Skip to main content
  • Research Article
  • Open access
  • Published:

T regulatory Cell-mediated Immunotherapy for Solid Organ Transplantation: A Clinical Perspective

Abstract

T regulatory cells (Tregs) play a vital role in suppressing heightened immune response, and thereby promote a state of immunological tolerance. Tregs modulate both innate and adaptive immunity, which makes them potential candidates for cell-based immunotherapy in the suppression of uncontrolled activation of graft-specific inflammatory cells and toxic mediators. Graft-specific inflammatory cells (T effector cells) and other inflammatory mediators (immunoglobulins, active complement mediators) are mainly responsible for graft vascular deterioration followed by acute/chronic rejection. Treg-mediated immunotherapy is under investigation to induce allospecific tolerance in various ongoing clinical trials in organ transplant recipients. Treg immunotherapy shows promising results; however, key issues regarding Treg immunotherapy remain unresolved, including the mechanism of action and specific Treg cell phenotypes responsible for a state of tolerance. This review highlights the involvement of various subsets of Tregs during immune suppression, the novelty of Treg functions, effects on angiogenesis, emerging technologies for effective Treg expansion, and plasticity and safety associated with clinical applications. Altogether, this information will assist in designing single/combined Treg-mediated therapies for successful clinical trials in solid organ transplantations.

Introduction

A typical immune response requires a firm balance between activation and attenuation, dependent upon the balance of T effector and regulatory T cell function, in turn dependent on molecular signaling. Alterations in the cell transcriptional phase are critical to the onset of immune self-tolerance (1). Likewise, immunotherapies for organ transplantation face challenges in achieving enough immunosuppression to prevent organ rejection while limiting autoreactivity, without impairing the host’s ability to guard against opportunistic infections and malignancies. The immune system defends the host from a broad range of pathogens and foreign tissue antigens while preventing unwarranted and exaggerated immune reactions that would be deleterious to the host tissue (2–4). During an immune response, T and B cells modulate an effective response against foreign tissue antigens, characterized by broad antigen recognition, high specificity, strong effector response and long-term immunologic memory (5,6). An effective immune response balances unresponsiveness to self-antigens (immunological self-tolerance) and the magnitude of adaptive immune responses to non-self-antigens, thereby preventing host tissue destruction (7–9) (Figure 1A). The model of immunotolerance explains how inadequate immune responses against tumor and microbial antigens in chronic infections can be augmented, or how aberrant immune responses to allograft can be regulated. Immunotolerance has been shown to modulate various populations of regulatory cells, which include T regulatory cells (CD4+ CD25+FOXP3+ Tregs) (5,10), B regulatory cells (CD19+CD24+CD38+ Bregs) (11,12), natural killer T cells (CD16+CD56+ NK T cells) (13) and, finally, dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin cells (DC-SIGN+ macrophages) (14).

Figure 1
figure 1

Development of Tregs and immune balance. (A) Treg develops from naïve CD4+ T cell population under the influence of IL-4 and IL-2 and characterized by surface expression of CD25 and nuclear expression of FOXP3 compared to other T cell lineages. (B) Immune balance between Tregs (graft-protective cells) and T-effector cells (graft-destructive cells) modulate the effective immune response and immunotolerance to foreign antigens.

Treg Subsets

Tregs, produced from naïve CD4+ T cells in the thymus as functionally mature CD4+ T cell subsets, play a vital role in providing immunological tolerance to self-antigens (15,16). The regulatory cells neutralize killer T cells during inflammation (17) and suppress heightened immune responses destructive to host tissue in organ transplant recipients (18–20).

Tregs (5–10% CD4+ T cells) are crucial to the regulation of self-tolerance and are capable of inhibiting antigen-specific inflammatory responses (7,21–24) (Figure 1B). Regulatory T cells, originally identified as antigen-specific T suppressor cells, uniquely express surface CD25 and the nuclear FOXP3 gene (25,26). The FOXP3 gene is required for immunosuppressive functions and regulation, acting through suppression of cytokines interleukin-2 (IL-2), interferon gamma (IFN-γ) and interleukin-4 (IL-4), and activation of interleukin-10 (IL-10), high-affinity IL-2R, CD25, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and glucocorticoid-induced TNFR-related protein family-related genes/proteins (20,21,26–29). The FOXP3 gene stimulates Treg-associated genes and stabilizes Treg features during antigen-specific activation while inhibiting expression of Th1-, Th2- and Th17-associated genes (26,30).

Distinct subsets of Tregs could play an important immunosuppressive role during rejection (31). Based on surface distribution of various expression proteins and state of origin, Treg subsets include natural Tregs (nTregs), inducible/adaptive Tregs (iTregs), inducible costimulator (ICOS+) Tregs, IL-10-producing type 1 Tregs (Tr1 cells), CD8+ Tregs, IL-17-producing Tregs and CD4+VEGFR1HIGH Tregs (32,33). These subsets share expression of the FOXP3 gene (except for Tr1 cells) and secretion of inhibitory cytokine IL-10 and/or tumor growth factor beta (TGF-β).

nTREGs. nTregs are characterized by CD4, CD25 and FOXP3 and are involved in inhibiting T cell proliferation, suppressing dendritic cells (DCs) and inhibiting effector Th1, Th2 and Th17 cells. They also suppress mast cells, basophils and eosinophils, interact with resident tissue cells and participate in tissue remodeling thorough the release of IL-10 and TGF-β (26,34).

ICOS+ Tregs. ICOS+ Tregs are generated from nTregs and are characterized by surface expression of CD4, CD25, FOXP3 and ICOS (35). They are involved in suppression of hapten-reactive CD8+ T cells and release of IL-10, IL-17 and IFN-γ (36,37).

iTregs. iTregs are generated in the periphery and express CD4 FOXP3 as surface markers. They act through IL-10 and TGF-β (38–40).

Tri Cells. Tr1 cells, which display CD4 and CD25, are generated from non-Treg cell precursors and draining lymph nodes. They suppress effector Th cell migration and function and suppress mast cells, basophils and eosinophils through the release of IL-10 (41).

CD8+ Tregs. CD8+ Tregs, which display unique CD8, FOXP3, CD25, CD28 and CD122, are generated from CD8 cells. They are involved in blocking activation of naive or effector T cells, suppression of IgG/IgE antibody responses and IL-4 expression, and proliferation of CD4+ T cells through IL-10, TNF-α and IFN-γ release (42,43).

IL-17-producing FOXP3+. IL-17-producing FOXP3+ Tregs, characterized by expression of CD4, FOXP3, chemokine receptor type 6 (CCR6) and RAR-related orphan receptor gamma transcription factor, are differentiated from CD4+− FOXP3+CCR6− Tregs in peripheral blood and lymphoid tissue. They are mainly involved in inhibiting proliferation of CD4+ effector T cells through IL-17 (44).

CD4+VEGFR1high Treg. Recently, a new CD4+VEGFR1high Treg subset has been reported to suppress proliferation of CD4+CD25− T cells as efficiently as CD4+CD25high natural Tregs in a contact-independent manner (32).

Treg-Mediated Immunosuppression

Treg activation is antigen-specific, which suggests that their immunosuppressive properties are a function of antigen exposure during an inflammatory response. In vitro experiments have shown that the suppressive property of Tregs requires T cell receptor (TCR)-mediated activation (45,46). In addition, antigen specificity modulates Treg proliferation and expansion in lymph nodes (47). Immunotolerance is linked with donor-specific Treg proliferation and expansion, possibly through the reconstitution of donor-antigen specific Tregs. This may prolong allograft survival and induce immunotolerance, as reported in different preclinical and clinical conditions of transplantation (48–52). Treg modulates antigen presentation of antigen-presenting cells (APCs) to conventional T-cells, which in turn become either anergic or regulatory cells (53–55). Therefore, empowering Treg at the expense of CD4+ T can induce a state of immune privilege that could facilitate long-term graft survival.

