Skip to main content
Self Nonself logoLink to Self Nonself
. 2011 Jan 1;2(1):26–34. doi: 10.4161/self.2.1.15422

Transplantation tolerance

Clinical potential of regulatory T cells

Yannick D Muller 1,2, Jörg D Seebach 2, Leo H Bühler 1, Manuel Pascual 3, Dela Golshayan 3,
PMCID: PMC3136901  PMID: 21776332

Abstract

The major challenge in transplantation medicine remains long-term allograft acceptance, with preserved allograft function under minimal chronic immunosuppression. To safely achieve the goal of sustained donor-specific T and B cell non-responsiveness, research efforts are now focusing on therapies based on cell subsets with regulatory properties. In particular the transfusion of human regulatory T cells (Treg) is currently being evaluated in phase I/II clinical trials for the treatment of graft versus host disease following hematopoietic stem cell transplantation, and is also under consideration for solid organ transplantation. The purpose of this review is to recapitulate current knowledge on naturally occurring as well as induced human Treg, with emphasis on their specific phenotype, suppressive function and how these cells can be manipulated in vitro and/or in vivo for therapeutic purposes in transplantation medicine. We highlight the potential but also possible limitations of Treg-based strategies to promote long-term allograft survival. It is evident that the bench-to-beside translation of these protocols still requires further understanding of Treg biology. Nevertheless, current data already suggest that Treg therapy alone will not be sufficient and needs to be combined with other immunomodulatory approaches in order to induce allograft tolerance.

Key words: transplantation, tolerance, regulatory T cells, cell therapy

Introduction

Following on the pioneering work of Medawar and colleagues more than 50 years ago, extensive data obtained in rodents and large animal experimental transplantation models have led to a better understanding of the mechanisms leading to graft rejection and transplantation tolerance.1,2 The advent of powerful immunosuppressive drugs that can control the rejection process since the mid 1980s has allowed the development of clinical transplantation with increasing success. Consequently, solid organ transplantation (SOT) has become the therapy of choice for end-stage organ diseases. Patient and allograft survival, as well as rates of acute allograft rejection episodes in the first year after transplantation have steadily improved. In contrast, concerns remain on the long-term outcome. Besides morbidity and mortality associated with chronic immune suppression and drug exposure, there is an inexorable loss of transplanted organs due to chronic allograft dysfunction.3,4 This process is mediated by both immunological (inadequate immunosuppression) and non-immunological (drug-related toxicity) factors.5 Thus, the ultimate goal in transplantation medicine remains the induction of specific tolerance to donor alloantigens and long-term graft acceptance with minimal immunosuppressive drug exposure. In this perspective, the development of tolerogenic protocols and in particular of regulatory cell-based therapies has stimulated much interest in recent years. In this review, we discuss the mechanisms of immune tolerance to self-antigens and how they could be exploited in the transplantation setting. In particular we scrutinize current knowledge on regulatory T cells (Treg) and their potential for cell-based immunotherapy in SOT.

Central and Peripheral Tolerance

The recognition of the allograft major histocompatibility complex (MHC)-mismatched antigens by circulating alloreactive T cells is the primary event that ultimately leads to graft rejection. T cells are therefore the main target of current immunomodulatory strategies. Transplantation tolerance is defined by a state of sustained donor-specific T and B cell non-responsiveness with preserved graft function, with no (operational tolerance) or only minimal (near-tolerance) chronic immunosuppressive drugs. The immune system is capable of distinguishing between self- and non-self antigens, leading to specific protective cell-mediated and humoral responses in the absence of autoimmunity. Several different experimental protocols were successful in inducing transplant tolerance to in rodents and large animals, demonstrating the possibility to exploit the mechanisms that normally maintain immune homeostasis and tolerance to self-antigens to induce tolerance to alloantigens. Furthermore, clinical reports of occasional “tolerant” recipients who had stopped taking their immunosuppressive drugs but still experienced prolonged allograft survival, suggest that immunological tolerance may indeed be a goal that can be achieved in transplantation medicine.69

Immune tolerance is principally mediated via central and peripheral mechanisms. Central tolerance normally leads to the intrathymic deletion of T cells recognizing thymus-expressed autoantigens with high avidity, so that potentially deleterious antigen-reactive T cells will not reach the periphery. Since the early observations by Medawar and colleagues,1,10 a large number of experimental as well as clinical studies have confirmed that central tolerance against alloantigens can be achieved in transplant recipients by the induction of full (donor cells reconstituting recipient's hematopoietic compartment) or mixed (donor cells coexisting with recipient cells) hematopoietic chimerism.1116 In these settings, cells of donor origin, either spontaneously released by the allograft (liver transplantation) or infused in therapeutic protocols (hematopoietic stem cells, HSCT or bone marrow transplantation, BMT), can migrate to the recipient's thymus and induce clonal deletion of donor-reactive host T cells. However, to achieve donor cell engraftment and full or even mixed hematopoietic chimerism in recipients with a fully functional immune system, some degree of cytoreductive conditioning is needed, unless very high doses of donor stem cells or BM are administered. Thus, these approaches are relatively toxic and not easily feasible, precluding their application in routine clinical practice. Furthermore, although encouraging data have been obtained in pre-clinical studies (non-human primates, pigs),17,18 and some small trials in humans,1416 in most cases mixed chimerism-based strategies have been difficult to translate successfully to large animal models. This indicates that central deletion may not be the only mechanism involved in the induction of robust tolerance in humans.

As not all antigens are expressed in the thymus (for instance tissue-specific antigens) and therefore evade central tolerance, peripheral tolerance usually prevents auto-immunity by various mechanisms. Peripheral tolerance to self- and non-self antigens can be achieved by deletion of activated T cells, T cell anergy and active regulation.19 The encounter of naive T cells with antigen-presenting cells (APC), mainly DC, modulates their differentiation into various subsets of polarized effector and regulatory T cells and is a major component in the nature of T-cell responsiveness. In particular, the type of DC and the local cytokine microenvironment determine the outcome of the immune response towards immunity or tolerance. Thus, alloreactive effector T cells could be either controlled by other cells with regulatory/suppressive functions or by modulating the immunogenicity of APC and the local cytokine milieu.

Increasing evidence suggest the existence of interactions between central and peripheral mechanisms to promote the induction and maintenance of transplantation tolerance. Indeed, the thymus is not only responsible for clonal deletion of autoreactive T cells but also for active peripheral regulation through the generation of natural Treg. Moreover, peripheral DC can migrate to the thymus to present donor-derived antigens and therefore participate in the education of the immune system towards alloantigens and the induction of central tolerance.20

Regulatory T Cells

Regulatory cells are defined by their functional ability to suppress immune responses. This concept was initially proposed following the observation in experimental transplantation models that donor-specific transplantation tolerance could be transferred to a new naïve host via the adoptive transfer of cells. In the 1970s, Gershon first described thymus-derived cells in mice that suppressed the response to an antigenic challenge with the injection of sheep red blood cells. He then developed the concepts of “infectious tolerance” and “suppressor cells”.21 However, due to poor experimental reproducibility in vitro, the “suppressor cells” fell into disrepute, although transferable tolerance remained a robust concept in vivo.22 Convincing evidence for the existence of Treg came in the mid 1980s from experiments in rodent models of autoimmune disease.2325 Further experiments by Sakaguchi and colleagues allowed the identification of a subset of naturally occurring CD4+ T cells with constitutive expression of the IL-2 receptor α-chain (CD25) that was essential for the prevention of autoimmunity.26 Since these early data, a huge body of literature has accumulated showing that CD4+CD25+ Treg are involved in the control of a wide variety of immune responses. Besides the naturally occurring CD4+CD25+ Treg that have been identified in non-manipulated animals and humans, uncommitted naïve CD4+ T cells can be skewed towards T cells with regulatory functions in the peripheral immune system under specific in vitro or in vivo conditions.

Foxp3+ T Regulatory Cells

Naturally occurring CD4+CD25+Foxp3+ Treg (nTreg) derive from the thymus and constitute 3–10% of the naïve peripheral CD4+ T cell population in humans. As CD25 is also upregulated on the surface of activated effector T cells, other specific markers are needed to identify nTreg. To date, the best marker of nTreg is the intracellular expression of the transcription factor forkhead box P3 (Foxp3). Foxp3, the cytokine IL-2 as well as CD25 as a component of the IL-2 receptor, are essential for the development, function and survival of nTreg, because mutations or polymorphisms in the genes encoding these molecules predispose to autoimmune or lymphoproliferative diseases.2730 Furthermore, the specific role of Foxp3 in the function of nTreg was highlighted by the fact that retroviral transduction of Foxp3 into CD4+CD25 T cells converts them to functional Treg that are able to suppress proliferation of other T cells in vitro and inhibit the development of autoimmune diseases mediated by pathogenic effector T cells in in vivo experimental models.31 It was also shown that sustained Foxp3 expression is required to confer the suppressive capacity of Treg. While Foxp3 expression constitutes a lineage specification of bonafide nTreg in mice, human activated effector T cells can transiently express Foxp3.32,33 Thus, more specific markers for human nTreg are still sought-after and the precise molecular role of Foxp3 in nTreg remains to be fully understood.

Human naturally occurring Treg.

As Foxp3 expression is not a reliable phenotype in human nTreg and cannot be used as a selection marker to isolate living cells due to its intracellular expression, numerous surface markers have been identified to help discriminate between activated effector T cells and nTreg. These include CD127, CD45RA/RO, inducible costimulatory protein (ICOS) and HLA class II. The IL-7 receptor α-chain (CD127) was shown to be downregulated on human peripheral nTreg and the combined use of CD4+, CD25+ and CD127low markers results in a highly purified population of suppressive cells, as opposed to CD4+CD25+CD127hi T cells that have been associated with pathogenic antigen-specific immune responses including chronic allograft rejection.34,35 It appears that human nTreg display at least two different states of activation: resting/naive (CD45RA+Foxp3low) and activated/differentiated (CD45RACD45RO+Foxp3hi).36,37 The proportion of these subpopulations differs between cord blood and adult peripheral blood, and in patients with immunological diseases. Activated/differentiated CD45RO+ nTreg have been described to be mainly ICOS positive whereas resting nTreg can be ICOS positive or negative. ICOS is a T cell costimulatory receptor and an activation marker and appears to define functionally distinct nTreg populations: ICOS+ nTreg produce IL-10 whereas ICOS nTreg mediate suppression predominantly via transforming growth factorβ (TGFβ).38 While HLA class II expression on CD25hi nTreg doesn't fully match ICOS or CD45RO, it was associated with a functionally distinct subset of “terminally activated” Treg. To date, the link between resting, activated, terminally activated and possibly memory nTreg remains unclear.39

Induced Treg.

Induced or adaptive Treg (iTreg) can be generated from naive T cells in vitro or induced in the periphery in vivo independently from thymic selection.40,41 Two main subtypes of CD4+ iTreg have been described: Tr1 cells producing IL-10 and TGFβ induced Foxp3+ iTreg. Tr1 cells are defined by their signature suppressive cytokine IL-10 but can transiently upregulate Foxp3 expression upon activation. They can be generated in vitro or in vivo by repeated antigenic stimulation in the presence of IL-10 and IFNα. Tr1 cells exert suppression mainly via the production of IL-10 and to a lesser degree by TGFβ secretion, as well as by modulating DC activation and cytokine production.42 Tr1 and nTreg might synergize to control alloresponses as nTreg can induce naive T cells to differentiate into Tr1 cells in vitro in the presence of allogeneic DC.43 In the initial experiments by Groux et al. Tr1 cells were able to suppress the development of colitis in SCID mice when co-transferred with CD4+CD45RBhi T cells.44 Subsequently, it was shown that Tr1 cells were also involved in the regulation of immune responses in transplantation, autoimmunity, inflammation and tumor progression.42

In the presence of TGFβ, in vitro TCR-mediated stimulation of peripheral CD4+CD25 naive T cells was shown to generate CD4+CD25+Foxp3+ T cells with all the phenotypical and functional characteristics of nTreg.45,46 In vivo, Foxp3+ iTreg could also be generated from naive T cells in the periphery with TGFβ playing a pivotal role.40,41,47 It is unclear at present whether Foxp3+ iTreg differ from TGFβ-secreting Th3 cells previously described in oral tolerance protocols.48 Various positive and negative stimuli were reported to influence the induction of Foxp3 expression in CD4+ T cells (Table 1). However, in contrast to nTreg, Foxp3 expression in iTreg seems to be less stable as reflected by their histone methylation, acetylation and microRNA status.4951 The in vitro regulation of Foxp3 in iTreg as well as their dynamics after transfer in vivo has been analyzed.52 Removal of TGFβ in vitro led to a loss of Foxp3 expression after a few days and, after adoptive transfer into wild-type mice most iTreg downregulated Foxp3 within 2 days only. Some studies have suggested that while TCR stimulation of human CD4+CD25 T cells in the presence of TGFβ induces high levels of stable Foxp3 expression, these iTreg are neither anergic nor suppressive and produce effector cytokines. Furthermore, while the presence of regulatory cytokines such as TGFβ and antigen-presentation by immature DC was shown to favor the generation of antigen-specific iTreg in vitro and in vivo, in a pro-inflammatory environment naïve T cells would rather differentiate into Th17 cells as suggested by recent data.5356 Thus, in vitro differentiated human iTreg might not be stable phenotypically and functionally, implying that in vivo transfer of iTreg for therapeutic purposes may give unexpected results and should be considered with caution.

Table 1.

Generation of Foxp3+ iTreg

Positive factors Negative factors
Cytokines TGFβ,45 IL-2,95 leukemia inhibitoy factor (LIF)96 IL-4, IFNγ, IL-21, IL-6.96,97
Costimulation PD-1/PD-L1.98 OX40,99 T cell-immunoglobuline mucin protein-1 (Tim-1)100
Proliferation Rapamycin (mTOR inhibitor)101,102 PI3K-AKT pathway activation (mTOR activation)101,102
Others Retinoic acid,103 IDO,104,105 activation of the aryl hydrocarbon receptor106

Superscript numbers are references.

Mechanisms of Suppression

There are numerous mechanisms of nTreg-mediated suppression that can be mainly subdivided into two categories: dependent on cell-cell contact and/or mediated by cytokines. The respective relevance of theses mechanisms are still a matter of debate which might be explained by differences in the experimental systems (in vitro vs. in vivo, rodent vs. human cells) and disease models that have been used. In vitro, nTreg were shown to inhibit the activation of effector CD4+CD25 T cells predominantly by cell-cell contact dependent mechanisms. Naturally occurring Treg constitutively express on their surface important molecules for their suppressive function such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), membrane-bound TGFβ latency-associated peptide (LAP), glucocorticoid induced tumor necrosis factor receptor (GITR), CD4-related lymphocyte activation gene 3 (LAG-3), galectin-1 and CD39.5763 Moreover, after activation, human nTreg were shown to be able to directly kill CD4+ and CD8+ T cells via the secretion of perforin and granzyme B.64 The role of regulatory cytokines such as IL-10 a TGFβ and more recently IL-35 in nTreg-mediated suppression of immune pathologies has mainly been described in in vivo experimental models.58,65,66 While the effector mechanisms of nTreg is still debated, there is less controversy, at least in in vitro models, on the resulting effect on the responding co-cultured CD4+CD25 T cells, namely inhibition of the transcription of IL-2.67 IL-2 is key in nTreg homeostasis as these cells are highly dependent on exogenous IL-2 for growth in vitro and in vivo. Experiments using transgenic mice have demonstrated that IL-2, although dispensable for nTreg development, was essential for their peripheral maintenance and competitive fitness.68,69 Different groups have also highlighted that IL-2 production by activated effector T cells was responsible for the maintenance of the peripheral pool of nTreg in vivo.70,71 Moreover, there is evidence for IL-2 “sequestration” by nTreg in the regulation of T-cell responses in vivo, i.e., nTreg would compete with activated effector T cells for the available IL-2 to maintain their peripheral pool.72,73 Collectively these data suggest the presence of an autoregulatory loop during immune responses where nTreg respond and expand via IL-2 to the inflammation that they regulate.74 However, it remains unclear which stimuli trigger nTreg suppression in vivo. It was originally thought to be dependent on specific TCR activation75 but recent in vitro experiments using cells from TCR-transgenic mice unravelled TCR-independent mediated suppression.76

Besides CD4+CD25 and CD8+ T cells, nTreg can modulate the effector function of other immune cells including DC, monocytes and B cells as well as NK cells.7780 By their constitutive surface expression of CTLA-4, nTreg can downregulate CD80/CD86 on APC and induce the expression of indoleamine-2,3-dioxygenase (IDO) in DC. IDO exerts immunomodulatory effects through multiple mechanisms including depletion of tryptophan (which is required for normal T cell functions) and production of tryptophan-derived catabolites that promote T and NK cell proliferative arrest and anergy. IDO-expressing immunoregulatory DC have been shown to promote immune tolerance, including in transplantation experimental models.81,82 Thus, by inhibiting the immunogenicity of other effector cells, nTreg could favor a tolerogenic environment that could promote the development of iTreg, contributing to the maintenance of tolerance (a phenomenon referred to as “infectious tolerance”).83 Altogether, the crosstalk between nTreg and alloantigen-presenting DC is important in determining the outcome of the immune response, tipping the balance in favor of regulation rather than immunity (Fig. 1).8486 Besides regulating APC function, previous in vitro and in vivo studies also reported a potential role of nTreg in inhibiting NK cell effector functions and innate immunity. This was mediated by nTreg membrane-bound TGFβ which downregulated NKG2D receptors on the NK cell surface.87

Figure 1.

Figure 1

Pathways of allorecognition, allograft rejection and mechanisms to induce transplantation tolerance.

Treg-Based Immunotherapy in Transplantation

Approaches to treat autoimmunity and to prevent allograft rejection have focused historically on potent immunosuppressive drugs that block the activation and expansion of pathogenic effector T cells. Although very potent, currently available drugs require chronic administration associated with toxicities and the impairment of protective immune responses against pathogens or tumors. The great progresses in our understanding of the basic processes that control immune tolerance as well as more recent characterization of professional nTreg open the door to their therapeutic application, either by enhancing their activity in autoimmune diseases, allograft rejection and graft versus host disease (GVHD), or by blocking their suppressive activity in tumor immunity and in vaccine development. In the transplantation setting, there is evidence that in many experimental protocols where robust peripheral tolerance can be achieved, immunoregulatory mechanisms dependent on donor-specific Treg are critical in the induction and maintenance of the tolerant state.

The possibility of using nTreg in immunotherapeutic protocols is however mainly limited by cell number. Indeed, the peripheral pool of nTreg only accounts for a small proportion of peripheral T cells in healthy individuals. For efficient suppression of alloreactive T cell responses, the pool of Treg needs to be expanded to obtain a peripheral Treg/T effector cells ratio that favors regulation.19

Expansion strategies.

Currently, three main approaches are being explored for Treg expansion in the perspective of therapeutic protocols: ex-vivo nTreg expansion, ex-vivo conversion of naïve T cells to iTreg and in vivo expansion of nTreg and/or induction of iTreg. The first method requires selection of highly purified nTreg prior to in vitro expansion for subsequent adoptive transfer. Purity is a critical issue as even a few contaminating effector T cells might expand in vivo and cause unwanted immune pathologies. As discussed, different surface markers have to be combined to purify human nTreg from the peripheral blood, including CD25hi and CD127low expression, CD45RA+, CD27, CD39, CD49b, folate receptor 4 (FR4) or PD-1.8891 Good manufacturing practice (GMP) accepted isolation strategies are based on CliniMACS (Miltenyi®) protocols, using antibody cocktails with magnetic microbeads and columns. However, these immunomagnetic techniques do not allow the same broad multiparameter selection as compared to flow cytometry cell sorting. Thus, these approaches may lead to poor nTreg purity and still need to be optimized. Once selected, nTreg have to be expanded to the yields needed for clinical application and transfer into patients. We and others have described robust protocols to expand nTreg in vitro in great numbers without loss of their suppressive function.9294 In brief, these strategies are based on the use of donor-derived APC, recipient-derived APC pulsed with donor antigens or surrogate APC (such as anti-CD3/CD28 coated beads) in the presence of high amounts of exogenous IL-2.

The second approach is based on the induction of iTreg in vitro from naïve CD4+ T cells as described in Table 1 or by forcing Foxp3 expression by viral transduction.31,95107 Finally, the third strategy consists in expanding nTreg and/or de novo generation of iTreg in vivo. This would alleviate the need for GMP cell isolation and cumbersome ex-vivo manipulations, thus rendering the therapy more clinically applicable. Blocking the T cell costimulatory signaling pathways (CD28:CD80/CD86, CD154:CD40, OX40:OX40L, ICOS:ICOSL, CD27:CD70) at the time of transplantation and donor-antigen encounter has been shown to facilitate donor-specific iTreg conversion and/or preferential proliferation of nTreg, while inducing anergy of alloantigen-specific effector T cells.108 Current studies suggest that effector T cells and nTreg have qualitative and quantitative differences in TCR stimulation and costimulatory molecules requirements, and thus could be differentially targeted.109,110 Besides costimulatory blockade, T-cell depletion induction therapies (e.g., anti-CD3, anti-CD52 monoclonal antibodies or polyclonal anti-thymocyte globulins) are used in clinical SOT to prevent acute rejection. These therapies induce profound and durable (weeks to months) reduction of circulating lymphocytes capable of mounting an alloresponse. Recent data suggest that T-cell depletion protocols allow preferential expansion of Treg once lymphocytes gradually repopulate the host, thus skewing the Treg/effector T cell ratio towards tolerance. In these studies, the increased frequency of Treg was neither fully explained by their homeostatic proliferation in a lymphopenic environment nor preferential sparing by the depleting antibody.111113 Although the underlying mechanisms need to be clarified, the induction of apoptotic cells in vivo (as would occur with cell-depleting agents) leads to TGFβ secretion by phagocytes (immature DC, macrophages) involved in clearing these cells, thus favoring iTreg generation and expansion. The uptake of apoptotic cells may also help to maintain DC in an immature state (low level of surface MHC II and costimulatory molecules), favoring tolerance.114116 Besides induction therapies, maintenance immunosuppressive drugs such as mammalian target of rapamycin (mTOR) inhibitors (e.g., sirolimus, everolimus), allow preferential expansion of nTreg and iTreg that promote antigen-specific transplantation tolerance.103,117119 Finally, as stressed before, in vivo homeostasis and expansion of Treg is highly dependent on IL-2. Thus, the administration of IL-2 could be combined to these immunomodulatory approaches and is under investigation in stringent experimental allotransplantation models.120122

Antigen specificity.

Naturally occurring Treg have been shown to have a polyclonal TCR repertoire primarily driven against self-antigens, but with cross-reactivity to transplantation antigens. We and others have shown that, on a cell-per-cell basis, antigen-specific nTreg were more potent suppressors than polyclonal nTreg in organ-specific autoimmune diseases and transplantation experimental models.92,93,123 Moreover, antigen-specificity dictates trafficking of Treg to the appropriate site i.e., the allograft. A potential benefit of donor-specific Treg in SOT is also supported by the fact that Treg-mediated tolerance was demonstrated to be dependent on a continuous supply of donor-derived alloantigens.124 Furthermore, the adoptive transfer of polyclonal Treg carries the risk of deleterious non-specific immune suppression. One noteworthy exception would be the infusion of donor polyclonal Treg to prevent GVHD after allogeneic BMT, as the disease is systemic with muti-organ involvement. In line with this concept, in vitro expanded donor polyclonal nTreg have been successfully used in mouse models of BMT to prevent GVHD while allowing graft-versus-leukemia effect and phase I/II clinical trials are underway.125127 The benefit vs. risk ratio of infusions of donor-derived iTreg (Tr1 cells) are also being evaluated in HSCT from haploidentical donors.128 Overall current available data indicate that the transfer of Treg (nTreg or iTreg) would be a feasible strategy in clinical transplantation with no apparent major side-effects.

Donor alloantigens are recognized by recipient CD4+ T cells either as intact MHC class II:peptide complex presented by donor APC (direct pathway of allorecognition) or after being processed and presented by recipient APC (indirect pathway) (Fig. 1). A series of experimental and clinical data indicate that the indirect pathway alloresponse is the main driver for chronic allograft rejection.129 As current immunosuppressive regimens have little effect on preventing chronic rejection, the control of T cells with indirect alloreactivity would promote transplantation tolerance. Treg with indirect allospecificity can be generated in vitro and were indeed shown to prevent acute and chronic rejection of skin and cardiac allografts respectively in rodent models.92,130132 We and others have also demonstrated that specificity to a single antigen was sufficient to convey protection for whole tissues expressing at least the same antigen together with other epitopes (a mechanism referred to as “linked suppression”).133 The use of antigen-specific Treg at the time of transplantation may be limited if the donor is cadaveric i.e., not known in advance, as time is required to generate and expand ex-vivo donor-specifc Treg. In the contrary, if a living donor is available (HSCT, kidney, liver transplantation), recipient (or donor in the case of HSCT) T cells could be isolated in advance and manipulated ex-vivo in the presence of donor-derived APC or peptides.

Limitations for clinical application.

To date the efficacy of Treg immunotherapy to induce tolerance in SOT has been mainly demonstrated in experimental models of adoptive transfer into lymphopenic animals.92,130 Our data indicated several limitations of donor-specific Treg under more stringent conditions such as donor-recipient full MHC-mismatch, in non-lymphopenic recipients or in regulating responses to more immunogenic skin instead of cardiac or islet allografts.92 In these situations, while the transfer of Treg prolonged allograft survival, it was not sufficient to induce robust tolerance on its own. This highlights the need for adjuvant immunomodulatory therapies to suppress strong immune activation and overcome the rapidly expanding pool of alloreactive T cells early after transplantation. As discussed, Treg transfer could be combined with drugs such as costimulatory blockade (e.g., belatacept) or mTOR inhibitors.19,120 Another option would be to combine robust central mechanisms of tolerance induction with peripheral transfer of Treg. Protocols based on hematopoietic mixed chimerism i.e., allogeneic BMT together with a solid organ from the same donor are robust experimental approaches for the induction of transplantation tolerance and were recently applied in kidney transplantation pilot clinical trials.1416 However, they imply toxic cytoreductive preconditioning of the recipient. Using a MHC-mismatched skin transplantation model in mice, Wekerle and colleagues recently reported that the infusion of recipient polyclonal Treg (both nTreg and iTreg) together with donor BM allowed the induction of long-term mixed chimerism and donor-specific transplantation tolerance. Interestingly, the recipient mice did not receive any cytotoxic drug and were conditioned only with a short-course of costimulation blockers and rapamycin.134 Thus, Treg therapy may allow mixed chimerism-based strategies to be translated more easily and with fewer risks into clinical practice.

There is experimental and clinical evidence that an inflammatory context such as post-operative ischemia-reperfusion injury of the graft or concomitant infections can trigger acute rejection episodes and prevent the induction of transplantation tolerance. Recent studies have highlighted the effect of the microenvironment in inhibiting or subverting nTreg function, as well as the plasticity of iTreg that could convert into pathogenic Th17 cells.135 This would partly explain the reported resistance to Treg-mediated suppression under some inflammatory conditions.136,137 An alternative strategy to promote tolerance would therefore be to skew the immune response away from Th17 or Th1 cells and towards Treg by modifying the microenvironment, for example by blocking critical cytokines.138 Finally, in a recent experimental study, a fraction of mouse nTreg failed to maintain Foxp3 expression in vivo, produced inflammatory cytokines and their adoptive transfer led to the rapid onset of autoimmunity.139 Thus, the in vivo homeostasis, lifespan and stability of nTreg and iTreg need to be clarified before clinical trials on Treg transfer can be considered.

Conclusion and Perspectives

Since the first description of suppressor cells in the 1970s, the field of immune regulation has become very complex, reflecting various distinct mechanisms to maintain tolerance. So far, dominant peripheral transplantation tolerance has mostly been associated with Treg. However other T cell subsets have also been described to have immunosuppressive capacities. In particular, CD8+CD28 suppressor T cells, double negative CD4CD8 T regulatory cells and NKT cells have been shown to be involved in the maintenance of peripheral allo- and xenotransplantation tolerance.140 Besides T cells, tolerogenic DC, myeloid-derived suppressor cells, mesenchymal stem cells, embryonic stem cells and more recently B cells were reported to have intrinsic or inducible regulatory properties.141143 The biology of all these cells as well as their potential in SOT need further investigation.

While therapies based on regulatory T or non-T cells proved promising in inhibiting donor-reactive T cell responses and promoting long-term allograft survival in experimental models, the bench-to-beside translation for SOT is still to be tested. Indeed, most of our current knowledge on regulatory cells is based on animal studies or restricted to in vitro assays when analyzing human cells. Therefore, many issues on the homeostasis and function of human regulatory cells still need to be addressed. Moreover, before implementing any new tolerance-inducing strategy in clinical practice, it needs to favorably compare to current well established immunospuppressive protocols in terms of feasibility, efficacy (rate of acute rejection episodes, long-term graft survival) and toxicity.

Acknowledgements

This work was supported by the Swiss National Science Foundation grant 32323B-111370/32003B-111371 (to D.G.), the Faculty of Biology and Medicine of Lausanne University (to D.G. and M.P.), Fondation Medi-CAL Futur (to D.G.) and an unrestricted grant from Astellas (to D.G. and M.P.). Y.M. and J.D.S. are supported by the Swiss National Science Foundation and the Ernst & Lucie Schmidheiny Foundation.

