Medvec A, Laszkowska M, Wray M, Mintz MA, Trotta E, Szot GL, Liu W, Lares A, et al. Clinical grade manufacturing of human alloantigen-reactive regulatory T cells for use in transplantation. Am J Transplant. 2013;13:3010–3020. doi: 10.1111/ajt.12433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Sawitzki B, Brunstein C, Meisel C, Schumann J, Vogt K, Appelt C, Curtsinger JM, Verneris MR, Miller JS, Wagner JE, et al. Prevention of graft-versus-host disease by adoptive T regulatory therapy is associated with active repression of peripheral blood toll-like receptor 5 mRNA expression. Biol Blood Marrow Transplant. 2014;20(2):173–182. doi: 10.1016/j.bbmt.2013.10.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Brunstein CG, Fuchs EJ, Carter SL, Karanes C, Costa LJ, Wu J, Devine SM, Wingard JR, Aljitawi OS, Cutler CS, et al. Alternative donor transplantation after reduced intensity conditioning: results of parallel phase 2 trials using partially HLA-mismatched related bone marrow or unrelated double umbilical cord blood grafts. Blood. 2011;118:282–288. doi: 10.1182/blood-2011-03-344853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Andolfi G, Fousteri G, Rossetti M, Magnani CF, Jofra T, Locafaro G, Bondanza A, Gregori S, Roncarolo MG. Enforced IL-10 expression confers type 1 regulatory T cell (Tr1) phenotype and function to human CD4(+) T cells. Mol Ther. 2012;20:1778–1790. doi: 10.1038/mt.2012.71. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES