Skip to main content
The Yale Journal of Biology and Medicine logoLink to The Yale Journal of Biology and Medicine
. 2011 Dec;84(4):345–351.

Learning to Live Together: Harnessing Regulatory T cells to Induce Organ Transplant Tolerance

Andrew Y Chang 1,*, Nupur Bhattacharya 1
PMCID: PMC3238321  PMID: 22180672

Abstract

The discovery of immune cells with regulatory effects has created considerable excitement for their potential use in inducing tolerance to transplanted tissues. Despite the fact that these cells possess essential functions in vivo, attempts to translate them into effective clinical therapies has proved challenging due to a number of unanticipated complexities in their behavior. This article provides a broad summary of research done to understand the largest of the regulatory cell subtypes, namely CD4+Foxp3+ Regulatory T cells (TRegs). Special attention will be paid to current and future difficulties in using TRegs clinically, as well as room for improvement and innovation in this field.

Keywords: TRegs, Regulatory T-cells, Transplant Tolerance, Transplant Immunology

Introduction

Though the concept that cells with the ability to downregulate the immune response has been around for a considerable time, their existence was strongly debated until the early 1990s, when a series of studies by Sakaguchi et al. described a population of CD4+ T cells expressing the IL-2 receptor CD25. When athymic mice were inoculated with T cell transfers depleted of these CD25+ cells, they developed severe autoimmunity in multiple organ systems [1]. Replacement of CD4+CD25+ cells suppressed the disease [1]. These “regulatory” T cells were subsequently suggested to be decreased in human patients with autoimmune diseases such as multiple sclerosis [2].

Researchers immediately recognized the therapeutic potential of these cells, not only in autoimmune disease, where their numbers were presumably decreased (and tolerance thus broken), but also in certain infections and tumors. The peripheral blood of epithelial cancer patients has elevated circulating regulatory T cells, and numerous mouse models have shown that manipulation of this cell population can increase or decrease immune-mediated tumor rejection [3,4]. Their tolerogenic effect also has been hypothesized to underlie the persistence of certain viral infections such as hepatitis C [5].

Particular interest in their ability to determine patient tolerance to non-self antigens was augmented by the discovery that antigen-specific CD4+ regulatory T cells were increased in mice, which tolerated allografted tissues long-term [6]. A number of human studies have since shown that a high number of circulating TRegs in kidney and liver transplant patients is correlated with the stability of graft acceptance [7,8,9]. As such, considerable excitement about the clinical usage of TRegs in organ transplantation has been drawn up in the past decade.

Nevertheless, a number of difficulties have arisen concerning the translation of these observational studies into useful human therapies; the system is much more complex than was initially expected. For example, the heterogeneity, plasticity, and context-dependent activity of TRegs have all stood in the way of developing an effective, yet safe, treatment option for transplant patients. In this review, we summarize the biology of CD4+ Foxp3+ TRegs and then discuss a framework for creating appropriate therapies in relation to the challenges presented. New approaches to apply these concepts in medicine also will be highlighted.

Current Landscape on Combating Transplant Rejection

The transplantation of donor tissues has been the dramatic last resort for intractable end-organ failure in a host of human diseases. Since the 1920s, however, physicians have observed rejection of foreign grafts, no doubt mediated by the immune system’s recognition of non-self protein targets [10]. Though a full discussion of the myriad mechanisms by which this process occurs is beyond the scope of this review, the major pathways are due to effector lymphocyte priming against donor HLA antigens, leading to cytotoxic effects (both direct cell-cell or humoral) on the parenchyma or vasculature of the graft [10]. The end result is progressive organ failure.

Before the discovery of regulatory immune cells and their role in promoting tolerance, the goal of creating durable organ transplant survival was focused on the elimination of the effector cells hostile to transplanted antigens. This concept is exemplified in the current repertoire of anti-rejection pharmaceuticals in clinical use, such as cyclosporine, a calcineurin inhibitor. Unfortunately, these therapies have a number of flaws preventing them from becoming acceptable permanent solutions to safe, long-term organ transplant acceptance. First, the drugs are nonspecific, generally dampening the immune system and leading to dangerous immunosuppressive side effects. Second, they are only a short-term solution; patients rarely achieve permanent tolerance and are dependent on these drugs ― and their side effects ― for life.

The recognition of TRegs has not only significantly altered the extant paradigm, but proposed an additional reason why long-term tolerance cannot be achieved with our current treatment modalities: the fact that these drugs do not distinguish between effector and regulatory players means that though the anti-graft response is prevented, so is the pro-graft tolerance response [11]. The ideal transplant induction therapy is thus one that Spoerl and Li define in their 2011 review as stable, self-perpetuating, and donor antigen-specific — factors that do not describe our treatments at the moment [12]. Taken together, these observations strongly suggest that more research must be conducted in order to understand how to heighten the activity of TRegs in transplant patients, either adoptively or endogenously.

Regulatory T cells

Much work has been carried out since the first studies examining broad CD4+ populations to characterize cells with regulatory properties. One of the first observations has been that there are numerous groups and subgroups of cells (both found in vivo and experimentally induced) with suppressive phenotypes of various potency including CD4+CD25+Foxp3+, CD8+Foxp3+, Tr1 cells, Tr35 cells, CD3+CD4-CD8- “Double-Negative” cells, and NKT cells [13,14,15]. For the purposes of this review, the term TReg will be defined as CD4+Foxp3+ cells, since they are the most numerous, naturally occurring, and well-studied of these various cell types.

The emphasis on CD4+Foxp3+ cells was heightened by the discovery of the Foxp3 (forkhead box P3) transcription factor, which regulates the initiation and maintenance of suppressive properties in TRegs. The importance of Foxp3 was demonstrated dramatically in scurfy mice, which are Foxp3-deficient. These animals have a lack of functional TRegs and suffer severe autoimmune effects in multiple organs [16]. A related condition in humans known as IPEX (immunodysregulation, polyendocrinopathy, and enteropathy, X-linked) is also associated with a mutated Foxp3 gene [17].

CD4+Foxp3+ TRegs are divided into two subgroups: “natural” TRegs (nTReg) and “induced” TRegs (iTReg). nTRegs are born in the thymus and are selected there by their specificities to self autoantigens, analogous to the process used to select effector T cells in the same organ [18]. iTRegs, on the other hand, come from existing CD4+Foxp3- T cells in the periphery that have been converted to tolerate, rather than to reject, their target antigen [19]. Their surprising origin implies not only that foreign antigens can become tolerated, but tolerance itself is a fluid, non-static process that is heavily context-dependent. In fact, these non-regulatory T cells are converted to iTRegs by a number of different factors, including TGF-β, IL-2, retinoic acid and leukemia inhibitory factor (LIF) [20,21,22]. Furthermore, the fact that both types of TRegs undergo a specificity-mediated selection process means that they are highly specific for individual antigens through engagements with their T cell Receptors (TCRs).

As for how TRegs suppress their target cells, the mechanisms are likely multiple and, as yet, not fully understood. For instance, it is known that within in vitro model systems, TRegs influence a whole host of immune subtypes, including CD4+ T cells, CD8+ T cells, natural killer T (NKT) cells, and B cells [23]. Their effect is not limited simply to effector cells, however, antigen-presenting cells such as dendritic cells and macrophages are also under TReg purview, as are osteoblasts, mast cells, and natural killer (NK) cells [23]. Their molecular toolbox for achieving their actions is thus similarly diverse, employing secreted suppressor cytokines (e.g., IL-10, TGF-ß, IL-35), consumption of local activating cytokines (e.g., IL-2), cell-surface molecule signaling (e.g., Galectin-1), and direct cell-cell killing (via the granzyme complex) [23]. Recent findings also show that TRegs are capable of altering cell surfaces by trans-endocytosing CD86 and CD80 co-stimulatory ligands on target antigen-presenting cells [24]. This is achieved by TReg CTLA-4, which recognizes those molecules and causes them to be internalized and digested by the TReg. Another feature of particular interest to transplant physicians in TRegs is that they also exhibit anti-inflammatory and anti-tissue remodeling effects, including the inhibition of transplant vasculopathy, a condition that accelerates the rejection of a donor organ [25].