References

  • 1.Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of foreign cells. Nature. 1953;172:603–606. doi: 10.1038/172603a0. [DOI] [PubMed] [Google Scholar]
  • 2.Sayegh MH, Carpenter CB. Transplantation 50 years later—progress, challenges and promises. N Engl J Med. 2004;351:2761–2766. doi: 10.1056/NEJMon043418. [DOI] [PubMed] [Google Scholar]
  • 3.Pascual M, Theruvath T, Kawai T, Tolkoff-Rubin N, Cosimi AB. Strategies to improve long-term outcomes after renal transplantation. N Engl J Med. 2002;346:580–590. doi: 10.1056/NEJMra011295. [DOI] [PubMed] [Google Scholar]
  • 4.Meier-Kriesche HU, Schold JD, Srinivas TR, Kaplan B. Lack of improvement in renal allograft survival despite a marked decrease in acute rejection rates over the most recent era. Am J Transplant. 2004;4:378–383. doi: 10.1111/j.1600-6143.2004.00332.x. [DOI] [PubMed] [Google Scholar]
  • 5.Hernandez-Fuentes MP, Lechler RI. Chronic graft loss. Immunological and non-immunological factors. Contrib Nephrol. 2005;146:54–64. doi: 10.1159/000082065. [DOI] [PubMed] [Google Scholar]
  • 6.Ashton-Chess J, Giral M, Brouard S, Soulillou JP. Spontaneous operational tolerance after immunosuppressive drug withdrawal in clinical renal allotransplantation. Transplantation. 2007;84:1215–1219. doi: 10.1097/01.tp.0000290683.54937.1b. [DOI] [PubMed] [Google Scholar]
  • 7.Starzl TE, Demetris AJ, Murase N, Ildstad S, Ricordi C, Trucco M. Cell migration, chimerism and graft acceptance. Lancet. 1992;339:1579–1582. doi: 10.1016/0140-6736(92)91840-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Strober S, Benike C, Krishnaswamy S, Engleman EG, Grumet FC. Clinical transplantation tolerance twelve years after prospective withdrawal of immunosuppressive drugs: studies of chimerism and anti-donor reactivity. Transplantation. 2000;69:1549–1554. doi: 10.1097/00007890-200004270-00005. [DOI] [PubMed] [Google Scholar]
  • 9.VanBuskirk AM, Burlingham WJ, Jankowska-Gan E, Chin T, Kusaka S, Geissler F, et al. Human allograft acceptance is associated with immune regulation. J Clin Invest. 2000;106:145–155. doi: 10.1172/JCI9171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Anderson DBR, Lampkin GH, Medawar P. Use of skin grafting to distinguish between monzygotic and dizygotic twins in cattle. Heredity. 1951;5:379. [Google Scholar]
  • 11.Sykes M. Mixed chimerism and transplant tolerance. Immunity. 2001;14:417–424. doi: 10.1016/s1074-7613(01)00122-4. [DOI] [PubMed] [Google Scholar]
  • 12.Ildstad ST, Sachs DH. Reconstitution with syngeneic plus allogeneic or xenogeneic bone marrow leads to specific acceptance of allografts or xenografts. Nature. 1984;307:168–170. doi: 10.1038/307168a0. [DOI] [PubMed] [Google Scholar]
  • 13.Wekerle T, Kurtz J, Ito H, Ronquillo JV, Dong V, Zhao G, et al. Allogeneic bone marrow transplantation with co-stimulatory blockade induces macrochimerism and tolerance without cytoreductive host treatment. Nat Med. 2000;6:464–469. doi: 10.1038/74731. [DOI] [PubMed] [Google Scholar]
  • 14.Buhler LH, Spitzer TR, Sykes M, Sachs DH, Delmonico FL, Tolkoff-Rubin N, et al. Induction of kidney allograft tolerance after transient lymphohematopoietic chimerism in patients with multiple myeloma and end-stage renal disease. Transplantation. 2002;74:1405–1409. doi: 10.1097/00007890-200211270-00011. [DOI] [PubMed] [Google Scholar]
  • 15.Scandling JD, Busque S, Dejbakhsh-Jones S, Benike C, Millan MT, Shizuru JA, et al. Tolerance and chimerism after renal and hematopoietic-cell transplantation. N Engl J Med. 2008;358:362–368. doi: 10.1056/NEJMoa074191. [DOI] [PubMed] [Google Scholar]
  • 16.Kawai T, Cosimi AB, Spitzer TR, Tolkoff-Rubin N, Suthanthiran M, Saidman SL, et al. HLA-mismatched renal transplantation without maintenance immunosuppression. N Engl J Med. 2008;358:353–361. doi: 10.1056/NEJMoa071074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fuchimoto Y, Huang CA, Yamada K, Shimizu A, Kitamura H, Colvin RB, et al. Mixed chimerism and tolerance without whole body irradiation in a large animal model. J Clin Invest. 2000;105:1779–1789. doi: 10.1172/JCI8721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kawai T, Sogawa H, Boskovic S, Abrahamian G, Smith RN, Wee SL, et al. CD154 blockade for induction of mixed chimerism and prolonged renal allograft survival in nonhuman primates. Am J Transplant. 2004;4:1391–1398. doi: 10.1111/j.1600-6143.2004.00523.x. [DOI] [PubMed] [Google Scholar]
  • 19.Lechler RI, Garden OA, Turka LA. The complementary roles of deletion and regulation in transplantation tolerance. Nat Rev Immunol. 2003;3:147–158. doi: 10.1038/nri1002. [DOI] [PubMed] [Google Scholar]
  • 20.Bonasio R, Scimone ML, Schaerli P, Grabie N, Lichtman AH, von Andrian UH. Clonal deletion of thymocytes by circulating dendritic cells homing to the thymus. Nat Immunol. 2006;7:1092–1100. doi: 10.1038/ni1385. [DOI] [PubMed] [Google Scholar]
  • 21.Gershon RK. A disquisition on suppressor T cells. Transplant Rev. 1975;26:170–185. doi: 10.1111/j.1600-065x.1975.tb00179.x. [DOI] [PubMed] [Google Scholar]
  • 22.Hall BM, Jelbart ME, Dorsch SE. Suppressor T cells in rats with prolonged cardiac allograft survival after treatment with cyclosporine. Transplantation. 1984;37:595–600. doi: 10.1097/00007890-198406000-00014. [DOI] [PubMed] [Google Scholar]
  • 23.Sakaguchi S, Fukuma K, Kuribayashi K, Masuda T. Organ-specific autoimmune diseases induced in mice by elimination of T cell subset. I. Evidence for the active participation of T cells in natural self-tolerance; deficit of a T cell subset as a possible cause of autoimmune disease. J Exp Med. 1985;161:72–87. doi: 10.1084/jem.161.1.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Fowell D, Mason D. Evidence that the T cell repertoire of normal rats contains cells with the potential to cause diabetes. Characterization of the CD4+ T cell subset that inhibits this autoimmune potential. J Exp Med. 1993;177:627–636. doi: 10.1084/jem.177.3.627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Powrie F, Mason D. OX-22 high CD4+ T cells induce wasting disease with multiple organ pathology: prevention by the OX-22low subset. J Exp Med. 1990;172:1701–1708. doi: 10.1084/jem.172.6.1701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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]
  • 27.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]
  • 28.Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. 2001;27:68–73. doi: 10.1038/83784. [DOI] [PubMed] [Google Scholar]
  • 29.Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20. doi: 10.1038/83707. [DOI] [PubMed] [Google Scholar]
  • 30.Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–21. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
  • 31.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]
  • 32.Wan YY, Flavell RA. Identifying Foxp3-expressing suppressor T cells with a bicistronic reporter. Proc Natl Acad Sci USA. 2005;102:5126–5131. doi: 10.1073/pnas.0501701102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Walker MR, Kasprowicz DJ, Gersuk VH, Benard A, Van Landeghen M, Buckner JH, et al. Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25− T cells. J Clin Invest. 2003;112:1437–1443. doi: 10.1172/JCI19441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, et al. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ Treg cells. J Exp Med. 2006;203:1701–1711. doi: 10.1084/jem.20060772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Codarri L, Vallotton L, Ciuffreda D, Venetz JP, Garcia M, Hadaya K, et al. Expansion and tissue infiltration of an allospecific CD4+CD25+CD45RO+IL-7Ralphahigh cell population in solid organ transplant recipients. J Exp Med. 2007;204:1533–1541. doi: 10.1084/jem.20062120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 30:899–911. doi: 10.1016/j.immuni.2009.03.019. [DOI] [PubMed] [Google Scholar]
  • 37.Fritzsching B, Oberle N, Pauly E, Geffers R, Buer J, Poschl J, et al. Naive regulatory T cells: a novel subpopulation defined by resistance toward CD95L-mediated cell death. Blood. 2006;108:3371–3378. doi: 10.1182/blood-2006-02-005660. [DOI] [PubMed] [Google Scholar]
  • 38.Ito T, Hanabuchi S, Wang YH, Park WR, Arima K, Bover L, et al. Two functional subsets of FOXP3+ regulatory T cells in human thymus and periphery. Immunity. 2008;28:870–880. doi: 10.1016/j.immuni.2008.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol. 2010;10:490–500. doi: 10.1038/nri2785. [DOI] [PubMed] [Google Scholar]