The mechanism of Treg suppressive functions remains contentious. The major differences between in vitro and in vivo outcomes, specifically with the IL-10 and TGF-β inhibitory cytokines, have fueled the dispute (56). Under antigen-stimulated in vitro conditions, Tregs suppress the proliferation and cytokine production of effector T cells irrespective of antigen specificity (56). Tregs also play specialized regulatory functions during inflammatory conditions and are key regulators in switching off the immune response after the onset of the inflammatory phase (18). Under normal circumstances nTregs prevent autoimmune diseases while iTregs actively modulate transplantation tolerance (9,57). In vivo and in vitro Tregs are characterized by an anergic state with suppressive functions and are able to inhibit multiple stages of target cell activities (7,16). nTregs have the potential to suppress proliferation and differentiation of naïve T (CD4+ and CD8+) cells into effector T cells, and also suppress effector activities, differentiation and functions of NK cells, NK T cells, B cells, macrophages, osteoclasts and DCs (5,20,58). As reported in preclinical and clinical studies, immune modulation through Treg regulation is a decisive factor in allografts due to the insufficient number of Tregs, which favor allograft injury and rejection in organ transplantations (59–62). Various modes of Tregmediated suppression have been proposed demonstrating that Tregs adopt various mechanisms for immunosuppression, including cell-contact-dependent secretion of immunosuppressive cytokines and local consumption of growth factors (8,58). Cell-cell interactions of Tregs and dendritic cells trigger the release of IFN-γ, a key inducer of indole amine 2, 3 dioxygenase, which catalyzes the conversion of tryptophan to kynurenine (63). In turn, release of kynurenine triggers generation of T regulatory cells (64,65).

During the process of immune inflammation, Tregs release a myriad of molecular mediators, which include several cytokines (TGF-β1, IL-35, IL-10), the cytotoxic molecule perforin and granzymes, to mitigate immunosuppressive T cells and control inflammation (45,66) (Figure 2). TGF-β1 is one of the key mediators, performing both offensive (67) and defensive (68,69) functions of Tregs during immunosuppression. Tregs utilize TGF-β1 to suppress T cell activation and differentiation to dampen inflammatory response (70). In addition, TGF-β1 released from Tregs has the potential not only to convert naive T cells into iTregs and Th17 to assist in their fight against local inflammatory condition, and defend Tregs against apoptosis and destabilization during inflammatory phase (68,69), but also to affect the activity of cytotoxic T cells and APCs (71) (Figure 2). During a regulatory response, TGF-β1 plays a key role in inflammation, T cell lineage, antibody production, immunosuppression and maintenance of tolerance (70). TGF-β1 is also critical to the development and differentiation of FOXP3+ Tregs (72–74). In addition, TGF-β1 is essential for the generation of IL-17-producing Th17 cells (70), and recent findings indicate that TGF-β1 is involved in the generation of IL-9-producing Th9 cells (75). These observations highlight the role of TGF-β1 in T cell proliferation and differentiation (75). Furthermore, TGF-β1 suppress cytokine released by activated CD4+ T cells without restricting differentiation and apoptosis, while IL-10 assists activated T cells to TGF- β1 response through the expression of TGF receptors (71). Various Treg surface markers have been proposed for this direct interaction. These include glucocorticoid-induced TNFR-related protein, CTLA-4, membrane-bound TGF-β, LAG-3 and the cytolytic molecules Fas and granzyme B (76). However, constitutive expression of CD25 by Tregs gives them an initial competitive advantage for the consumption of IL-2 over naïve T cells, which express CD25 only after TCR stimulation (58). As reported earlier, nTregs predominantly produce immunosuppressive IL-35, a new member of the IL-12 family, which confers a suppressive activity of Tregs (77).

Figure 2
figure 2

Array of Treg-mediated immunosuppression. This demonstrates different mediators of Treg-mediated immunosuppression, which mainly includes IL-10, TGF-β and IL-35, consumption of IL-2, IL-4, IL-7 and IL-35, or release of perforins/granzymes. TGF-β also plays both offensive (immunosuppression) and defensive (Treg protection) roles as it suppress T-effector functions but protects Tregs from the surrounding inflammatory environment.

Tregs are also involved in growth factor consumption and cytokine deprivation, and thus favor target cell apoptosis. This phenomenon of Treg-mediated immunosuppression involves competitive consumption of IL-2. However, under in vitro conditions, Tregs can immunosuppress IL-2R-deficient T cells in the presence of exogenous IL-2. This favors target T cell proliferation in the presence of Tregs while endogenous target T cell production of IL-2 remains suppressed (58). Tregs have been characterized as displaying a wide range of immunosuppressive mediators, including TGF-β, CTLA-4, IL-10 and galectin-1, although it is still uncertain which mode of immunosuppression is the main mediator of immunoregulatory properties (78). In addition, Tregs actively produce extracellular adenosine (ADP and AMP) and promote suppression of T effector cells through adenosine receptor signaling (79,80). CTLA-4 is a costimulation receptor and plays a crucial role in the development of T cell anergy. Some studies report that engagement of CTLA-4 with B7 on the APC leads to activation of the Treg (81), which further facilitates the release of several inhibitory cytokines such as IL-10 and TGF-β (82). Furthermore, proliferation of Tregs can be induced, leading to a positive feedback mechanism that ultimately results in the downregulation of T effector cell response in an antigenspecific manner (83). Tregs can either inhibit effector activity of conventional T cells or downregulate APC function of target cells (58). Additionally, Tregmediated immunosuppression can also operate through various mechanisms that involve not only cellular components but also unique proteins known as sirtuins (84). These cellular and molecular signals require close spatial proximity between Tregs and effector T cells. Sirtuin 1 (Srt1) has antiinflammatory properties, and its therapeutic targeting may be a valuable factor in organ transplantation (84–87). Activated Treg cells show downregulated Sirt1, and this may be a key process in stabilizing FOXP3 expression and Treg phenotypes (86), which may have clinical benefits in autoimmunity and transplantation (84,85,88). Information regarding the role of sertuins in the immune system has been sparse. However, enhanced Treg immunosuppressive activity and attenuated immune responses as a result of Srt1 deletion in CD4+ T and Treg cells have been reported, and both Srt1 deletion and Srt1 inhibition cause prolonged allograft survival (88,89). These investigations explain that the loss of Srt1 led to upregulation of proinflammatory cytokines and was required for deacetylating RelA/p65 in Tregs, which is required for the increased immunosuppressive capacity of Tregs (89). Therefore, Srt1 targeting can offer important therapeutic options for T cell-dependent immune responses in experimental models of transplantation, which enable allograft survival through enhanced Treg function in the fl-Sirt1/FOXP3+ cre model (88–90). Notably, effector T cell function was practically unaffected by Srt1 deletion. Furthermore, transfection of Srt1 and FOXP3+ into HEK 293 cancer cells prevents its proteasomal degradation through the deacetylation of FOXP3+ (85,86). Studies in sirtuin1 knockout mice reported that native Tregs express high FOXP3+, and inhibition or deletion of Srt1 can favor formation of acetylated FOXP3+, which is protected from proteasomal degradation (91).

Tregs and Angiogenesis

The process of angiogenesis in ischemic tissues is controlled by immune cells, which require Tregs and macrophages, with chemokines playing a key role in new vessel growth (92). Tregs play a key role in suppressing excessive immune response during an inflammatory response, and also support vascular repair at different levels (93). Loss of microvasculature may be an unappreciated root cause of chronic rejection for all solid-organ transplants (2,94). In clinical conditions, the ischemic phase favors the process of neovascularization (including vasculogenesis and angiogenesis) and characterizes the tissue microvascular repair and remodeling during allograft rejection (94–97). In addition to tissue-specific initial activators, neovascularization requires growth factors, chemokines and proteases that play distinct roles in promoting and refining tissue repair and regeneration. Most of the cellular machinery of the immune system plays a key role during the process of microvascular repair (98). The involvement of T lymphocytes is also shown in microvascular and vessel development and, as reported, T cell-deficient nude mice exhibit a distinct reduction in microvascular and vessel growth (99,100). Furthermore, it has been proposed that CD4+ and CD8+ cells play a key role in vascular remodeling, as CD4- and CD8-deficient mice display a major reduction in vessel growth (101). In addition, leukocytes release angiogenic mediators, including vascular endothelial growth factor (VEGF) and proinflammatory cytokines TNF-α and IL-1β, which initiate neovascularization and microvascular establishment and organize the tissue response to ischemic conditions (102,103). A number of cytokines secreted by pathogenic T cells affect the survival and function of Treg cells, specifically IL-2 released by peripheral pathogenic T cells after CD28 interaction multiplies the Treg cell population (72). Numerous costimulatory signals have been involved in inflammatory T cell activation and differentiation, of which the B7/CD28 and CD40L/CD40 pathways play key roles, which suggests that loss of Treg cell-mediated self-tolerance affects both T and B cell-mediated tolerance (24). Of note, CD28 interactions with the ligands B7-1 (CD80) and B7-2 (CD86) are also vital for the development of CD4+CD25+FOXP3+ Treg cells (104). In addition, release of immunosuppressive cytokines TGF-β and IL-10 by Treg cells suppresses dendritic cells, which further inactivates effector T cells and monocytes (8,71). Expression of IL-10 and TGF-β by Tregs offers a possible mechanism to explain how these cells limit inflammatory injury and possibly accelerate recovery (1). Furthermore, Tregs also modulate inflammatory responses of innate immune cells, including macrophages, monocytes, DCs, NK cells and the complement activation system, which highlights that the immunoregulatory role of Tregs is not limited to the adaptive immune system (105).