Current Use of TReg Therapy in Transplant Models

The potent aforementioned properties of TRegs have attracted researchers to begin animal and preliminary clinical tests to bring a therapy closer to reality. A number of murine studies have shown that TRegs can be generated in different ways and confer allograft tolerance [25,25,27,28]. Regulatory T cells induced in vivo, in vitro, or expanded ex vivo produced some impressive results, including beating heart graft survivals past a 100-day observation period [25]. Most of these studies, however, have had several key limitations. First, the animal subjects had their existing immune systems downregulated in some fashion, either by sublethal irradiation or the depletion of lymphocytes or CD4 T cells with monoclonal antibody pretreatment. Second, although most of these studies were able to stave off acute transplant rejection, chronic rejection still occurred, or in the absence of frank chronic rejection, histological evidence of inflammatory infiltration in the graft was still noted.

Nevertheless, the possibility of using immunoablation followed by bone marrow transplantation (BMT) as a means of “resetting” the immune system and transferring tolerance to solid organ transplants has led to several small human trials with promising results [29,30,31]. Co-transplantation of both bone marrow and kidneys into patients showed not only cases of long-term graft acceptance, but the complete discontinuation of immunosuppressive therapies for some. As for the association between BMT and TRegs, the Kawai et al. study also detected that Foxp3 mRNA levels in renal biopsies of stable immunosuppression-free patients were about 6 times higher than those from the stable-with-immunosuppression group [31]. Thus, even in the absence of more specific immune-tailoring, fostering the growth of endogenous TRegs or possibly transferring ex vivo TRegs could be beneficial to human patients as well.

Methods for Harvesting and Expanding TRegs

Creating a feasible, more specific TReg therapy for human transplant tolerance is primarily limited, however, by the difficulty of expanding TReg populations to sizes large enough to tip the effector-regulatory balance. They are not particularly numerous; CD4+25+ regulatory cells constitute only 5 to 10 percent of peripheral CD4+ T cells [32]. To this end, a number of experimental strategies are being investigated for ways to grow TRegs, both in vitro and in vivo. The first major approach involves identifying samples highly purified for naturally occurring TRegs. Since Foxp3 is an intracellular molecule, it is not available as a cell surface marker. As such, a cocktail of antibodies (to CD25, CD45RA, CD27, CD39, CD49b, FR4, or PD-1), with magnetic microbeads and columns, must be used to select them [33]. Thereafter, they can be expanded ex vivo using donor or recipient antigen-presenting cells (APCs) or anti-CD3/CD28 coated beads [33]. The resultant TRegs can be reintroduced into the patient. The second method involves the conversion of isolated effector CD4+ T cells into induced TRegs. This can be achieved by exposing them to a whole host of cytokines, costimulatory factors, and growth factors, including TGF-ß, IL-2, LIF, PD-1/PD-L1, retinoic acid, and IDO [20,21,22,34,35,36]. As with the prior strategy, these converted ex vivo TRegs can then be administered to the patient.

The third major approach would be to expand TRegs in vivo with a variety of growth signals, a process that is made more attractive because it avoids the difficulties of purifying TRegs and then coaxing them to expand in vitro. However, though this method seems as simple as introducing growth factors to the patient, the signals are not specific for just TRegs and may result in systemic side effects. A possible workaround is to somehow preferentially encourage the growth of regulatory T cells over effector T cells; indeed, inhibiting effector T cell costimulatory pathways such as CD28, CD40, OX40, ICOS, and CD27 with monoclonal antibodies has been able to induce tolerance to solid organ transplants in murine experimental systems [37].