  • 40.Karim M, Kingsley CI, Bushell AR, Sawitzki BS, Wood KJ. Alloantigen-induced CD25+CD4+ regulatory T cells can develop in vivo from CD25-CD4+ precursors in a thymus-independent process. J Immunol. 2004;172:923–928. doi: 10.4049/jimmunol.172.2.923. [DOI] [PubMed] [Google Scholar]
  • 41.Cobbold SP, Castejon R, Adams E, Zelenika D, Graca L, Humm S, et al. Induction of foxP3+ regulatory T cells in the periphery of T cell receptor transgenic mice tolerized to transplants. J Immunol. 2004;172:6003–6010. doi: 10.4049/jimmunol.172.10.6003. [DOI] [PubMed] [Google Scholar]
  • 42.Roncarolo MG, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K, Levings MK. Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol Rev. 2006;212:28–50. doi: 10.1111/j.0105-2896.2006.00420.x. [DOI] [PubMed] [Google Scholar]
  • 43.Jonuleit H, Schmitt E, Kakirman H, Stassen M, Knop J, Enk AH. Infectious tolerance: human CD25(+) regulatory T cells convey suppressor activity to conventional CD4(+) T helper cells. J Exp Med. 2002;196:255–260. doi: 10.1084/jem.20020394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Groux H, O'Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, et al. 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]
  • 45.Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al. Conversion of peripheral CD4+CD25− naive T cells to CD4+CD25+ regulatory T cells by TGFbeta induction of transcription factor Foxp3. J Exp Med. 2003;198:1875–1886. doi: 10.1084/jem.20030152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fantini MC, Becker C, Monteleone G, Pallone F, Galle PR, Neurath MF. Cutting edge: TGFbeta induces a regulatory phenotype in CD4+CD25− T cells through Foxp3 induction and downregulation of Smad7. J Immunol. 2004;172:5149–5153. doi: 10.4049/jimmunol.172.9.5149. [DOI] [PubMed] [Google Scholar]
  • 47.Peng Y, Laouar Y, Li MO, Green EA, Flavell RA. TGFbeta regulates in vivo expansion of Foxp3-expressing CD4+CD25+ regulatory T cells responsible for protection against diabetes. Proc Natl Acad Sci USA. 2004;101:4572–4577. doi: 10.1073/pnas.0400810101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Weiner HL. Induction and mechanism of action of transforming growth factorbeta-secreting Th3 regulatory cells. Immunol Rev. 2001;182:207–214. doi: 10.1034/j.1600-065x.2001.1820117.x. [DOI] [PubMed] [Google Scholar]
  • 49.Baron U, Floess S, Wieczorek G, Baumann K, Grützkau A, Dong J, et al. DNA demethylation in the human FOXP3 locus discriminates regulatory T cells from activated FOXP3(+) conventional T cells. Eur J Immunol. 2007;37:2378–2389. doi: 10.1002/eji.200737594. [DOI] [PubMed] [Google Scholar]
  • 50.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]
  • 51.Lu LF, Thai TH, Calado DP, Chaudhry A, Kubo M, Tanaka K, et al. Foxp3-dependent microRNA155 confers competitive fitness to regulatory T cells by targeting SOCS1 protein. Immunity. 2009;30:80–91. doi: 10.1016/j.immuni.2008.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Selvaraj RK, Geiger TL. A kinetic and dynamic analysis of Foxp3 induced in T cells by TGFbeta. J Immunol. 2007;178:7667–7677. doi: 10.4049/jimmunol.178.12.7667. [DOI] [PubMed] [Google Scholar]
  • 53.Mangan PR, Harrington LE, O'Quinn DB, Helms WS, Bullard DC, Elson CO, et al. Transforming growth factor-beta induces development of the T (H) 17 lineage. Nature. 2006;441:231–234. doi: 10.1038/nature04754. [DOI] [PubMed] [Google Scholar]
  • 54.Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24:179–189. doi: 10.1016/j.immuni.2006.01.001. [DOI] [PubMed] [Google Scholar]
  • 55.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–238. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
  • 56.Tran DQ, Ramsey H, Shevach EM. Induction of FOXP3 expression in naive human CD4+FOXP3 T cells by T-cell receptor stimulation is transforming growth factor-beta dependent but does not confer a regulatory phenotype. Blood. 2007;110:2983–2990. doi: 10.1182/blood-2007-06-094656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Read S, Malmstrom V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med. 2000;192:295–302. doi: 10.1084/jem.192.2.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kingsley CI, Karim M, Bushell AR, Wood KJ. CD25+CD4+ regulatory T cells prevent graft rejection: CTLA-4- and IL-10-dependent immunoregulation of alloresponses. J Immunol. 2002;168:1080–1086. doi: 10.4049/jimmunol.168.3.1080. [DOI] [PubMed] [Google Scholar]
  • 59.Nakamura K, Kitani A, Strober W. Cell contact-dependent immunosuppression by CD4(+)CD25(+) regulatory T cells is mediated by cell surface-bound transforming growth factor beta. J Exp Med. 2001;194:629–644. doi: 10.1084/jem.194.5.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.McHugh RS, Whitters MJ, Piccirillo CA, Young DA, Shevach EM, Collins M, et al. CD4(+)CD25(+) immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity. 2002;16:311–323. doi: 10.1016/s1074-7613(02)00280-7. [DOI] [PubMed] [Google Scholar]
  • 61.Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, et al. Role of LAG-3 in regulatory T cells. Immunity. 2004;21:503–513. doi: 10.1016/j.immuni.2004.08.010. [DOI] [PubMed] [Google Scholar]
  • 62.Garin MI, Chu CC, Golshayan D, Cernuda-Morollon E, Wait R, Lechler RI. Galectin-1: a key effector of regulation mediated by CD4+CD25+ T cells. Blood. 2007;109:2058–2065. doi: 10.1182/blood-2006-04-016451. [DOI] [PubMed] [Google Scholar]
  • 63.Borsellino G, Kleinewietfeld M, Di Mitri D, Sternjak A, Diamantini A, Giometto R, et al. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood. 2007;110:1225–1232. doi: 10.1182/blood-2006-12-064527. [DOI] [PubMed] [Google Scholar]
  • 64.Grossman WJ, Verbsky JW, Barchet W, Colonna M, Atkinson JP, Ley TJ. Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity. 2004;21:589–601. doi: 10.1016/j.immuni.2004.09.002. [DOI] [PubMed] [Google Scholar]
  • 65.Collison LW, Chaturvedi V, Henderson AL, Giacomin PR, Guy C, Bankoti J, et al. IL-35-mediated induction of a potent regulatory T cell population. Nat Immunol. 2010;11:1093–1101. doi: 10.1038/ni.1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Bettini M, Vignali DA. Regulatory T cells and inhibitory cytokines in autoimmunity. Curr Opin Immunol. 2009;21:612–618. doi: 10.1016/j.coi.2009.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Thornton AM, Shevach EM. CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting Interleukin 2 production. J Exp Med. 1998;188:287–296. doi: 10.1084/jem.188.2.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Fontenot JD, Rasmussen JP, Gavin MA, Rudensky AY. A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat Immunol. 2005;6:1142–1151. doi: 10.1038/ni1263. [DOI] [PubMed] [Google Scholar]
  • 69.D'Cruz LM, Klein L. Development and function of agonist-induced CD25+Foxp3+ regulatory T cells in the absence of interleukin 2 signaling. Nat Immunol. 2005;6:1152–1159. doi: 10.1038/ni1264. [DOI] [PubMed] [Google Scholar]
  • 70.Furtado GC, Curotto de Lafaille MA, Kutchukhidze N, Lafaille JJ. Interleukin 2 signaling is required for CD4(+) regulatory T cell function. J Exp Med. 2002;196:851–857. doi: 10.1084/jem.20020190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Malek TR, Bayer AL. Tolerance, not immunity, crucially depends on IL-2. Nat Rev Immunol. 2004;4:665–674. doi: 10.1038/nri1435. [DOI] [PubMed] [Google Scholar]
  • 72.Stockinger B, Kassiotis G, Bourgeois C. Homeostasis and T cell regulation. Curr Opin Immunol. 2004;16:775–779. doi: 10.1016/j.coi.2004.09.003. [DOI] [PubMed] [Google Scholar]
  • 73.Barthlott T, Moncrieffe H, Veldhoen M, Atkins CJ, Christensen J, O'Garra A, et al. CD25+CD4+ T cells compete with naive CD4+ T cells for IL-2 and exploit it for the induction of IL-10 production. Int Immunol. 2005;17:279–288. doi: 10.1093/intimm/dxh207. [DOI] [PubMed] [Google Scholar]
  • 74.Maloy KJ, Powrie F. Fuelling regulation: IL-2 keeps CD4+ Treg cells fit. Nat Immunol. 2005;6:1071–1072. doi: 10.1038/ni1105-1071. [DOI] [PubMed] [Google Scholar]
  • 75.Thornton AM, Shevach EM. Suppressor effector function of CD4+CD25+ immunoregulatory T cells is antigen nonspecific. J Immunol. 2000;164:183–190. doi: 10.4049/jimmunol.164.1.183. [DOI] [PubMed] [Google Scholar]