The complement pathway is a unique part of innate immunity, and the receptors C3aR and C5aR are present in a variety of cells, including Tregs, and signaling through both C3aR and C5aR on nTregs cells has been reported to inhibit regulatory functions of Tregs (106). Inhibition or genetic deficiency of both C3aR and C5aR on nTreg cells augments their in vitro and in vivo suppressive activity and prolongs skin allograft survival (106). In addition, C3aR/C5aR deficiency or inhibition further triggers the activation of murine iTreg cells, stabilizes expression of the FOXP3 gene and precludes iTreg conversion to IFN-γ/TNF-α-producing T effector cells, thereby limiting graftversus-host disease (106). Liu et al. showed an antagonistic relation between CD4+CD25− T cells and CD4+CD25+ Treg cells on the polarization of macrophage phenotypes (107). They highlighted that CD4+CD25+ Treg favors M2 macrophage polarization, whereas M1 macrophages can be induced by CD4+CD25− T effector cells (108). Phenotypically, M2 macrophages play a crucial antiinflammatory and reparative role by secretion of IL-10, IL-1β and TGF-β, which regulate tissue repair and promote angiogenesis through VEGF secretion (109). Tregs affect angiogenesis through both indirect and direct mechanisms and have the potential to stimulate angiogenesis indirectly by Th1 cell suppression through the release of TNFα and IFN-γ cytokines, as well as interferon-induced chemokines such as CXCL9, ™10 and ™11 (110). Further, CD4+CD25+ Tregs release a surplus of VEGF at the steady state as well as under hypoxic conditions when compared with CD4+CD25−T cells, while demonstrating capillary formation in vitro through VEGF signaling (111). Also, it was recently reported in a mouse model of orthotopic tracheal transplantation that adoptive transfer of CD4+CD25+FOXP3+ Tregs ameliorated functional microvascular blood flow between donor and recipient grafts, which further facilitated allograft recovery from the severe hypoxia phase (112,113). In addition, supernatants of hypoxic Tregs were able to promote angiogenesis in vivo in cell-free Matrigel implants (114). As reported in a left lung ischemia model, an increase in CD4+CD25+FOXP3+ Tregs cells was observed 3–5 d after the onset of ischemia in C57Bl/6 WT mice (93). Further experiments with adoptive transfer of CD4+CD25+ lymphocytes into FOXP3+ Treg-depleted mice showed almost complete recovery of the angiogenic phenotype (115). Furthermore, recent studies highlighted a relationship between Tregs and vascular wall function in cardiovascular disease (116), showing that an increase in apoptotic Tregs triggers induction of vascular inflammation and impaired endothelium-dependent relaxation in coronary arterioles in hypertension, whereas reconstitution of Tregs subdues arterial blood pressure and improves coronary arteriolar endothelial function through the release of IL-10 (116) (Figure 3).

Figure 3
figure 3

Treg therapy and immunotolerance. This demonstrates ex vivo and in vivo expansion of freshly isolated Tregs for cell therapy to rescue allograft rejection. Adoptive transfer of Tregs shows downregulation of CD4+ T cells followed by upregulation of Th2 responses, which favor microvascular and tissue repair in rejecting allograft.

Treg-Mediated Immunotherapy in Transplants

Organ transplantation can be a life-saving procedure for patients with end-stage disease of the lung, heart, kidney or liver. Unfortunately, this treatment strategy is limited by chronic rejection of the transplanted organ, which occurs when the patient’s immune system continually attacks and impairs the organ and ceases vascular flow required for graft survival (117,118). This process affects nearly all patients in the first 10 years following transplantation, and there is no effective therapy for this condition. Treg cell-based therapy can be achieved by administering Tregs cells to diseased patients (119). Tregs (CD4+CD25+FOXP3+) play a crucial role in self-tolerance and graft immunity as well as in controlling infections, and outcomes of preclinical models have recognized them as a vital candidate for cell therapy, e.g., for the treatment of transplant-related complications such as graft-versus-host disease following allogeneic stem cell transplantation (15,120). Currently, different translational approaches have been utilized to induce Tregs though anti-TNF receptor super family member 25 (121), IL-2/mAb complexes (122), anti-CD45RB (123), rapamycin-mediated (124), mitomycin C-incubated myeloid blood cells (MICs), regulatory macrophages (Mregs) (125) and regulatory dendritic cells (DCregs) (126), which show positive therapeutic effects in preclinical studies (127,128). Tregs are mainly isolated and expanded ex vivo, but few preclinical studies have successfully expanded Treg populations through in vivo treatment with either anti-CD45RB antibody or anti-TNF receptor super family member 25 antibody or IL-2-IL2 complex or recombinant IL-33 (123,129–132) (Figure 3). TNFRSF25, also known as DR3, is constitutively and highly expressed by CD4+FOXP3+ Tregs, which are mostly involved in autoimmunity (121), while CD45 is a protein tyrosine phosphatase receptor type C that regulates T and B cell antigen receptor signaling and lymphocyte activation (133), and anti-CD45RB is a potent tolerogenic molecule that works by boosting the Treg number (123). This immune modulation of Tregs occurs by specifically inducing proliferative expansion of Tregs in vivo, and recent data have suggested that this occurs through specific enhancement of interactions between Tregs and APCs via an unknown mechanism but could potentially be a milestone discovery for in vivo Treg expansion if replicated in humans (123). In contrast, ex vivo Treg cell-based therapy is a clinically approved strategy to harness the immunosuppressive properties of Tregs for therapeutic use (134). In this therapeutic approach, Tregs are isolated from a patient, enriched, expanded ex vivo and adoptively transferred to the patient (135) (Figure 3). This cell-based therapeutic approach is beneficial because the expanded Treg cell population can be screened phenotypically and functionally prior to adoptive transfer under controlled conditions (135).

Several immunotherapeutic strategies implicating the use of Tregs have been developed, some of which have been used in clinical trials in organ transplantation (Table 1) (135). Preclinical and clinical studies have demonstrated the therapeutic relevance of Tregs in allograft survival in different transplantation models (52,136–138). Several clinical studies have shown an increase in peripheral CD4+CD25high T cells in operationally tolerant liver transplant recipients (60,139,140), and an Initial observation that CD4+CD25high T cells play a pivotal role in transplantation tolerance has been well demonstrated in mouse models (20). Sakaguchi et al. showed depletion of CD4+CD25high T cells from enhanced graft rejection, while dose-dependent reconstitution of CD4+CD25high T cells prolonged allograft survival (59,141,142). Clinical studies in transplantation have investigated the number and functional properties of regulatory CD4+CD25high Tregs in relation to immunological quiescence, tolerance and acute or chronic rejection (143,144). Moreover, Tregs known to be crucial in the maintenance of peripheral immune tolerance are a critical modulator of post-ischemic neovascularization in a hind limb ischemia model (145). In humans, there are only few distinctive markers to distinguish CD4+CD25high T cells from conventional activated T cells. However, the α-chain of the IL-7 receptor (CD127) allows a clear variation between Treg-activated CD4+CD25+ T cells (146,147); further, this marker could also be used in patients after solid organ transplantation of liver and kidney (127,128), and CD127-based characterized Treg and activated T cell subsets have been reported to be differentially distributed in healthy individuals as compared to transplant recipients (148). However, the ratio of activated T cell subsets among CD4+CD25high T cells was augmented in stable liver and kidney transplant recipients as compared to healthy individuals (149). The use of expanded Tregs and iTregs compared to nTregs requires more phenotypic evaluation for safety and quality control. CD127 is less useful after strong activation, for example, as it can downregulate on conventional T cells (150). As discussed earlier, the plasticity and safety of expanded Tregs mainly depends on their state of FOXP3 expression (151). The standard therapeutic intervention after transplantation should induce tolerance, and regulatory T cells play a pivotal role in maintaining homeostasis and self-tolerance through the modulation of immune effector functions (7,16). Treg cells can be found inside the tolerated graft, and these cells can have indirect allospecificity for donor antigens (151). In patients transplanted with lung, liver or kidney grafts, a positive correlation between graft survival and the number of circulating Treg cells has been reported in both preclinical and clinical conditions (152), and based on these observations, it is widely accepted that Tregs play a pivotal role in the induction of transplantation tolerance (153–155). Therefore, this supports the possibility of using Tregs as a biological therapy to maintain tolerance to alloantigens. Preclinical research findings report that reconstitution of Tregs has been shown to ameliorate graft-versus-host disease and facilitate engraftment of the bone marrow (156–158). Originally, natural Tregs had to maintain immunological self-tolerance, but deficiency or dysfunction of these cells may lead to the onset of autoimmune disease (5). However, it was later realized that a decline in their number or function can also provoke tumor immunity (159), whereas their antigen-specific subset expansion can potentiate transplantation tolerance (5). Furthermore, Treg-mediated immunotolerance has been implicated in other pathological conditions including allergies, microbial infections and fetomaternal tolerance (120,160,161), and in organ transplantation (52). Based on these outcomes, elevating Treg numbers or their suppressive properties may be key in treating autoimmune disease and preventing allograft rejection. On the other hand, depletion of Treg cells or inhibition of their regulatory function could enhance immunity against tumors and chronic infectious agents (162).

