Abstract
Purpose of review
Achieving allograft tolerance is the holy grail of transplantation. However, tolerance and rejection are two extreme ends of a scale that can be tipped in either direction. We review the novel effector and regulatory mechanisms involved and factors that tip the balance in favor of rejection or regulation.
Recent findings
It is increasingly recognized that established T cell phenotypes could change their commitments. New data point to the plasticity of Th17 cells in vivo with a reciprocal balance of Th17 cells and regulatory T cells driven by the local cytokine environment. Regulatory T cell profiles have been linked to acute and chronic allograft outcomes, and emerging data also indicate a novel role of a regulatory B cell population. Current research efforts are looking into factors that tip the balance towards allograft tolerance by targeting cytokines, novel co-stimulatory pathways such as T cell Ig mucin molecules, and components of innate immunity, particularly dendritic cells.
Summary
The balance of effector and regulatory mechanisms contributing to allograft outcome is very complex. It is likely that targeting multiple pathways will be required to achieve tolerance. Further studies are warranted to define this balance and identify optimal combination of therapeutic interventions.
Keywords: Allograft rejection, allograft tolerance, transplantation, regulatory cells
Introduction
Transplantation tolerance is defined as the absence of pathologic immune response against a graft in an immunocompetent host not using maintenance immunosuppressive drugs while still maintaining protective responses against third party antigens (1). Significant research contributions have been made to the field since the first description of acquired tolerance to allograft antigens was published in a landmark nature article by Billingham et al in 1953 (2). It is well recognized now that the fate of the allograft depends on the balance of effector and regulatory immune mechanisms, with allograft tolerance and rejection being two opposite ends of the scale. While most research in the past decade have focused on the balance of effector memory and regulatory T cells and how they are affected by co-stimulation, there is increasing effort to understand the contribution of newly discovered populations such as T helper 17 and Regulatory B cells. There is also a renewed interest in defining the role of innate immunity in tipping the balance towards allograft rejection through inflammatory monocytes or tolerance through tolerogenic dendritic cells. The purpose of this review is to summarize recently discovered novel effector and regulatory mechanisms and the various over-riding pathways that affect their balance and determine the fate of the allograft.
A. Effector Mechanisms
The role of monocytes in alloimmunity, and memory T cells and their exhaustive differentiation is discussed in detail by Mannon et al. and Li et al. in the accompanying articles. We provide below a brief overview of other new emerging players in rejection and tolerance.
1. T Helper 17 (Th17) Cells
T helper 1 (Th1) cells have long been considered the classic effector T cell phenotype implicated in allograft rejection (3). However, the development of cardiac allograft rejection in a mouse model lacking the Th1 lineage led to the implication of Th17 cells as a novel T cell subset characterized by the production of Interleukin-17 (IL-17) in allograft rejection (4). Increasingly, IL-17 secreting cells are being identified in human allograft recipients as we continue to understand their significance and contribution to allograft outcomes (5–9). Recently, Hester et al have identified an increase of IL-17A producing cells in kidney transplant recipients treated with sirolimus monotherapy compared to sirolimus and mycophenolate mofetil following alemtuzumab induction. This finding would be concerning except for the fact that it was compensated for by an increased functionally suppressive Treg population in the same subjects (6). It is important to note that Th17 cells likely develop from the same precursors as induced regulatory T cells (iTregs) depending on the cytokine environment. The presence of Interleukin-6 (IL-6) has been shown to inhibit Treg generation and induce Th17 differentiation (4), which underscores the significant influence of innate immunity in tipping the balance towards rejection in the setting of inflammation. Th17 cells also exhibit considerable plasticity in vitro and in vivo (10). Depending on the cytokine milieu, Th17 cells can pursue various fates: in inflammatory settings, Th17 cells may remain stable, but in some immune diseases, may convert to Th1-like cells producing IFN-γ, shown to be the main pathogenic force in EAE, while, in contrast, Th17 cells can completely cease IL-17 production in anti-inflammatory conditions, both “ex-Th17” cells “invisible” to methods of detection based on cytokine expression. The role of Th17 plasticity, if any, is currently unknown in alloimmunity. In sum, our understanding of the mechanisms underlying both rejection and tolerance has evolved with the discovery of novel T cell subsets such as Tregs and Th17 cells and the concept of plasticity. Th17 cells are reviewed in more detail by Ansari et al. later in this issue.