  • 76.Szymczak-Workman AL, Workman CJ, Vignali DA. Cutting edge: regulatory T cells do not require stimulation through their TCR to suppress. J Immunol. 2009;182:5188–5192. doi: 10.4049/jimmunol.0803123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cederbom L, Hall H, Ivars F. CD4+CD25+ regulatory T cells downregulate co-stimulatory molecules on antigen-presenting cells. Eur J Immunol. 2000;30:1538–1543. doi: 10.1002/1521-4141(200006)30:6<1538::AID-IMMU1538>3.0.CO;2-X. [DOI] [PubMed] [Google Scholar]
  • 78.Tang Q, Adams JY, Tooley AJ, Bi M, Fife BT, Serra P, et al. 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]
  • 79.Maloy KJ, Salaun L, Cahill R, Dougan G, Saunders NJ, Powrie F. CD4+CD25+ T(R) cells suppress innate immune pathology through cytokine-dependent mechanisms. J Exp Med. 2003;197:111–119. doi: 10.1084/jem.20021345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Bystry RS, Aluvihare V, Welch KA, Kallikourdis M, Betz AG. B cells and professional APCs recruit regulatory T cells via CCL4. Nat Immunol. 2001;2:1126–1132. doi: 10.1038/ni735. [DOI] [PubMed] [Google Scholar]
  • 81.Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol. 2004;4:762–774. doi: 10.1038/nri1457. [DOI] [PubMed] [Google Scholar]
  • 82.Li J, Meinhardt A, Roehrich ME, Golshayan D, Dudler J, Pagnotta M, et al. Indoleamine-2,3-dioxygenase gene transfer prolongs cardiac allograft survival. Am J Physiol Heart Circ Physiol. 2007;293:3415–3423. doi: 10.1152/ajpheart.00532.2007. [DOI] [PubMed] [Google Scholar]
  • 83.Waldmann H, Adams E, Fairchild P, Cobbold S. Infectious tolerance and the long-term acceptance of transplanted tissue. Immunol Rev. 2006;212:301–313. doi: 10.1111/j.0105-2896.2006.00406.x. [DOI] [PubMed] [Google Scholar]
  • 84.Lanzavecchia A, Sallusto F. Regulation of T cell immunity by dendritic cells. Cell. 2001;106:263–266. doi: 10.1016/s0092-8674(01)00455-x. [DOI] [PubMed] [Google Scholar]
  • 85.Lechler R, Ng WF, Steinman RM. Dendritic cells in transplantation: Friend or foe? Immunity. 2001;14:357–368. doi: 10.1016/s1074-7613(01)00116-9. [DOI] [PubMed] [Google Scholar]
  • 86.Morelli AE, Thomson AW. Tolerogenic dendritic cells and the quest for transplant tolerance. Nature Rev Immunol. 2007;7:610–621. doi: 10.1038/nri2132. [DOI] [PubMed] [Google Scholar]
  • 87.Ghiringhelli F, Ménard C, Terme M, Flament C, Taieb J, Chaput N, et al. CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factorbeta-dependent manner. J Exp Med. 2005;202:1075–1085. doi: 10.1084/jem.20051511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA. CD4+CD25high regulatory cells in human peripheral blood. J Immunol. 2001;167:1245–1253. doi: 10.4049/jimmunol.167.3.1245. [DOI] [PubMed] [Google Scholar]
  • 89.Yamaguchi T, Hirota K, Nagahama K, Ohkawa K, Takahashi T, Nomura T, et al. Control of immune responses by antigen-specific regulatory T cells expressing the folate receptor. Immunity. 2007;l:145–159. doi: 10.1016/j.immuni.2007.04.017. [DOI] [PubMed] [Google Scholar]
  • 90.Hoffmann P, Eder R, Kunz-Schughart LA, Andreesen R, Edinger M. Large-scale in vitro expansion of polyclonal human CD4(+)CD25high regulatory T cells. Blood. 2004;104:895–903. doi: 10.1182/blood-2004-01-0086. [DOI] [PubMed] [Google Scholar]
  • 91.Coenen JJ, Koenen HJ, van Rijssen E, Hilbrands LB, Joosten I. Rapamycin and not cyclosporin A, preserves the highly suppressive CD27+ subset of human CD4+CD25+ regulatory T cells. Blood. 2006;107:1018–1023. doi: 10.1182/blood-2005-07-3032. [DOI] [PubMed] [Google Scholar]
  • 92.Golshayan D, Jiang S, Tsang J, Garin MI, Mottet C, Lechler RI. In vitro-expanded donor alloantigen-specific CD4+CD25+ regulatory T cells promote experimental transplantation tolerance. Blood. 2007;109:827–835. doi: 10.1182/blood-2006-05-025460. [DOI] [PubMed] [Google Scholar]
  • 93.Tang Q, Henriksen KJ, Bi M, Finger EB, Szot G, Ye J, et al. In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes. J Exp Med. 2004;199:1455–1465. doi: 10.1084/jem.20040139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Peters JH, Preijers FW, Woestenenk R, Hilbrands LB, Koenen HJ, Joosten I. Clinical grade Treg: GMP isolation, improvement of purity by CD127 Depletion, Treg expansion and Treg cryopreservation. PLoS One. 2008;3:3161. doi: 10.1371/journal.pone.0003161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Allan SE, Song-Zhao GX, Abraham T, McMurchy AN, Levings MK. Inducible reprogramming of human T cells into Treg cells by a conditionally active form of FOXP3. Eur J Immunol. 2008;38:3282–3289. doi: 10.1002/eji.200838373. [DOI] [PubMed] [Google Scholar]
  • 96.Zheng SG, Wang J, Wang P, Gray JD, Horwitz DA. IL-2 is essential for TGF-beta to convert naive CD4+CD25− cells to CD25+Foxp3+ regulatory T cells and for expansion of these cells. J Immunol. 2007;178:2018–2027. doi: 10.4049/jimmunol.178.4.2018. [DOI] [PubMed] [Google Scholar]
  • 97.Gao W, Thompson L, Zhou Q, Putheti P, Fahmy TM, Strom TB, et al. Treg versus Th17 lymphocyte lineages are cross-regulated by LIF versus IL-6. Cell Cycle. 2009;8:1444–1450. doi: 10.4161/cc.8.9.8348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Korn T, Mitsdoerffer M, Croxford AL, Awasthi A, Dardalhon VA, Galileos G, et al. IL-6 controls Th17 immunity in vivo by inhibiting the conversion of conventional T cells into Foxp3+ regulatory T cells. Proc Natl Acad Sci USA. 2008;105:18460–18465. doi: 10.1073/pnas.0809850105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK, et al. PD-L1 regulates the development, maintenance and function of induced regulatory T cells. J Exp Med. 2009;206:3015–3029. doi: 10.1084/jem.20090847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Vu MD, Xiao X, Gao W, Degauque N, Chen M, Kroemer A, et al. OX40 costimulation turns off Foxp3+ Tregs. Blood. 2007;110:2501–2510. doi: 10.1182/blood-2007-01-070748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Mariat C, Degauque N, Strom TB. TIM-1: a new player in transplant immunity. Transplantation. 2009;87:84–86. doi: 10.1097/TP.0b013e3181a2ba83. [DOI] [PubMed] [Google Scholar]
  • 102.Haxhinasto S, Mathis D, Benoist C. The AKT-mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells. J Exp Med. 2008;205:565–574. doi: 10.1084/jem.20071477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zeiser R, Negrin RS. Interleukin-2 receptor downstream events in regulatory T cells: implications for the choice of immunosuppressive drug therapy. Cell Cycle. 2008;7:458–462. doi: 10.4161/cc.7.4.5454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Hill JA, Hall JA, Sun CM, Cai Q, Ghyselinck N, Chambon P, et al. Retinoic acid enhances Foxp3 induction indirectly by relieving inhibition from CD4+CD44hi Cells. Immunity. 2008;29:758–770. doi: 10.1016/j.immuni.2008.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Chung DJ, Rossi M, Romano E, Ghith J, Yuan J, Munn DH, et al. Indoleamine-2,3-dioxygenase-expressing mature human monocyte-derived dendritic cells expand potent autologous regulatory T cells. Blood. 2009;114:555–563. doi: 10.1182/blood-2008-11-191197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Baban B, Chandler PR, Sharma MD, Pihkala J, Koni PA, Munn DH, et al. IDO activates regulatory T cells and blocks their conversion into Th17-like T cells. J Immunol. 2009;183:2475–2483. doi: 10.4049/jimmunol.0900986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Gandhi R, Kumar D, Burns EJ, Nadeau M, Dake B. Activation of the aryl hydrocarbon receptor induces human type 1 regulatory T cell-like and Foxp3(+) regulatory T cells. Nat Immunol. 2010;11:846–853. doi: 10.1038/ni.1915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Wekerle T, Kurtz J, Bigenzahn S, Takeuchi Y, Sykes M. Mechanisms of transplant tolerance induction using co-stimulatory blockade. Curr Opin Immunol. 2002;14:592–600. doi: 10.1016/s0952-7915(02)00378-3. [DOI] [PubMed] [Google Scholar]
  • 109.Ndhlovu LC, Takeda I, Sugamura K, Ishii N. Expanding role of T-cell costimulators in regulatory T-cell function: recent advances in accessory molecules expressed on both regulatory and nonregulatory T cells. Crit Rev Immunol. 2004;24:251–266. doi: 10.1615/critrevimmunol.v24.i4.30. [DOI] [PubMed] [Google Scholar]