Table 1 List of clinical trials of T regulatory cell-based immunotherapy.

Treg Plasticity and Safety

FOXP3+ plays an important role in the development and immunosuppressive function of Tregs (8,80,163). Treg functions are modulated through transcription as well as post-translation modifications, including lysine residue acetylation, which protects FOXP3+ from degradation and thus promotes optimum Treg function (163). The acetylation process of the FOXP3+ gene is structured by the histone/protein acetyltransferases and histone/protein deacetylases (89). Interestingly, targeted deletion of sirtuin1 upregulates the process of acetylation and ultimately the expression of FOXP3+ and augments the immunosuppressive mechanism of Tregs (88). However, deletion or inhibition of sirtuin1 attenuates allograft rejection and prolongs survival of murine cardiac allografts (85). Based on the origin, two different forms of FOXP3 Tregs exist, of which naturally occurring CD4+CD25+FOXP3+ Tregs (nTregs) originate in the thymus after TCR stimulation through MHC self-antigen complex interaction, followed by signaling through CD28 and CD25 (164), while induced Tregs (iTregs) develop from naive CD4+ T cells through TGF-β1 stimulation (164), and more specifically, generation of iTregs has been explained in GALT, spleen, lymph node, chronically inflamed and transplanted tissues (165,166). Functionally, under Th17 or Th1 cell-polarizing conditions, nTreg cells differentiated to a substantial fraction of IL-17+FOXP3+ or IFNγ+FOXP3+ cells arose without downregulation of FOXP3 expression (167–169). In contrast, under Th17 cell-polarizing conditions, TGF-β-induced Tregs lost their FOXP3 expression and acquired IL-17 expression (168). There is a notion that the difference in FOXP3 stability between nTreg cells and iTreg cells is due to epigenetic modifications at the FOXP3 locus (170), which contains a highly conserved CpG-rich region upstream of exon −1 and is referred to as the Treg cell-specific demethylated region (171–173). This specific FOXP3 locus is fully demethylated in nTreg cells but remains methylated in iTreg cells and activated human conventional T cells that transiently express FOXP3 (164).

Tregs have been shown to display a unique feature that depends on their FOXP3 expression and the demethylation state of the conserved non-coding region 2/Treg-specific demethylated region and Treg cell representative regions. This feature is the indicator of Treg exposure to specific antigens, the strength and duration of costimulation, and T cell receptor signaling. Therefore, genetically the Treg population is a mishmash of FOXP3+epigenome+ stable Tregs, potential Tregs (FOXP 3−epigenome+) and transient Tregs (FOXP3+epigenome−). The stability and plasticity of Tregs are usually affected by the balance between their intrinsic FOXP3 expression, stabilizing and destabilizing signals that regulate their effective immunosuppressive function. The association of these key functions is likely to be apparent and influenced by the developmental, environmental and inflammatory microenvironment and state of tolerance, thereby generating different mediators to initiate Treg stability or instability (174).

Conclusions

Recent research has highlighted the cellular and molecular basis of Treg development and function, and implicated dysregulation of Tregs in major immunological diseases, including allograft rejection (19,23). Tregs are instrumental in establishing immune tolerance and are important cellular mediators of cell-based therapy for clinical applications (135). Efforts to unravel the complexity of Tregs are only just beginning, and further understanding of their biology and characterization of targets will undoubtedly enhance future therapeutic opportunities (10,88,134). Increasing evidence indicates that Tregs could be used to inhibit pathogenic anti-transplant immunity (in the absence of immune suppression), but mechanisms to accomplish this goal are hampered by inadequate understanding, Treg expansion, cost of GMP Treg manufacturing, safety and difficulties with the stability of the Treg phenotype after adoptive transfer. The present and future therapeutic scope of Treg-based therapy will hopefully minimize the drug burden of immunosuppression on solid organ transplant patients. In coming years, Treg cell-based therapy will provide a novel therapeutic platform in transplantation as well as in other diseases. This will further assist both clinicians and researchers in testing the combinations of current drug-based therapies with Tregs, which will further minimize the side effects and thus morbidity caused by these toxic immunosuppressants (175). Treg-based immunotherapy assures antigen-specific immunosuppression and cell dosage can be tightly controlled and affect long-lasting regulation in vivo, and can be individualized to each patient with very limited side effects (135). Tregs possess unique defensive and offensive mechanisms, antigen specificity and promising therapeutic potential, as reported in both preclinical and clinical studies of solid organ transplantation, but these cells do not have sufficient efficacy as a stand-alone therapy to prevent chronic rejection in solid organ transplantation, and certain factors, including dose, specificity and plasticity, remain uncertain and challenging areas to uncover (176). The future of Treg-based immunotherapy is dependent on effective clinical trials, technological advancements in Treg manufacture/expansion and better mechanistic understanding of Treg biology and transplantation tolerance in humans.

In summary, Treg-based immune control can be implemented as a potential pharmaceutical tool through an adoptive cell therapy protocol to rescue patients with inflammatory diseases, chronic inflammation and transplantation. This review highlights the clinical significance of Tregs and also emphasizes the difficulties encountered in transitioning from bench to bedside. Although Treg therapy is very effective and does not present side effects, various challenges, including different methods of Treg expansion, the cost of GMP-Treg manufacturing, safety, and difficulties with stability of the Treg phenotypes after adoptive transfer mean that its use as a standard therapy remains a challenge.

Disclosure

The author declares that he has no competing interests as defined by Molecular Medicine, or other interests that might be perceived to influence the results and discussion reported in this paper.

References

  1. Jager A, Kuchroo VK. (2010) Effector and regulatory T-cell subsets in autoimmunity and tissue inflammation. Scand. J. Immunol. 72:173–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Khan MA, Nicolls MR. (2013) Complement-mediated microvascular injury leads to chronic rejection. Adv. Exp. Med. Biol. 734:233–46.

    Article  CAS  Google Scholar 

  3. Janeway CA, Jr., Medzhitov R. (2002) Innate immune recognition. Annu. Rev. Immunol. 20:197–216.

    Article  CAS  PubMed  Google Scholar 

  4. Kew RR, Ghebrehiwet B, Janoff A. (1987) Characterization of the third component of complement (C3) after activation by cigarette smoke. Clin. Immunol. 44:248–58.

    CAS  Google Scholar 

  5. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. (2008) Regulatory T cells and immune tolerance. Cell. 133:775–87.

    Article  CAS  Google Scholar 

  6. Dinavahi R, Heeger PS. (2008) T-cell immune monitoring in organ transplantation. Curr. Opin. Organ Transplant. 13:419–24.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Fehervari Z, Sakaguchi S. (2004) CD4+ Tregs and immune control. J. Clin. Invest. 114:1209–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sakaguchi S, Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T. (2009) Regulatory T cells: how do they suppress immune responses? Int. Immunol. 21:1105–11.

    Article  CAS  PubMed  Google Scholar 

  9. Shalev I, Selzner N, Shyu W, Grant D, Levy G. (2012) Role of regulatory T cells in the promotion of transplant tolerance. Liver Transpl. 18:761–70.

    Article  PubMed  Google Scholar 

  10. Onishi H, Morisaki T, Katano M. (2012) Immunotherapy approaches targeting regulatory T-cells. Anticancer Res. 32:997–1003.

    CAS  PubMed  Google Scholar 

  11. Nouel A, Simon Q, Jamin C, Pers JO, Hillion S. (2014) Regulatory B cells: an exciting target for future therapeutics in transplantation. Front. Immunol. 5: 11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Chesneau M, et al. (2015) Tolerant Kidney Transplant Patients Produce B Cells with Regulatory Properties. Clin. J. Am. Soc. Nephrol. 26:2588–98.