2. T Helper 9 (Th9) Cells
A unique subset of CD4+ T helper cells secreting interleukin-9 (IL-9) has recently been identified and implicated in allergic and autoimmune disease models (11). Wong et al have shown that TGF-β and IL-4 drive the generation of this subset from human CD4+ T cells (12). This underscores the importance of the local cytokine environment in shaping the immune response given that TGF-β is also involved in the differentiation of both Th17 and Tregs in the presence of other cytokines. The role of this subset in transplantation and allograft rejection remains unclear. Interestingly, Poulin et al observed massive eosinophilic infiltration in rejected MHC class II mismatched mouse cardiac allografts in donor grafts from IL-9 transgenic mice compared to wild type animals, suggesting that overexpression of IL-9 within the graft may influence transplant outcomes (13). We have unpublished preliminary data indicating that blockade of IL-9 signaling abrogates the tolerogenic effects of CTLA4-Ig (Abstract by Boenisch et al, ATC 2011). These data in aggregate highlight a new regulatory function of IL-9 in solid organ transplantation.
3. T Follicular Helper (Tfh) Cells
Tfh cells provide a link between the cellular and humoral arms of the adaptive immune systems. Those cells can be found in the germinal centers of lymphoid tissue and instruct B cells to generate affinity-matured antibodies through their production of IL-21 (14). Recently, Choi et al have shown that activation of naïve CD4+ cells by dendritic cells via ICOS signals upregulated Bcl-6 expression; the characteristic marker of the Tfh subset. Maintenance of this population was dependent on the presence of B cells also via ICOS signals (15). Targeting these interactions and/or modulating the expression of Tfh specific markers can provide new means for suppressing anti-donor humoral responses following transplantation.
B. Regulatory Mechanisms
Regulatory T cells are the classic population linked to allograft tolerance but increasing emphasis is currently directed towards defining a regulatory B cell population.
1. Regulatory T Cells (Tregs)
In the context of alloimmunity, the balance between effector and regulatory cells dictates whether the outcome of a transplant is rejection or tolerance. New data indicate that Tregs can exhibit considerable plasticity (16); unstable Tregs can play an effector rather than regulatory role, depending on the inflammatory conditions (17). These new insights have led to a re-evaluation of transplantation biology and form the basis for the development of novel therapeutic approaches for preventing rejection.
Debate continues regarding the use of Tregs as biomarkers for tolerance in transplantation. Recent human data from renal transplant recipient biopsies reported by Kollins et al failed to correlate the level of intragraft FoxP3+ cells with the severity of graft rejection or renal function at 1 or 2 years (18). Batsford et al had similar findings when they showed that even though FoxP3+ Treg cells within the infiltrating T-cell population can increase transiently during episodes of acute rejection, the phenomenon was not consistently seen in acute cellular rejection and the information did not correlate with serum creatinine at the time of biopsy, at 3 months or 1 year later (19). On the contrary, when Iwase et al utilized real time PCR to detect peripheral blood FoxP3 mRNA in renal transplant recipients up to 1-year post transplantation they noted significantly lower levels of FoxP3 in those with chronic rejection compared to patients with stable graft function (20). Similarly Bhorade et al found that a threshold of 3.2% CD4+FoxP3+ cells in the broncho-alveolar lavage of lung transplant recipients distinguished stable recipients from those subsequently developing Bronchiolitis Obliterans within the first 2 years post transplantation (21). These observations highlight the complexities of using Tregs as a biomarker for acute rejection in human allograft recipients but show promising results in correlating FoxP3+ Treg levels to chronic allograft outcomes. This correlation may differ between different organ types and sampling sources used to detect Tregs (22, 23)
Recent research efforts have focused on identifying novel medications or combinations of current immunosuppressants to expand regulatory T cells in vivo and increase the Treg/T effector ratio to tip the allograft outcome towards tolerance (24–28). Bestard et al used a combination of rATG, belatacept (Bela) and sirolimus (SRL) in a group of renal transplant recipients and compared them to other groups treated with a CNI-based (rATG/tacrolimus/MMF), and two other Bela-based regimens (rATG/Bela/MMF and basiliximab/Bela/MMF/steroids). Their results were highlighted by an increased ratio of Tregs to memory T cells in the rATG/Bela/SRL arm of the study during the first year of follow up (29). Our group has also shown in vitro that low dose rATG is able to expand human Tregs (30). While we still await mechanistic results from low dose rATG induction clinical trials, Gurkan et al described the immune profile in pediatric and adult renal transplant recipients following rATG induction therapy and demonstrated an expansion of circulating regulatory T cells in vivo (31).
Further studies in this area are warranted to identify the safest and most effective combination strategy, however, given the complexity of immune pathways, it is likely that individually tailored regimens guided by accurate biomarkers of rejection and tolerance will be the ultimate goal.
2. Regulatory B Cells (Bregs)
B cell deficiency or depletion worsens disease in mouse models of EAE, IBD, and contact hypersensitivity, suggesting that B cells can exhibit immunomodulatory function (32–34). The importance of B cells in human transplantation tolerance is becoming a subject of increasing research efforts. This is highlighted by Newell et al when they identified, using gene expression profiles and peripheral blood lymphocyte subset analysis, a characteristic B cell “signature” in a cohort of tolerant renal transplant recipients who had stable graft function and received no immunosuppression for more than 1 year (35).
Harnessing the tolerance inducing properties of regulatory B cells requires the identification of their unique properties and surface markers to facilitate their isolation and expansion for therapeutic purposes. Carter et al recently reported their findings in an IL-10−/− B cell deficient mouse model where they noted exacerbated arthritis compared to the wild type control. They found a decrease in the absolute numbers of FoxP3+ Tregs and increased IFN-γ and IL-17A producing CD4+ T cells in the IL-10−/− B cell deficient mice that was reset by adoptive transfer of wild type Bregs confirming a role of this subset in modulating the balance of effector and regulatory T cell subsets (36). Rothstein et al (37) have recently shown that the surface marker TIM-1 identifies a large majority of B cells capable of IL-4 and IL-10 expression. Those TIM-1+ Bregs are induced by TIM-1 ligation and can transfer long-term acceptance of islet allografts to untreated recipients in an IL-10 dependent fashion. Therefore, both the surface expression of TIM-1 and cytokine production of IL-10 provides attractive options for future studies of Bregs.
C. Tipping the Balance
The balance between effector and regulatory mechanisms is strongly influenced by the cytokine milieu and the pattern of co-stimulation. APCs, particularly DCs, are the fulcrum upon which the balance between tolerance and immunity pivots (38). DCs play a key role in antigen presentation, and, especially in the context of endogenous or exogenous “danger signals” from pattern recognition receptors, are capable of orchestrating CD4+ T helper cell differentiation through costimulatory interaction and/or soluble cytokines (39, 40).
1. Cytokines
Naïve T cells commitment to an effector or regulatory phenotype as well as the “plasticity” of Tregs/Th17 cells are strongly influenced by the local cytokine environment (41). Park et al exploited this plasticity to promote regulatory T cell expansion by directed cytokine delivery using nanoparticles (42). Inflammatory cytokines of the innate system (IL-6, TNF-α, and IL1-β) have been associated with both chronic renal and cardiac allograft rejection (43, 44). Data from our group has revealed that the pro-inflammatory cytokine IL-6 plays a critical role in allograft rejection in a fully mismatched mouse cardiac transplant model. CTLA4Ig treatment led to allograft acceptance in IL-6 −/− mice while wild type recipients rejected the graft suggesting that intact IL-6 production contributed to rejection (45). It remains to be seen whether blocking humanized IL-6 receptor antibodies such as tocilizumab, which has recently been used for treatment in rheumatoid arthritis (46), will be effective in human transplantation. Finally, there are emerging data about the role of IL-23 in allograft rejection (47). IL-12 and IL-23 share the p40 subunit and are crucial for the development of Th1 and Th17 cell responses in acute allograft rejection. A recent work demonstrated that treatment with anti-p40 inhibited Th1- and Th17-cell responses and prolonged survival of cardiac allograft in mice (48). There are also new data indicating IL23/IL17 axis in bronchiolitis obliterans syndrome, the leading cause of death after lung transplantation(49).
2. Co-stimulatory molecules
Costimulatory molecules play an important role in determining the balance between effector and regulatory mechanisms and ultimately the fate of the allograft. This topic was extensively reviewed in an excellent article by Li et al (40).
Novel ligands such as T cell Ig mucin (TIM) molecules are increasingly recognized as promising co-stimulatory targets in transplantation, as there is compelling evidence that molecules in the TIM family may broadly regulate Treg/Breg and Th1/Th2/Th17 activity (37, 40, 50, 51). TIM molecules are type I transmembrane glycoproteins, that can be expressed on antigen presenting cells (52), T cells (53, 54) and B cells (37). Our group demonstrated prolonged survival of fully MHC-mismatched vascularized mouse cardiac allografts using a short course of antagonistic TIM-1-specific antibody monotherapy. The prolongation was associated with inhibition of alloreactive Th1 responses and preservation of Th2 responses and the effect was abrogated by depletion of natural Tregs (51). Targeting the same TIM-1 molecule with a specific mAb with agonist properties (3B3) prevented allograft tolerance as shown by Degauque et al in diabetic C57BL/6 mice transplanted with DBA/2 pancreatic islet and treated with an anti-CD154 mAb. The authors performed in vitro experiments where the use of TIM-1 agonist 3B3 prevented the induction of CD4+FoxP3+ Tregs from FoxP3− precursors and was associated with a shift towards Th17 differentiation (53). TIM-3: galectin-9 pathway has been shown to affect Th1/Th17 effector T cells. Recent studies from one group of investigators demonstrated that administration of recombinant mouse or human gal-9 can prolong MHC-mismatched skin and heart transplant survival (55–58). Graft prolongation was accompanied by a decrease in the overall number of TIM-3 expressing, CD4+ and CD8+ T cells in the draining lymph nodes, as well as a decline of INF-γ and IL-17 mRNA in the grafts. Based on pro-apoptotic effects of recombinant gal-9 seen in vitro, the authors propose the possibility that recombinant gal-9 may selectively induce the death of TIM-3-expressing Th1/Th17 cells in vivo. Recently, our group studied the physiological role of TIM-3: gal-9 pathway in cardiac allograft rejection, by interrupting the interaction of TIM-3 with endogenous gal-9 using a blocking anti-TIM-3 monoclonal antibody (RMT3–23) (50). We demonstrated that blockade of TIM-3 accelerates allograft rejection by increasing IL-6 production in recipient CD4+ T cells. Accelerated rate of rejection is also accompanied by increased frequencies of alloreactive splenic Th1/Th17 T cells, a rise in Th17 lymphocytes and a decline in the frequency of allograft Tregs. These data point to TIM-3 as a central master switch, with the unique ability to broadly modulate CD4+ effector T cell differentiation, possibly by its ability to regulate IL-6 production by CD4+ T cells. Activation of innate immunity and tissue inflammation has an important impact on the differentiation of naïve T cells into Th1/Th17/Tregs. Targeting costimulatory pathways that could control innate immune cells might therefore be important in Treg induction and/or effector T cell differentiation in vivo, and consequently critical for tolerance induction. TIM-3 is expressed by some innate immune cells and may play a role in their function, thus indirectly affecting adaptive immunity (59). The reader is directed to this review by Yeung et al for more information on the role of TIM molecules in transplantation (60).