Additional Challenges and Opportunities

In addition to the issue of efficiently generating a large number of TRegs for therapy, several factors inherent to their physiology make it challenging to envision regulatory T cells in clinic in the near future. For one, the difference between natural n TRegs and induced iTRegs described above could have significant ramifications in approaches that exclusively use one subset of TRegs or the other. nTRegs, because they are tailored to recognize and accept self antigens, may cause immunodeficiencies if expanded beyond their normal repertoire of tolerance. iTRegs avoid this problem but suffer from another unique property: They are far more unstable than their nTRegs brethren. iTRegs are much more likely, under various stresses, to lose Foxp3 expression, a process probably due to epigenetic differences [38]. In fact, iTRegs have been noted to turn into anti-tolerant TH17 T cells when exposed to inflammatory cytokines, the direct opposite of properties desired in transplant therapy [39].

Furthermore, one must not forget that TReg activity is the suppression of an existing active process; much of their effectiveness is due to the fact that they can systematically shut down effector cells. As such, effector cells can find ways to evade TRegs-mediated suppression. One example is memory T cells, which maintain a certain resistance to the activities of regulatory T cells [40]. NK cells, on the other hand, take a more direct approach by lysing TRegs [41]. As such, therapies that seek to use TRegs for tolerizing patients to transplanted organs also will need to overcome the anti-suppressive impulses of other immune cells. For example, Afzali et al. suggest that the resistance of memory T cells to downregulation can be counteracted by infusing TRegs prior to transplantation, thus preempting the development of these resilient cells [40]. NK cells could (cautiously) be targeted for depletion with monoclonal antibodies.

Even if TRegs are able to be easily expanded and the anti-regulatory response sufficiently reduced, more theoretical challenges still exist. First, there is the risk of uncontrolled adoptive TReg proliferation; inadvertent suppression of the normal immune response may cause unregulated growth of infectious agents and tumor cells. A possible way around this problem is the engineering of self-limiting or self-destructing TRegs that stop growing after the therapeutic goal is achieved. It is yet an unexplored field, but a cell-surface receptor sensitive to the tolerance-immunocompromise balance (perhaps via circulating cytokine detection) could be coupled to the apoptotic pathway of a TReg to maintain an appropriate population size. Another unresolved issue is that of crosstalk between the numerous regulatory T cell subsets named above (CD8+Foxp3+, Tr1 cells, Tr35 cells, CD3+CD4-CD8- “Double-Negative” cells, NKT cells). Immunosuppression is a sophisticated tightrope to walk, and it is highly unlikely that the different suppressor cell types do not communicate with each other to decide this important concern. As of yet, there is little experimental investigation into this subject.

Lastly, there is the issue of TRegs and the current practice of medicine. As was discussed before, the extant standards of treatment involve the use of broad immunosuppressive pharmaceuticals. Calcineurin inhibitors like cyclosporine suppress TCR signaling, blocking the conversion of effector T cells into induced TRegs [42]. All is not lost, however, as other drug classes such as mTOR inhibitors (exemplified by rapamycin/sirolimus) have shown surprisingly positive effects on the development of tolerance [42]. A mouse model utilizing rapamycin, costimulatory blockade, and BMT showed that the therapy could induce mixed chimerism and subsequent graft tolerance without the need for dangerous immunoablative therapies [43]. On the human side, renal transplant patients receiving low-dose rapamycin had increased circulating TRegs, suggesting that future immunosuppressive drug regimens should take into account those that are more TReg-friendly [44].

Conclusions and Outlook

It is undeniable that regulatory T cells are a powerful and important cellular actor in the establishment of tolerance in the human body. Nevertheless, major strides need to be made in overcoming both technical and mechanistic challenges to turn the existing research into a coherent and specific therapy for transplant patients. Particular attention needs to be paid to expanding TRegs efficiently, controlling their fickle suppressive-inflammatory duality, and overcoming endogenous resistance to regulatory action. If these goals are achieved, a novel and considerable force will have been recruited not only for the treatment of transplant patients, but the understanding and future conquest of autoimmune diseases, cancer, and infection.