    Article  CAS  Google Scholar 

  13. Mandal A, Viswanathan C. (2015) Natural killer cells: In health and disease. Hematol. Oncol. Stem Cell Ther. 8:47–55.

    Article  CAS  PubMed  Google Scholar 

  14. Conde P, et al. (2015) DC-SIGN(+) Macrophages Control the Induction of Transplantation Tolerance. Immunity. 42:1143–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bilate AM, Lafaille JJ. (2012) Induced CD4+Foxp3+ regulatory T cells in immune tolerance. Annu. Rev. Immunol. 30:733–58.

    Article  CAS  PubMed  Google Scholar 

  16. Josefowicz SZ, Lu LF, Rudensky AY. (2012) Regulatory T cells: mechanisms of differentiation and function. Annu. Rev. Immunol. 30:531–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Issa F, Wood KJ. (2010) CD4+ regulatory T cells in solid organ transplantation. Curr. Opin. Organ Transpl. 15:757–64.

    Article  Google Scholar 

  18. Sakaguchi S. (2011) Regulatory T cells: history and perspective. Methods Mol. Biol. 707:3–17.

    Article  CAS  PubMed  Google Scholar 

  19. Sakaguchi S, et al. (2001) Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol. Rev. 182:18–32.

    Article  CAS  PubMed  Google Scholar 

  20. Wood KJ, Sakaguchi S. (2003) Regulatory T cells in transplantation tolerance. Nat. Rev. Immunol. 3:199–210.

    Article  CAS  PubMed  Google Scholar 

  21. Khan MA, Moeez S, Akhtar S. (2013) T-regulatory cell-mediated immune tolerance as a potential immunotherapeutic strategy to facilitate graft survival. Blood Transfus. 11:357–63.

    PubMed  PubMed Central  Google Scholar 

  22. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. (1995) 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. 155:1151–64.

    CAS  Google Scholar 

  23. Sakaguchi S. (2004) Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses. Ann. Rev. Immunol. 22:531–62.

    Article  CAS  Google Scholar 

  24. Sakaguchi S. (2000) Regulatory T cells: key controllers of immunologic self-tolerance. Cell. 101:455–58.

    Article  CAS  PubMed  Google Scholar 

  25. Gershon RK, Kondo K. (1970) Cell interactions in the induction of tolerance: the role of thymic lymphocytes. Immunology. 18:723–37.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Sakaguchi S, et al. (2006) Foxp3+ CD25+ CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol. Rev. 212:8–27.

    Article  CAS  Google Scholar 

  27. Tiemessen MM, et al. (2007) CD4+CD25+Foxp3+ regulatory T cells induce alternative activation of human monocytes/macrophages. Proc. Natl. Acad. Sci. USA. 104:19446–51.

    Article  CAS  PubMed  Google Scholar 

  28. Hall BM, Jelbart ME, Gurley KE, Dorsch SE. (1985) Specific unresponsiveness in rats with prolonged cardiac allograft survival after treatment with cyclosporine. Mediation of specific suppression by T helper/inducer cells. J. Exp. Med. 162:1683–94.

    Article  CAS  PubMed  Google Scholar 

  29. Shevach EM. (2009) Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity. 30:636–45.

    Article  CAS  PubMed  Google Scholar 

  30. Roncarolo MG, Gregori S. (2008) Is FOXP3 a bona fide marker for human regulatory T cells? Eur. J. Immunol. 38:925–27.

    Article  CAS  PubMed  Google Scholar 

  31. Zelenika D, et al. (2001) The role of CD4+ T-cell subsets in determining transplantation rejection or tolerance. Immunol. Rev. 182:164–79.

    Article  CAS  PubMed  Google Scholar 

  32. Shin JY, et al. (2015) CD4+VEGFR1(HIGH) T cell as a novel Treg subset regulates inflammatory bowel disease in lymphopenic mice. Cell. Mol. Immunol. 12:592–603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhang H, et al. (2014) Subsets of regulatory T cells and their roles in allergy. J. Transl. Med. 12:125.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Sakaguchi S, et al. (2003) Thymic generation and selection of CD25+CD4+ regulatory T cells: implications of their broad repertoire and high self-reactivity for the maintenance of immunological self-tolerance. Novartis Found. Symp. 252:6–16; discussion 16–23, 106–14.

    CAS  PubMed  Google Scholar 

  35. Ito T, et al. (2008) Two functional subsets of FOXP3+ regulatory T cells in human thymus and periphery. Immunity. 28:870–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Sim GC, et al. (2014) IL-2 therapy promotes suppressive ICOS+ Treg expansion in melanoma patients. J. Clin. Invest. 124:99–110.

    Article  CAS  PubMed  Google Scholar 

  37. Coquerelle C, et al. (2009) Anti-CTLA-4 treatment induces IL-10-producing ICOS+ regulatory T cells displaying IDO-dependent anti-inflammatory properties in a mouse model of colitis. Gut. 58: 1363–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hsu P, et al. (2015) IL-10 Potentiates Differentiation of Human Induced Regulatory T Cells via STAT3 and Foxo1. J. Immunol. 195:3665–74.

    Article  CAS  PubMed  Google Scholar 

  39. Hippen KL, et al. (2011) Generation and large-scale expansion of human inducible regulatory T cells that suppress graft-versus-host disease. Am. J. Transpl. 11:1148–57.

    Article  CAS  Google Scholar 

  40. Peng X, et al. (2014) CD4+CD25+FoxP3+ Regulatory Tregs inhibit fibrocyte recruitment and fibrosis via suppression of FGF-9 production in the TGF-beta1 exposed murine lung. Front. Pharmacol. 5:80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Cobbold SP, Graca L, Lin CY, Adams E, Waldmann H. (2003) Regulatory T cells in the induction and maintenance of peripheral transplantation tolerance. Transplant Int. 16:6675.

    Article  Google Scholar 

  42. Li H, et al. (2015) Interferon-gamma and tumor necrosis factor-alpha promote the ability of human placenta-derived mesenchymal stromal cells to express programmed death ligand-2 and induce the differentiation of CD4(+)interleukin-10(+) and CD8(+)interleukin-10(+)Treg subsets. Cytotherapy. 17:1560–71.

    Article  CAS  PubMed  Google Scholar 

  43. Li S, et al. (2014) A naturally occurring CD8(+) CD122(+) T-cell subset as a memory-like Treg family. Cell. Mol. Immunol. 11:326–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Voo KS, et al. (2009) Identification of IL-17-producing FOXP3+ regulatory T cells in humans. Proc. Natl. Acad. Sci. USA. 106:4793–98.

    Article  CAS  PubMed  Google Scholar 

  45. Thornton AM, Shevach EM. (1998) CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J. Exp. Med. 188:287–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Takahashi T, et al. (1998) Immunologic self-tolerance maintained by CD25+CD4+ naturally anergic and suppressive T cells: induction of autoimmune disease by breaking their anergic/suppressive state. Int. Immunol. 10:1969–80.

    Article  CAS  PubMed  Google Scholar 

  47. Yamazaki S, et al. (2003) Direct expansion of functional CD25+ CD4+ regulatory T cells by antigen-processing dendritic cells. J. Exp. Med. 198:235–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Trenado A, et al. (2003) Recipient-type specific CD4+CD25+ regulatory T cells favor immune reconstitution and control graft-versus-host disease while maintaining graft-versus-leukemia. J. Clin. Invest. 112:1688–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Golshayan D, et al. (2007) In vitro-expanded donor alloantigen-specific CD4+CD25+ regulatory T cells promote experimental transplantation tolerance. Blood. 109:827–35.

    Article  CAS  PubMed  Google Scholar 

  50. Joffre O, et al. (2008) Prevention of acute and chronic allograft rejection with CD4+ CD25+Foxp3+ regulatory T lymphocytes. Nat. Med. 14:88–92.

    Article  CAS  PubMed  Google Scholar 

  51. Sagoo P, et al. (2011) Human regulatory T cells with alloantigen specificity are more potent inhibitors of alloimmune skin graft damage than polyclonal regulatory T cells. Sci. Transl. Med. 3:83ra42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Todo S, et al. (2016) A Pilot Study of Operational Tolerance with a Regulatory T Cell-Based Cell Therapy in Living Donor Liver Transplantation. Hepatology. 64:632–43.