3. Dendritic cells (DCs)
Dendritic cells are the prototypical antigen presenting cells and have the capacity to direct the adaptive immune responses towards an inflammatory or tolerant state (61). The development of recipient DCs to an inflammatory or tolerogenic phenotype in vitro was related to HLA-C genotype as shown by Hanvesakul et al (62). In their study of 760 renal transplant recipients, those with HLA-C2 genotype had significantly better long term graft outcome compared to those with HLA-C1 genotype. In vitro, the HLA-C2 genotype was associated with less mature “tolerogenic” DCs secreting anti-inflammatory cytokines while the HLA-C1 genotype was associated with pro-inflammatory cytokine production and enhanced DCs maturation.
It comes as no surprise then that investigators are attempting to modulate DCs into a tolerogenic phenotype for clinical application. Lan and Ge et al utilized soluble CD83 as a DC modulating agent capable of attenuating DC maturation. Those immature DCs were successful in inducing donor-specific allograft tolerance in both a cardiac and renal mouse transplant models (63, 64). Another approach involving an adenovirus encoding PD-L1 viral vector was used by Peng et al to transfect DCs and later transfer them into an MHC-mismatched rat kidney transplant model (65). The approach was successful in suppressing the proliferation and cytokine secretion of CD8+ T cells in recipient rats and also slowed the development of proteinuria and prolonged their survival.
Conclusion
Despite the identification of new regulatory cell subsets and better understanding of co-stimulatory pathways and the contribution of innate immunity to allograft outcomes, achieving tolerance still remains an elusive goal. Future research efforts should focus on harnessing the benefits of regulatory T and B cells, and tolerogenic dendritic cells while selectively depleting effector memory T cells. To further maintain established tolerance, efforts should be directed at engaging tolerogenic costimulatory pathways and minimizing the negative influence of a pro-inflammatory cytokine environment as seen in infection and ischemia-reperfusion injury.
Key points.
Allograft outcome (rejection vs. tolerance) is determined by the balance of effector and regulatory mechanisms.
Both naïve and established T cell phenotypes can change their commitment providing a potential therapeutic target to expand regulatory T cells but also posing a challenge to establishing long-term tolerance.
The balance of positive and negative co-stimulatory molecule activation is critical to the fate of the allograft and new promising targets such as the TIM molecules are currently being investigated.
The innate immune system is intricately linked to the adaptive immune system and can determine allograft outcomes through inflammatory cytokines and tolerogenic dendritic cells.
Acknowledgments
This work was supported by NIH grant RO1AI070820 and R56AI089777.
Footnotes
Conflicts of interest:
None declared
References and Recommended Reading
Papers of particular interest, published within the annual period of review, have been highlighted as:
* of special interest
** of outstanding interest
- 1.Suthanthiran M. Transplantation tolerance: fooling mother nature. Proc Natl Acad Sci U S A. 1996 Oct 29;93(22):12072–5. doi: 10.1073/pnas.93.22.12072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Billingham RE, Brent L, Medawar PB. Actively acquired tolerance of foreign cells. Nature. 1953 Oct 3;172(4379):603–6. doi: 10.1038/172603a0. [DOI] [PubMed] [Google Scholar]
- 3.Safinia N, Afzali B, Atalar K, Lombardi G, Lechler RI. T-cell alloimmunity and chronic allograft dysfunction. Kidney Int Suppl. 2010 Dec;(119):S2–12. doi: 10.1038/ki.2010.416. [DOI] [PubMed] [Google Scholar]