Glossary

BMT

Bone Marrow Transplantation

CD

Cluster of Differentiation

CTLA

Cytotoxic T-Lymphocyte Antigen

Foxp3

Forkhead Box P3

HLA

Human Leukocyte Antigen

IDO

Indolamine 2,3-dioxygenase

IL

Interleukin

IPEX

Immunodysregulation, Polyendocrinopathy, and Enteropathy, X-linked

LIF

Leukemia Inhibitory Factor

NK

Natural Killer

TCR

T cell Receptor

TGF

Transforming Growth Factor

TReg

Regulatory T cell

References

  1. 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(3):1151–1164. [PubMed] [Google Scholar]
  2. Viglietta V, Baecher-Allan C, Weiner HL, Hafler DA. Loss of Functional Suppression by CD4+CD25+ Regulatory T Cells in Patients with Multiple Sclerosis. J Exp Med. 2004;199(7):971–979. doi: 10.1084/jem.20031579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Wolf AM, Wolf D, Steurer M, Gastl G, Gunsilius E, Grubeck-Loebenstein B. Increase of Regulatory T Cells in the Peripheral Blood of Cancer Patients. Clin Cancer Res. 2003;9(2):606–612. [PubMed] [Google Scholar]
  4. Zou W. Regulatory T cells, tumour immunity and immunotherapy. Nat Rev Immunol. 2006;6(4):295–307. doi: 10.1038/nri1806. [DOI] [PubMed] [Google Scholar]
  5. Cabrera R, Tu Z, Xu Y, Firpi RJ, Rosen HR, Liu C. et al. An immunomodulatory role for CD4+CD25+ regulatory T lymphocytes in hepatitis C virus infection. Hepatology. 2004;40(5):1062–1071. doi: 10.1002/hep.20454. [DOI] [PubMed] [Google Scholar]
  6. Cobbold S, Waldmann H. Infectious tolerance. Curr Opin Immunol. 1998;10(5):518–524. doi: 10.1016/s0952-7915(98)80217-3. [DOI] [PubMed] [Google Scholar]
  7. Salama AD, Najafian N, Clarkson MR, Harmon WE, Sayegh MH. Regulatory CD25+ T Cells in Human Kidney Transplant Recipients. J Am Soc Nephrol. 2003;14(6):1643–1651. doi: 10.1097/01.asn.0000057540.98231.c1. [DOI] [PubMed] [Google Scholar]
  8. Martínez-Llordella M, Puig-Pey I, Orlando G, Ramoni M, Tisone G, Rimola A. et al. Multiparameter Immune Profiling of Operational Tolerance in Liver Transplantation. Am J Transplant. 2007;7(2):309–319. doi: 10.1111/j.1600-6143.2006.01621.x. [DOI] [PubMed] [Google Scholar]
  9. Bestard O, Cruzado JM, Mestre M, Caldés A, Bas J, Carrera M. et al. Achieving Donor-Specific Hyporesponsiveness Is Associated with FOXP3+ Regulatory T Cell Recruitment in Human Renal Allograft Infiltrates. J Immunol. 2007;179(7):4901–4909. doi: 10.4049/jimmunol.179.7.4901. [DOI] [PubMed] [Google Scholar]
  10. Sayegh MH, Carpenter CB. Transplantation 50 years later―progress, challenges, and promises. N Engl J Med. 2004;351(26):2761–2766. doi: 10.1056/NEJMon043418. [DOI] [PubMed] [Google Scholar]
  11. Lechler RI, Garden OA, Turka LA. The complementary roles of deletion and regulation in transplantation tolerance. Nat Rev Immunol. 2003;3(2):147–158. doi: 10.1038/nri1002. [DOI] [PubMed] [Google Scholar]
  12. Spoerl S, Li XC. Regulatory T cells and the quest for transplant tolerance. Discov Med. 2011;11(56):25–34. [PubMed] [Google Scholar]
  13. 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(6652):737–742. doi: 10.1038/39614. [DOI] [PubMed] [Google Scholar]
  14. Zhang Z-X, Yang L, Young KJ, DuTemple B, Zhang L. Identification of a previously unknown antigen-specific regulatory T cell and its mechanism of suppression. Nat Med. 2000;6(7):782–789. doi: 10.1038/77513. [DOI] [PubMed] [Google Scholar]