    Article  CAS  PubMed  Google Scholar 

  53. Walker LS. (2004) CD4+ CD25+ Treg: divide and rule? Immunology. 111:129–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Adalid-Peralta L, Fragoso G, Fleury A, Sciutto E. (2011) Mechanisms underlying the induction of regulatory T cells and its relevance in the adaptive immune response in parasitic infections. Int. J. Biol. Sci. 7:1412–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Walsh PT, Taylor DK, Turka LA. (2004) Tregs and transplantation tolerance. J. Clin. Invest. 114:1398–1403.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Scheffold A, Murphy KM, Hofer T. (2007) Competition for cytokines: T(reg) cells take all. Nat. Immunol. 8:1285–87.

    Article  CAS  PubMed  Google Scholar 

  57. Schmidt A, Oberle N, Krammer PH. (2012) Molecular mechanisms of Treg-mediated T cell suppression. Front. Immunol. 3:51.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Sojka DK, Huang YH, Fowell DJ. (2008) Mechanisms of regulatory T-cell suppression: a diverse arsenal for a moving target. Immunology. 124:13–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kingsley CI, Karim M, Bushell AR, Wood KJ. (2002) CD25+CD4+ regulatory T cells prevent graft rejection: CTLA-4- and IL-10-dependent immunoregulation of alloresponses. J. Immunol. 168:1080–86.

    Article  CAS  PubMed  Google Scholar 

  60. Koshiba T, et al. (2007) Clinical, immunological, and pathological aspects of operational tolerance after pediatric living-donor liver transplantation. Transpl. Immunol. 17:94–97.

    Article  CAS  PubMed  Google Scholar 

  61. Bunnag S, et al. (2008) FOXP3 expression in human kidney transplant biopsies is associated with rejection and time post transplant but not with favorable outcomes. Am. J. Transpl. 8:1423–33.

    Article  CAS  Google Scholar 

  62. Muthukumar T, et al. (2005) Messenger RNA for FOXP3 in the urine of renal-allograft recipients. N. Engl. J. Med. 353:2342–51.

    Article  CAS  PubMed  Google Scholar 

  63. Boasso A, Herbeuval JP, Hardy AW, Winkler C, Shearer GM. (2005) Regulation of indoleamine 2,3-dioxygenase and tryptophanyl-tRNA-synthetase by CTLA-4-Fc in human CD4+ T cells. Blood. 105:1574–81.

    Article  CAS  PubMed  Google Scholar 

  64. Mezrich JD, et al. (2010) An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J. Immunol. 185:3190–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Xie FT, et al. (2015) IDO expressing dendritic cells suppress allograft rejection of small bowel transplantation in mice by expansion of Foxp3+ regulatory T cells. Transpl. Immunol. 33:69–77.

    Article  CAS  PubMed  Google Scholar 

  66. Li MO, Wan YY, Flavell RA. (2007) T cell-produced transforming growth factor-beta1 controls T cell tolerance and regulates Th1- and Th17-cell differentiation. Immunity. 26:579–91.

    Article  CAS  PubMed  Google Scholar 

  67. Probst-Kepper M, et al. (2009) GARP: a key receptor controlling FOXP3 in human regulatory T cells. J. Cell. Mol. Med. 13:3343–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Wang H, et al. (2008) TGF-beta-dependent suppressive function of Tregs requires wild-type levels of CD18 in a mouse model of psoriasis. J. Clin. Invest. 118:2629–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Wang R, Wan Q, Kozhaya L, Fujii H, Unutmaz D. (2008) Identification of a regulatory T cell-specific cell surface molecule that mediates suppressive signals and induces Foxp3 expression. PloS One. 3:e2705.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Tran DQ. (2012) TGF-beta: the sword, the wand, and the shield of FOXP3(+) regulatory T cells. J. Mol. Cell Biol. 4:2937.

    Article  CAS  Google Scholar 

  71. Taylor A, Verhagen J, Blaser K, Akdis M, Akdis CA. (2006) Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: the role of T regulatory cells. Immunology. 117:433–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Zheng SG, Wang J, Wang P, Gray JD, Horwitz DA. (2007) 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. 178:2018–27.

    Article  CAS  PubMed  Google Scholar 

  73. Zheng SG, Wang JH, Gray JD, Soucier H, Horwitz DA. (2004) Natural and induced CD4+CD25+ cells educate CD4+CD25− cells to develop suppressive activity: the role of IL-2, TGF-beta, and IL-10. J. Immunol. 172:5213–21.

    Article  CAS  PubMed  Google Scholar 

  74. Zheng SG. (2008) The Critical Role of TGF-beta1 in the Development of Induced Foxp3+ Regulatory T Cells. Int. J. Clin. Exper. Mede 1:192–202.

    CAS  Google Scholar 

  75. Jones CP, Gregory LG, Causton B, Campbell GA, Lloyd CM. (2012) Activin A and TGF-beta promote T(H)9 cell-mediated pulmonary allergic pathology. J. Allergy Clin. Immunol. 129:1000–10, e1003.

    Google Scholar 

  76. Schmetterer KG, Neunkirchner A, Pickl WF. (2012) Naturally occurring regulatory T cells: markers, mechanisms, and manipulation. FASEB J. 26:2253–76.

    Article  CAS  PubMed  Google Scholar 

  77. Collison LW, et al. (2007) The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature. 450:566–69.

    Article  CAS  PubMed  Google Scholar 

  78. Kubach J, et al. (2007) Human CD4+CD25+ regulatory T cells: proteome analysis identifies galectin-10 as a novel marker essential for their anergy and suppressive function. Blood. 110:1550–58.

    Article  CAS  PubMed  Google Scholar 

  79. Brandenburg S, et al. (2008) IL-2 induces in vivo suppression by CD4(+)CD25(+)Foxp3(+) regulatory T cells. Eur. J. Immunol. 38:1643–53.

    Article  CAS  PubMed  Google Scholar 

  80. Ohta A, et al. (2012) The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front. Immunol. 3:190.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Chen L, Flies DB. (2013) Molecular mechanisms of T cell co-stimulation and co-inhibition. Nature reviews. Immunology. 13:227–42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Takahashi T, et al. (2000) Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J. Exp. Med. 192:303–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Corthay A. (2009) How do regulatory T cells work? Scand. J. Immunol. 70:326–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Kong S, McBurney MW, Fang D. (2012) Sirtuin 1 in immune regulation and autoimmunity. Immunol. Cell Biol. 90:613.

    Article  CAS  Google Scholar 

  85. Beier UH, et al. (2011) Sirtuin-1 targeting promotes Foxp3+ T-regulatory cell function and prolongs allograft survival. Mol. Cell. Biol. 31:1022–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. van Loosdregt J, et al. (2010) Regulation of Treg functionality by acetylation-mediated Foxp3 protein stabilization. Blood. 115:965–74.

    Article  PubMed  CAS  Google Scholar 

  87. Fairchild RL. (2016) Juicing Tregs in situ to improve kidney allograft outcomes. Kidney Int. 89:976–78.

    Article  CAS  PubMed  Google Scholar 

  88. Akimova T, et al. (2014) Targeting sirtuin-1 alleviates experimental autoimmune colitis by induction of Foxp3+ T-regulatory cells. Mucosal Immunol. 7:1209–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Beier UH, Akimova T, Liu Y, Wang L, Hancock WW. (2011) Histone/protein deacetylases control Foxp3 expression and the heat shock response of T-regulatory cells. Curr. Opin. Immunol. 23:670–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Beier UH, et al. (2012) Histone deacetylases 6 and 9 and sirtuin-1 control Foxp3+ regulatory T cell function through shared and isoform-specific mechanisms. Sci. Signal. 5:ra45.

    Article  PubMed  PubMed Central  Google Scholar 

  91. van Loosdregt J, et al. (2011) Rapid temporal control of Foxp3 protein degradation by sirtuin-1. PloS One. 6:e19047.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Owen JL, Mohamadzadeh M. (2013) Macrophages and chemokines as mediators of angiogenesis. Front. Physiol. 4:159.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. D’Alessio FR, Zhong Q, Jenkins J, Moldobaeva A, Wagner EM. (2015) Lung Angiogenesis Requires CD4(+)Forkhead Homeobox Protein-3(+) Regulatory T Cells. Am. J. Respir. Cell Mol. Biol. 52:603–10.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Khan MA, et al. (2011) CD4+ T cells and complement independently mediate graft ischemia in the rejection of mouse orthotopic tracheal transplants. Circ. Res. 109:1290–1301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Cravedi P, et al. (2013) Immune cell-derived C3a and C5a costimulate human T cell alloimmunity. Am. J. Transpl. 13:2530–39.