  • 110.Bluestone JA, Liu W, Yabu JM, Laszik ZG, Putnam A, Belingheri M, et al. The effect of costimulatory and interleukin 2 receptor blockade on regulatory T cells in renal transplantation. Am J Transplant. 2008;8:2086–2096. doi: 10.1111/j.1600-6143.2008.02377.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Lopez M, Clarkson MR, Albin M, Sayegh MH, Najafian N. A novel mechanism of action for antithymocyte globulin: induction of CD4+CD25+Foxp3+ regulatory T-cells. J Am Soc Nephrol. 2006;17:2844–2853. doi: 10.1681/ASN.2006050422. [DOI] [PubMed] [Google Scholar]
  • 112.Bloom DD, Chang Z, Fechner JH, Dar W, Polster SP, Pascual J, et al. CD4+CD25+FOXP3+ regulatory T cells increase de novo in kidney transplant patients after immunodepletion with Campath-1H. Am J Transplant. 2008;8:793–802. doi: 10.1111/j.1600-6143.2007.02134.x. [DOI] [PubMed] [Google Scholar]
  • 113.Noris M, Casiraghi F, Todeschini M, Cravedi P, Cugini D, Monteferrante G, et al. Regulatory T cells and T cell depletion: role of immunosuppressive drugs. J Am Soc Nephrol. 2007;18:1007–1018. doi: 10.1681/ASN.2006101143. [DOI] [PubMed] [Google Scholar]
  • 114.Belghith M, Bluestone JA, Barriot S, Mégret J, Bach JF, Chatenoud L. TGFbeta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes. Nat Med. 2003;9:1202–1208. doi: 10.1038/nm924. [DOI] [PubMed] [Google Scholar]
  • 115.Perruche S, Zhang P, Liu Y, Saas P, Bluestone JA, Chen W. CD3-specific antibody-induced immune tolerance involves transforming growth factor-beta from phagocytes digesting apoptotic T cells. Nat Med. 2008;14:528–535. doi: 10.1038/nm1749. [DOI] [PubMed] [Google Scholar]
  • 116.Morelli AE. The immune regulatory effect of apoptotic cells and exosomes on dendritic cells: its impact on transplantation. Am J Transplant. 2006;6:254–261. doi: 10.1111/j.1600-6143.2005.01197.x. [DOI] [PubMed] [Google Scholar]
  • 117.Wells AD, Li XC, Li Y, Walsh M, Zeng XX, Wu Z, et al. Requirement for T cell apoptosis in the induction of peripheral transplantation tolerance. Nat Med. 1999;5:1039–1046. doi: 10.1038/15260. [DOI] [PubMed] [Google Scholar]
  • 118.Battaglia M, Stabilini A, Roncarolo MG. Rapamycin selectively expands CD4+CD25+FoxP3+ regulatory T cells. Blood. 2005;105:4743–4748. doi: 10.1182/blood-2004-10-3932. [DOI] [PubMed] [Google Scholar]
  • 119.Muller YD, Mai G, Morel P, Serre-Beinier V, Gonelle-Gispert C, Yung GP, et al. Anti-CD154 mAb and rapamycin induce T regulatory cell mediated tolerance in rat-to-mouse islet transplantation. PLoS One. 2010;5:10352. doi: 10.1371/journal.pone.0010352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Zheng XX, Sanchez-Fueyo A, Sho M, Domenig C, Sayegh MH, Strom TB. Favorably tipping the balance between cytopathic and regulatory T-cells to create transplantation tolerance. Immunity. 2003;19:503–514. doi: 10.1016/s1074-7613(03)00259-0. [DOI] [PubMed] [Google Scholar]
  • 121.Zorn E, Nelson EA, Mohseni M, Porcheray F, Kim H, Litsa D, et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood. 2006;108:1571–1579. doi: 10.1182/blood-2006-02-004747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Webster KE, Walters S, Kohler RE, Mrkvan T, Boyman O, Surh CD, et al. In vivo expansion of Treg 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]
  • 123.Tarbell KV, Yamazaki S, Olson K, Toy P, Steinman RM. CD25+CD4+ T cells, expanded with dendritic cells presenting a single autoantigenic peptide, suppress autoimmune diabetes. J Exp Med. 2004;99:1467–1477. doi: 10.1084/jem.20040180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Sayegh M, Zheng X, Magee C, Hancock W, Turka L. Donor antigen is necessary for the prevention of chronic rejection in CTLA-4-Ig-treated murine cardiac allografts. Transplantation. 1997;64:1646–1650. doi: 10.1097/00007890-199712270-00003. [DOI] [PubMed] [Google Scholar]
  • 125.Taylor PA, Lees CJ, Blazar BR. The infusion of ex vivo activated and expanded CD4(+)CD25(+) immune regulatory cells inhibits graft-versus-host disease lethality. Blood. 2002;99:3493–3499. doi: 10.1182/blood.v99.10.3493. [DOI] [PubMed] [Google Scholar]
  • 126.Edinger M, Hoffmann P, Ermann J, Drago K, Fathman CG, Strober S, et al. CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med. 2003;9:1144–1150. doi: 10.1038/nm915. [DOI] [PubMed] [Google Scholar]
  • 127.Trzonkowski P, Bieniaszewska M, Juscinska J, Dobyszuk A, Krzystyniak A, et al. First-in-man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+CD25+CD127− T regulatory cells. Clin Immunol. 2009;133:22–26. doi: 10.1016/j.clim.2009.06.001. [DOI] [PubMed] [Google Scholar]
  • 128.Jiang S, Tsang J, Tam P. Regulatory T cell immunotherapy for transplantation tolerance: step into clinic. Int Immunopharmacol. 2010;10:1486–1490. doi: 10.1016/j.intimp.2010.08.007. [DOI] [PubMed] [Google Scholar]
  • 129.Gökmen MR, Lombardi G, Lechler RI. The importance of the indirect pathway of allorecognition in clinical transplantation. Curr Opin Immunol. 2008;20:568–574. doi: 10.1016/j.coi.2008.06.009. [DOI] [PubMed] [Google Scholar]
  • 130.Joffre O, Santolaria T, Calise D, Al Saati T, Hudrisier D, Romagnoli P, et al. Prevention of acute and chronic allograft rejection with CD4+CD25+Foxp3+ regulatory T lymphocytes. Nat Med. 2008;14:88–92. doi: 10.1038/nm1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Jiang S, Golshayan D, Tsang J, Lombardi G, Lechler RI. In vitro expanded alloantigen-specific CD4+CD25+ regulatory T cell treatment for the induction of donor-specific transplantation tolerance. Int Immunopharmacol. 2006;6:1879–1882. doi: 10.1016/j.intimp.2006.07.025. [DOI] [PubMed] [Google Scholar]
  • 132.Tsang JY, Tanriver Y, Jiang S, Xue SA, Ratnasothy K, Chen D, et al. Conferring indirect allospecificity on CD4+CD25+ Tregs by TCR gene transfer favors transplantation tolerance in mice. J Clin Invest. 2008;118:3619–3628. doi: 10.1172/JCI33185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wise MP, Bemelman F, Cobbold SP, Waldmann H. Linked suppression of skin graft rejection can operate through indirect recognition. J Immunol. 1998;161:5813–5816. [PubMed] [Google Scholar]
  • 134.Pilat N, Baranyi U, Klaus C, Jaeckel E, Mpofu N, Wrba F, et al. Treg-therapy allows mixed chimerism and transplantation tolerance without cytoreductive conditioning. Am J Transplant. 2010;10:751–762. doi: 10.1111/j.1600-6143.2010.03018.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Yang XO, Nurieva R, Martinez GJ, Kang HS, Chung Y, Pappu BP, et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity. 2008;29:44–56. doi: 10.1016/j.immuni.2008.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Chen L, Ahmed E, Wang T, Wang Y, Ochando J, Chong AS, et al. TLR signals promote IL6/IL-17-dependent transplant rejection. J Immunol. 2009;182:6217–6225. doi: 10.4049/jimmunol.0803842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Ahmed EB, Daniels M, Alegre ML, Chong AS. Bacterial infections, alloimmunity and transplantation tolerance. Transplant Rev. 2011;25:27–35. doi: 10.1016/j.trre.2010.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Hanidziar D, Koulmanda M. Inflammation and the balance of Treg and Th17 cells in transplant rejection and tolerance. Curr Opin Organ Transplant. 2010;15:411–415. doi: 10.1097/MOT.0b013e32833b7929. [DOI] [PubMed] [Google Scholar]
  • 139.Zhou X, Bailey-Bucktrout SL, Jeker LT, Penaranda C, Martínez-Llordella M, Ashby M, et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol. 2009;10:1000–1007. doi: 10.1038/ni.1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Muller YD, Golshayan D, Ehirchiou D, Wekerle T, Seebach JD, Bühler LH. T regulatory cells in xenotransplantation. Xenotransplantation. 2009;16:121–128. doi: 10.1111/j.1399-3089.2009.00531.x. [DOI] [PubMed] [Google Scholar]
  • 141.Morelli AE, Thomson AW. Tolerogenic dendritic cells and the quest for transplant tolerance. Nat Rev Immunol. 2007;7:610–621. doi: 10.1038/nri2132. [DOI] [PubMed] [Google Scholar]
  • 142.Dilek N, van Rompaey N, Le Moine A, Vanhove B. Myeloid-derived suppressor cells in transplantation. Curr Opin Organ Transplant. 2010;15:765–768. doi: 10.1097/MOT.0b013e3283401742. [DOI] [PubMed] [Google Scholar]
  • 143.Newell KA, Asare A, Kirk AD, Gisler TD, Bourcier K, Suthanthiran M, et al. Immune Tolerance Network ST507 Study Group. Identification of a B cell signature associated with renal transplant tolerance in humans. J Clin Invest. 2010;120:1836–1847. doi: 10.1172/JCI39933. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Self Nonself are provided here courtesy of Taylor & Francis

RESOURCES