  15. Seino K, Fukao K, Muramoto K, Yanagisawa K, Takada Y, Kakuta S. et al. Requirement for natural killer T (NKT) cells in the induction of allograft tolerance. Proc Nat Acad Sci USA. 2001;98(5):2577–2581. doi: 10.1073/pnas.041608298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Walker MR, Kasprowicz DJ, Gersuk VH, Bènard 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(9):1437–1443. doi: 10.1172/JCI19441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bacchetta R, Passerini L, Gambineri E, Dai M, Allan SE, Perroni L. et al. Defective regulatory and effector T cell functions in patients with FOXP3 mutations. J Clin Invest. 2006;116(6):1713–1722. doi: 10.1172/JCI25112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Fontenot JD, Rasmussen JP, Williams LM, Dooley JL, Farr AG, Rudensky AY. Regulatory T Cell Lineage Specification by the Forkhead Transcription Factor Foxp3. Immunity. 2005;22(3):329–341. doi: 10.1016/j.immuni.2005.01.016. [DOI] [PubMed] [Google Scholar]
  19. Curotto de Lafaille MA, Lafaille JJ. Natural and Adaptive Foxp3+ Regulatory T Cells: More of the Same or a Division of Labor? Immunity. 2009;30(5):626–635. doi: 10.1016/j.immuni.2009.05.002. [DOI] [PubMed] [Google Scholar]
  20. Chen W, Jin W, Hardegen N, Lei K, Li L, Marinos N. et al. Conversion of Peripheral CD4+CD25− Naive T Cells to CD4+CD25+ Regulatory T Cells by TGF-β Induction of Transcription Factor Foxp3. J Exp Med. 2003;198(12):1875–1886. doi: 10.1084/jem.20030152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Benson MJ, Pino-Lagos K, Rosemblatt M, Noelle RJ. All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation. J Exp Med. 2007;204(8):1765–1774. doi: 10.1084/jem.20070719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. 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(9):1444–1450. doi: 10.4161/cc.8.9.8348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Shevach EM. Mechanisms of Foxp3+ T Regulatory Cell-Mediated Suppression. Immunity. 2009;30(5):636–645. doi: 10.1016/j.immuni.2009.04.010. [DOI] [PubMed] [Google Scholar]
  24. Qureshi OS, Zheng Y, Nakamura K, Attridge K, Manzotti C, Schmidt EM. et al. Trans-Endocytosis of CD80 and CD86: A Molecular Basis for the Cell-Extrinsic Function of CTLA-4. Science. 2011;332(6029):600–603. doi: 10.1126/science.1202947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Joffre O, Santolaria T, Calise D, Saati TA, Hudrisier D, Romagnoli P. et al. Prevention of acute and chronic allograft rejection with CD4+CD25+Foxp3+ regulatory T lymphocytes. Nat Med. 2008;14(1):88–92. doi: 10.1038/nm1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Bushell A, Morris PJ, Wood KJ. Transplantation tolerance induced by antigen pretreatment and depleting anti‐CD4 antibody depends on CD4+ T cell regulation during the induction phase of the response. Eur J Immunol. 1995;25(9):2643–2649. doi: 10.1002/eji.1830250936. [DOI] [PubMed] [Google Scholar]
  27. 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(3):1080–1086. doi: 10.4049/jimmunol.168.3.1080. [DOI] [PubMed] [Google Scholar]
  28. Xia G, He J, Leventhal JR. Ex Vivo‐Expanded Natural CD4+CD25+ Regulatory T Cells Synergize With Host T‐Cell Depletion to Promote Long‐Term Survival of Allografts. Am J Transplant. 2008;8(2):298–306. doi: 10.1111/j.1600-6143.2007.02088.x. [DOI] [PubMed] [Google Scholar]