- 4.Heidt S, Segundo DS, Chadha R, Wood KJ. The impact of Th17 cells on transplant rejection and the induction of tolerance. Curr Opin Organ Transplant. 2010 Aug;15(4):456–61. doi: 10.1097/MOT.0b013e32833b9bfb. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Chadha R, Heidt S, Jones ND, Wood KJ. Th17: contributors to allograft rejection and a barrier to the induction of transplantation tolerance? Transplantation. 2011 May 15;91(9):939–45. doi: 10.1097/TP.0b013e3182126eeb. [DOI] [PubMed] [Google Scholar]
- 6*.Hester J, Mills N, Shankar S, Carvalho-Gaspar M, Friend P, Wood KJ. Th17 cells in alemtuzumab-treated patients: the effect of long-term maintenance immunosuppressive therapy. Transplantation. 2011 Apr 15;91(7):744–50. doi: 10.1097/TP.0b013e31820c85dc. This study shows an icrease in IL-17A producing T cells in kidney transplant recipients maintained on sirolimus monotherapy following alemtuzumab induction compared to patients maintained on sirolimus and mycophenolate mofetil. This was compensated for by an increase in Treg frequency and number in the monotherapy group. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kappel LW, Goldberg GL, King CG, Suh DY, Smith OM, Ligh C, et al. IL-17 contributes to CD4-mediated graft-versus-host disease. Blood. 2009 Jan 22;113(4):945–52. doi: 10.1182/blood-2008-08-172155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fabrega E, Lopez-Hoyos M, San Segundo D, Casafont F, Pons-Romero F. Changes in the serum levels of interleukin-17/interleukin-23 during acute rejection in liver transplantation. Liver Transpl. 2009 Jun;15(6):629–33. doi: 10.1002/lt.21724. [DOI] [PubMed] [Google Scholar]
- 9.Wang S, Li J, Xie A, Wang G, Xia N, Ye P, et al. Dynamic changes in Th1, Th17, and FoxP3+ T cells in patients with acute cellular rejection after cardiac transplantation. Clin Transplant. 2011 Mar–Apr;25(2):E177–86. doi: 10.1111/j.1399-0012.2010.01362.x. [DOI] [PubMed] [Google Scholar]
- 10**.Hirota K, Duarte JH, Veldhoen M, Hornsby E, Li Y, Cua DJ, et al. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat Immunol. 2011 Mar;12(3):255–63. doi: 10.1038/ni.1993. This study describes the generation of a novel reporter mouse able to trace the fate of IL-17A expressing cell. The model is useful in demonstrating the plasticity of Th17 cells under different inflammatory experimental conditions. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Soroosh P, Doherty TA. Th9 and allergic disease. Immunology. 2009 Aug;127(4):450–8. doi: 10.1111/j.1365-2567.2009.03114.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wong MT, Ye JJ, Alonso MN, Landrigan A, Cheung RK, Engleman E, et al. Regulation of human Th9 differentiation by type I interferons and IL-21. Immunol Cell Biol. 2010 Aug;88(6):624–31. doi: 10.1038/icb.2010.53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Poulin LF, Richard M, Le Moine A, Kiss R, McKenzie AN, Goldman M, et al. Interleukin-9 promotes eosinophilic rejection of mouse heart allografts. Transplantation. 2003 Aug 15;76(3):572–7. doi: 10.1097/01.TP.0000071201.32424.D2. [DOI] [PubMed] [Google Scholar]
- 14.King C, Tangye SG, Mackay CR. T follicular helper (TFH) cells in normal and dysregulated immune responses. Annu Rev Immunol. 2008;26:741–66. doi: 10.1146/annurev.immunol.26.021607.090344. [DOI] [PubMed] [Google Scholar]