    Article  CAS  Google Scholar 

  96. Jiang X, et al. (2011) Adenovirus-mediated HIF-1alpha gene transfer promotes repair of mouse airway allograft microvasculature and attenuates chronic rejection. J. Clin. Invest. 121:2336–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Khan MA, et al. (2013) Targeting complement component 5a promotes vascular integrity and limits airway remodeling. Proc. Natl. Acad. Sci. USA. 110:6061–66.

    Article  CAS  PubMed  Google Scholar 

  98. Babu AN, et al. (2007) Microvascular destruction identifies murine allografts that cannot be rescued from airway fibrosis. J. Clin. Invest. 117:3774–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Waeckel L, et al. (2005) Impairment in postischemic neovascularization in mice lacking the CXC chemokine receptor 3. Circ. Res. 96:576–82.

    Article  CAS  PubMed  Google Scholar 

  100. Silvestre JS, Mallat Z, Tedgui A, Levy BI. (2008) Post-ischaemic neovascularization and inflammation. Cardiovasc. Res. 78:242–49.

    Article  CAS  PubMed  Google Scholar 

  101. Stabile E, et al. (2006) CD8+ T lymphocytes regulate the arteriogenic response to ischemia by infiltrating the site of collateral vessel development and recruiting CD4+ mononuclear cells through the expression of interleukin-16. Circulation. 113:118–24.

    Article  PubMed  Google Scholar 

  102. Thurston G, Gale NW. (2004) Vascular endothelial growth factor and other signaling pathways in developmental and pathologic angiogenesis. Int. J. Hematol. 80:7–20.

    Article  CAS  PubMed  Google Scholar 

  103. Liu X, et al. (2014) IL-1beta Upregulates IL-8 Production in Human Muller Cells Through Activation of the p38 MAPK and ERK1/2 Signaling Pathways. Inflammation. 37:1486–95.

    Article  CAS  PubMed  Google Scholar 

  104. Zeng M, Guinet E, Nouri-Shirazi M. (2009) B7-1 and B7-2 differentially control peripheral homeostasis of CD4(+)CD25(+)Foxp3(+) regulatory T cells. Transpl. Immunol. 20:171–79.

    Article  CAS  PubMed  Google Scholar 

  105. Andre S, Tough DF, Lacroix-Desmazes S, Kaveri SV, Bayry J. (2009) Surveillance of antigen-presenting cells by CD4+ CD25+ regulatory T cells in autoimmunity: immunopathogenesis and therapeutic implications. Am. J. Pathol. 174:1575–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Khan MA, Hsu JL, Assiri AM, Broering DC. (2015) Targeted complement inhibition and microvasculature in transplants: A therapeutic perspective. Clin. Exper. Immunol. 183:175–86.

    Article  Google Scholar 

  107. Liu G, et al. (2011) Phenotypic and functional switch of macrophages induced by regulatory CD4+CD25+ T cells in mice. Immunol. Cell Biol. 89:130–42.

    Article  CAS  PubMed  Google Scholar 

  108. Yu X, Li H, Ren X. (2012) Interaction between regulatory T cells and cancer stem cells. Int. J. Cancer. 131:1491–98.

    Article  CAS  PubMed  Google Scholar 

  109. Kyurkchiev D, et al. (2014) Secretion of immunoregulatory cytokines by mesenchymal stem cells. World J. Stem Cells. 6:552–70.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Muller-Hermelink N, et al. (2008) TNFR1 signaling and IFN-gamma signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell. 13:507–18.

    Article  PubMed  CAS  Google Scholar 

  111. Facciabene A, Motz GT, Coukos G. (2012) T-regulatory cells: key players in tumor immune escape and angiogenesis. Cancer Res. 72:2162–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Afzal Khan M, Ahmed HA, Hasan AF, Altuhami A, Assiri AM, Clemens D, Broering DC. (2016) The therapeutic potential of Treg cells in preserving microvascular health in a mouse model of orthotopic tracheal transplantation. J. Clin. Cell. Immunol. 7:89.

    Google Scholar 

  113. Khan MA, et al. (2017) FOXP3+ regulatory T cell ameliorates microvasculature in the rejection of mouse orthotopic tracheal transplants. Clin. Immunol. 174:84–98.

    Article  CAS  PubMed  Google Scholar 

  114. Facciabene A, et al. (2011) Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature. 475:226–30.

    Article  CAS  PubMed  Google Scholar 

  115. Kwon HS, et al. (2012) Three novel acetylation sites in the Foxp3 transcription factor regulate the suppressive activity of regulatory T cells. J. Immunol. 188:2712–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Kassan M, Galan M, Partyka M, Trebak M, Matrougui K. (2011) Interleukin-10 released by CD4(+)CD25(+) natural regulatory T cells improves microvascular endothelial function through inhibition of NADPH oxidase activity in hypertensive mice. Arterioscler. Thromb. Vasc. Biol. 31:2534–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Luckraz H, et al. (2004) Microvascular changes in small airways predispose to obliterative bronchiolitis after lung transplantation. J. Heart Lung Transpl. 23:527–31.

    Article  Google Scholar 

  118. Luckraz H, et al. (2006) Is obliterative bronchiolitis in lung transplantation associated with microvascular damage to small airways? Ann. Thorac. Surg. 82:1212–18.

    Article  PubMed  Google Scholar 

  119. Di Ianni M, et al. (2011) Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation. Blood. 117:3921–28.

    Article  PubMed  CAS  Google Scholar 

  120. Chatila TA. (2005) Role of regulatory T cells in human diseases. J. Allergy Clin. Immunol. 116:949–59.

    Article  CAS  PubMed  Google Scholar 

  121. Schreiber TH, et al. (2010) Therapeutic Treg expansion in mice by TNFRSF25 prevents allergic lung inflammation. J. Clin. Invest. 120:3629–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. McDonald-Hyman C, et al. (2016) Therapeutic regulatory T-cell adoptive transfer ameliorates established murine chronic GVHD in a CXCR5 dependent manner. Blood. 128:1013–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Camirand G, et al. (2014) CD45 ligation expands Tregs by promoting interactions with DCs. J. Clin. Invest. 124:4603–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Rossetti M, et al. (2015) Ex vivo-expanded but not in vitro-induced human regulatory T cells are candidates for cell therapy in autoimmune diseases thanks to stable demethylation of the FOXP3 regulatory T cell-specific demethylated region. J. Immunol. 194:113–24.

    Article  CAS  PubMed  Google Scholar 

  125. Riquelme P, Geissler EK, Hutchinson JA. (2012) Alternative approaches to myeloid suppressor cell therapy in transplantation: comparing regulatory macrophages to tolerogenic DCs and MDSCs. Transpl. Res. 1:17.

    Article  Google Scholar 

  126. Thomson AW, et al. (2016) Prospective Clinical Testing of Regulatory Dendritic Cells in Organ Transplantation. Front. Immunol. 7:15.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Dittmar L, et al. (2015) Immunosuppressive properties of mitomycin C-incubated human myeloid blood cells (MIC) in vitro. Hum. Immunol. 76:480–87.

    Article  CAS  PubMed  Google Scholar 

  128. Morath C, et al. (2015) Cell therapy for immunosuppression after kidney transplantation. Langenbecks Arch. Surg. 400:541–50.

    Article  PubMed  Google Scholar 

  129. Wolf D, et al. (2012) Tregs expanded in vivo by TNFRSF25 agonists promote cardiac allograft survival. Transplantation. 94:569–74.

    Article  CAS  PubMed  Google Scholar 

  130. Webster KE, et al. (2009) 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. 206:751–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Matta BM, Turnquist HR. (2016) Expansion of Regulatory T Cells In Vitro and In Vivo by IL-33. Methods Mol. Biol. 1371:29–41.

    Article  CAS  PubMed  Google Scholar 

  132. Biton J, et al. (2016) In Vivo Expansion of Activated Foxp3+ Regulatory T Cells and Establishment of a Type 2 Immune Response upon IL-33 Treatment Protect against Experimental Arthritis. J. Immunol. 197:1708–19.

    Article  CAS  PubMed  Google Scholar 

  133. Huntington ND, Tarlinton DM. (2004) CD45: direct and indirect government of immune regulation. Immunol. Lett. 94:167–74.

    Article  CAS  PubMed  Google Scholar 

  134. June CH, Blazar BR. (2006) Clinical application of expanded CD4+25+ cells. Sem. Immunol. 18:7888.

    Article  CAS  Google Scholar 

  135. Marek-Trzonkowska N, et al. (2012) Administration of CD4+CD25highCD127− regulatory T cells preserves beta-cell function in type 1 diabetes in children. Diabetes Care. 35:1817–20.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Brennan TV, et al. (2011) Requirements for prolongation of allograft survival with regulatory T cell infusion in lymphosufficient hosts. J. Surg. Res. 169:e69–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Lee MK, et al. (2004) Promotion of allograft survival by CD4+CD25+ regulatory T cells: evidence for in vivo inhibition of effector cell proliferation. J. Immunol. 172:6539–44.