  29. Bühler 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(10):1405–1409. doi: 10.1097/00007890-200211270-00011. [DOI] [PubMed] [Google Scholar]
  30. 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(4):362–368. doi: 10.1056/NEJMoa074191. [DOI] [PubMed] [Google Scholar]
  31. 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(4):353–361. doi: 10.1056/NEJMoa071074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Ann Rev Immunol. 2004;22:531–562. doi: 10.1146/annurev.immunol.21.120601.141122. [DOI] [PubMed] [Google Scholar]
  33. Muller YD, Seebach JD, Bühler LH, Pascual M, Golshayan D. Transplantation tolerance: Clinical potential of regulatory T cells. Self Nonself. 2011;2(1):26–34. doi: 10.4161/self.2.1.15422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Zheng SG, Wang J, Wang P, Gray JD, Horwitz DA. IL-2 Is Essential for TGF-β to Convert Naive CD4+CD25- Cells to CD25+Foxp3+ Regulatory T Cells and for Expansion of These Cells. J Immunol. 2007;178(4):2018–2027. doi: 10.4049/jimmunol.178.4.2018. [DOI] [PubMed] [Google Scholar]
  35. 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(13):3015–3029. doi: 10.1084/jem.20090847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. 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(3):555–563. doi: 10.1182/blood-2008-11-191197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Wekerle T, Kurtz J, Bigenzahn S, Takeuchi Y, Sykes M. Mechanisms of transplant tolerance induction using costimulatory blockade. Curr Opin Immunol. 2002;14(5):592–600. doi: 10.1016/s0952-7915(02)00378-3. [DOI] [PubMed] [Google Scholar]
  38. Lal G, Bromberg JS. Epigenetic mechanisms of regulation of Foxp3 expression. Blood. 2009;114(18):3727–3735. doi: 10.1182/blood-2009-05-219584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGF[beta] in the Context of an Inflammatory Cytokine Milieu Supports De Novo Differentiation of IL-17-Producing T Cells. Immunity. 2006;24(2):179–189. doi: 10.1016/j.immuni.2006.01.001. [DOI] [PubMed] [Google Scholar]
  40. Afzali B, Mitchell PJ, Scottà C, Canavan J, Edozie FC, Fazekasova H. et al. Relative Resistance of Human CD4+ Memory T Cells to Suppression by CD4+CD25+ Regulatory T Cells. Am J Transplant. 2011;11(8):1734–1742. doi: 10.1111/j.1600-6143.2011.03635.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Roy S, Barnes PF, Garg A, Wu S, Cosman D, Vankayalapati R. NK Cells Lyse T Regulatory Cells That Expand in Response to an Intracellular Pathogen. J Immunol. 2008;180(3):1729–1736. doi: 10.4049/jimmunol.180.3.1729. [DOI] [PubMed] [Google Scholar]
  42. Gao W, Lu Y, El Essawy B, Oukka M, Kuchroo VK, Strom TB. Contrasting Effects of Cyclosporine and Rapamycin in De Novo Generation of Alloantigen‐Specific Regulatory T Cells. Am J Transplant. 2007;7(7):1722–1732. doi: 10.1111/j.1600-6143.2007.01842.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. 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(4):751–762. doi: 10.1111/j.1600-6143.2010.03018.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. 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(3):1007–1018. doi: 10.1681/ASN.2006101143. [DOI] [PubMed] [Google Scholar]

Articles from The Yale Journal of Biology and Medicine are provided here courtesy of Yale Journal of Biology and Medicine

RESOURCES