    Article  CAS  PubMed  Google Scholar 

  138. Zhang ZX, et al. (2011) Adoptive transfer of DNT cells induces long-term cardiac allograft survival and augments recipient CD4(+)Foxp3(+) Treg cell accumulation. Transpl. Immunol. 24:119–26.

    Article  PubMed  CAS  Google Scholar 

  139. Tokita D, et al. (2008) High PD-L1/CD86 ratio on plasmacytoid dendritic cells correlates with elevated T-regulatory cells in liver transplant tolerance. Transplantation. 85:369–77.

    Article  PubMed  Google Scholar 

  140. Pons JA, et al. (2008) FoxP3 in peripheral blood is associated with operational tolerance in liver transplant patients during immunosuppression withdrawal. Transplantation. 86:1370–78.

    Article  CAS  PubMed  Google Scholar 

  141. Takasato F, et al. (2014) Prevention of allogeneic cardiac graft rejection by transfer of ex vivo expanded antigen-specific regulatory T-cells. PloS One. 9:e87722.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Hori S, Nomura T, Sakaguchi S. (2003) Control of regulatory T cell development by the transcription factor Foxp3. Science. 299:1057–61.

    Article  CAS  Google Scholar 

  143. Salama AD, Najafian N, Clarkson MR, Harmon WE, Sayegh MH. (2003) Regulatory CD25+ T cells in human kidney transplant recipients. J. Am. Soc. Nephrol. 14:1643–51.

    Article  PubMed  Google Scholar 

  144. He Q, Fan H, Li JQ, Qi HZ. (2011) Decreased circulating CD4+CD25highFoxp3+ T cells during acute rejection in liver transplant patients. Transpl. Proc. 43:1696–1700.

    Article  CAS  Google Scholar 

  145. Zouggari Y, et al. (2009) Regulatory T cells modulate postischemic neovascularization. Circulation. 120:1415–25.

    Article  PubMed  Google Scholar 

  146. Liu W, et al. (2006) CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J. Exp. Med. 203:1701–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Putnam AL, et al. (2009) Expansion of human regulatory T-cells from patients with type 1 diabetes. Diabetes. 58:652–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. van de Berg PJ, et al. (2012) Circulating lymphocyte subsets in different clinical situations after renal transplantation. Immunology. 136:198–207.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  149. Vallotton L, et al. (2011) Monitoring of CD4+ CD25highIL-7Ralphahigh activated T cells in kidney transplant recipients. Clin. J. Am. Soc. Nephrol. 6:2025–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. (2010) FOXP3+ regulatory T cells in the human immune system. Nat. Rev. Immunol. 10:490–500.

    Article  CAS  PubMed  Google Scholar 

  151. Tang Q, Bluestone JA, Kang SM. (2012) CD4(+) Foxp3(+) regulatory T cell therapy in transplantation. Mol. Cell Biol. 4:11–21.

    Article  CAS  Google Scholar 

  152. Demirkiran A, et al. (2006) Low circulating regulatory T-cell levels after acute rejection in liver transplantation. Liver Transpl. 12:277–84.

    Article  PubMed  Google Scholar 

  153. Jiang S, Lechler RI, He XS, Huang JF. (2006) Regulatory T cells and transplantation tolerance. Human Immunol. 67:765–76.

    Article  CAS  Google Scholar 

  154. Jiang S, et al. (2006) Generation and expansion of human CD4+ CD25+ regulatory T cells with indirect allospecificity: Potential reagents to promote donor-specific transplantation tolerance. Transplantation. 82:1738–43.

    Article  CAS  PubMed  Google Scholar 

  155. Jiang S, Tsang J, Lechler RI. (2006) Adoptive cell therapy using in vitro generated human CD4+ CD25+ regulatory t cells with indirect allospecificity to promote donor-specific transplantation tolerance. Transpl. Proc. 38:3199–3201.

    Article  CAS  Google Scholar 

  156. Taylor KN, Shinde-Patil VR, Cohick E, Colson YL. (2007) Induction of FoxP3+CD4+25+ regulatory T cells following hemopoietic stem cell transplantation: role of bone marrow-derived facilitating cells. J. Immunol. 179:2153–62.

    Article  CAS  PubMed  Google Scholar 

  157. Steiner D, et al. (2006) Overcoming T cell-mediated rejection of bone marrow allografts by T-regulatory cells: synergism with veto cells and rapamycin. Exper. Hematol. 34:802–08.

    Article  CAS  Google Scholar 

  158. Colonna L, Sega EI, Negrin RS. (2011) Natural and expanded CD4(+)CD25(+) regulatory T cells in bone marrow transplantation. Biol. Blood Marrow Transplant. 17:S58–62.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Gallimore A, Godkin A. (2008) Regulatory T cells and tumour immunity: observations in mice and men. Immunology. 123:157–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Benson A, et al. (2012) Microbial infection-induced expansion of effector T cells overcomes the suppressive effects of regulatory T cells via an IL-2 deprivation mechanism. J. Immunol. 188:800–10.

    Article  CAS  PubMed  Google Scholar 

  161. Leavy O. (2012) Tolerance: Induced T(Reg) cells evolved to protect the fetus. Nat. Rev. Immunol. 12:554–55.

    Article  CAS  PubMed  Google Scholar 

  162. Johansson M, Denardo DG, Coussens LM. (2008) Polarized immune responses differentially regulate cancer development. Immunol. Rev. 222:145–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Wing K, Sakaguchi S. (2010) Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat. Immunol. 11:7–13.

    Article  CAS  PubMed  Google Scholar 

  164. Bettini ML, Vignali DA. (2010) Development of thymically derived natural regulatory T cells. Ann. NY Acad. Sci. 1183:1–12.

    Article  CAS  PubMed  Google Scholar 

  165. Sun CM, et al. (2007) Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J. Exp. Med. 204:1775–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Beres A, Komorowski R, Mihara M, Drobyski WR. (2011) Instability of Foxp3 expression limits the ability of induced regulatory T cells to mitigate graft versus host disease. Clin. Cancer Res. 17:3969–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Zhou L, Chong MM, Littman DR. (2009) Plasticity of CD4+ T cell lineage differentiation. Immunity. 30:646–55.

    Article  CAS  PubMed  Google Scholar 

  168. Yang XO, et al. (2008) Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity. 29:44–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Wei G, et al. (2009) Global mapping of H3K4me3 and H3K27me3 reveals specificity and plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity. 30:155–67.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  170. Huehn J, Polansky JK, Hamann A. (2009) Epigenetic control of FOXP3 expression: the key to a stable regulatory T-cell lineage? Nature reviews. Immunology. 9:83–89.

    Article  CAS  PubMed  Google Scholar 

  171. Floess S, et al. (2007) Epigenetic control of the foxp3 locus in regulatory T cells. PLoS Biol. 5:e38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  172. Kim HP, Leonard WJ. (2007) CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNA methylation. J. Exp. Med. 204:1543–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Baron U, et al. (2007) DNA demethylation in the human FOXP3 locus discriminates regulatory T cells from activated FOXP3(+) conventional T cells. Eur. J. Immunol. 37:2378–89.

    Article  CAS  PubMed  Google Scholar 

  174. Starr TK, Jameson SC, Hogquist KA. (2003) Positive and negative selection of T cells. Annu. Rev. Immunol. 21:139–76.

    Article  CAS  PubMed  Google Scholar 

  175. Pazetti R, Pego-Fernandes PM, Jatene FB. (2013) Adverse effects of immunosuppressant drugs upon airway epithelial cell and mucociliary clearance: implications for lung transplant recipients. Drugs. 73:1157–69.

    Article  CAS  PubMed  Google Scholar 

  176. Tang Q, Bluestone JA. (2013) Regulatory T-cell therapy in transplantation: moving to the clinic. Cold Spring Harb. Perspect. Med. 3.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Mohammad Afzal Khan.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, and provide a link to the Creative Commons license. You do not have permission under this license to share adapted material derived from this article or parts of it.

The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this license, visit (http://creativecommons.org/licenses/by-nc-nd/4.0/)

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Khan, M.A. T regulatory Cell-mediated Immunotherapy for Solid Organ Transplantation: A Clinical Perspective. Mol Med 22, 892–904 (2016). https://doi.org/10.2119/molmed.2016.00050

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2119/molmed.2016.00050

Keywords