- 15*.Choi YS, Kageyama R, Eto D, Escobar TC, Johnston RJ, Monticelli L, et al. ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity. 2011 Jun 24;34(6):932–46. doi: 10.1016/j.immuni.2011.03.023. This study demonstrates the critical role of ICOS signalling in inducing the transcription factor bcl6 to drive the differentiation of Tfh cells even in the absence of B cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhou X, Bailey-Bucktrout SL, Jeker LT, Penaranda C, Martinez-Llordella M, Ashby M, et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol. 2009 Sep;10(9):1000–7. doi: 10.1038/ni.1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lee YK, Mukasa R, Hatton RD, Weaver CT. Developmental plasticity of Th17 and Treg cells. Curr Opin Immunol. 2009 Jun;21(3):274–80. doi: 10.1016/j.coi.2009.05.021. [DOI] [PubMed] [Google Scholar]
- 18.Kollins D, Stoelcker B, Hoffmann U, Bergler T, Reinhold S, Banas MC, et al. FOXP3+ regulatory T-cells in renal allografts: correlation with long-term graft function and acute rejection. Clin Nephrol. 2011 Feb;75(2):91–100. [PubMed] [Google Scholar]
- 19.Batsford S, Dickenmann M, Durmuller U, Hopfer H, Gudat F, Mihatsch M. Is monitoring of FOXP3 Treg cells in renal transplants during acute cellular rejection episodes useful? Clin Nephrol. 2011 Feb;75(2):101–6. [PubMed] [Google Scholar]
- 20*.Iwase H, Kobayashi T, Kodera Y, Miwa Y, Kuzuya T, Iwasaki K, et al. Clinical significance of regulatory T-cell-related gene expression in peripheral blood after renal transplantation. Transplantation. 2011 Jan 27;91(2):191–8. doi: 10.1097/TP.0b013e3181ffbab4. The authors used qPCR to measure mRNA expression levels of various Treg related molecules in peripheral blood from renal transplant recipients with stable graft function and recipients with chronic rejection and found FoxP3 mRNA expression to be significantly lower in the chronic rejection group. [DOI] [PubMed] [Google Scholar]
- 21.Bhorade SM, Chen H, Molinero L, Liao C, Garrity ER, Vigneswaran WT, et al. Decreased percentage of CD4+FoxP3+ cells in bronchoalveolar lavage from lung transplant recipients correlates with development of bronchiolitis obliterans syndrome. Transplantation. 2010 Sep 15;90(5):540–6. doi: 10.1097/TP.0b013e3181e8dabe. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Semiletova NV, Shen XD, Baibakov B, Andakyan A. Intensity of transplant chronic rejection correlates with level of graft-infiltrating regulatory cells. J Heart Lung Transplant. 2010 Mar;29(3):335–41. doi: 10.1016/j.healun.2009.08.003. [DOI] [PubMed] [Google Scholar]
- 23.Moraes-Vieira PM, Silva HM, Takenaka MC, Monteiro SM, Lemos F, Saitovitch D, et al. Differential monocyte STAT6 activation and CD4(+)CD25(+)Foxp3(+) T cells in kidney operational tolerance transplanted individuals. Hum Immunol. 2010 May;71(5):442–50. doi: 10.1016/j.humimm.2010.01.022. [DOI] [PubMed] [Google Scholar]
- 24.De Serres SA, Sayegh MH, Najafian N. Immunosuppressive drugs and Tregs: a critical evaluation! Clin J Am Soc Nephrol. 2009 Oct;4(10):1661–9. doi: 10.2215/CJN.03180509. [DOI] [PubMed] [Google Scholar]
- 25.Raimondi G, Sumpter TL, Matta BM, Pillai M, Corbitt N, Vodovotz Y, et al. Mammalian target of rapamycin inhibition and alloantigen-specific regulatory T cells synergize to promote long-term graft survival in immunocompetent recipients. J Immunol. 2010 Jan 15;184(2):624–36. doi: 10.4049/jimmunol.0900936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.D’Addio F, Yuan X, Habicht A, Williams J, Ruzek M, Iacomini J, et al. A novel clinically relevant approach to tip the balance toward regulation in stringent transplant model. Transplantation. 2010 Aug 15;90(3):260–9. doi: 10.1097/tp.0b013e3181e64217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lim DG, Koo SK, Park YH, Kim Y, Kim HM, Park CS, et al. Impact of immunosuppressants on the therapeutic efficacy of in vitro-expanded CD4+CD25+Foxp3+ regulatory T cells in allotransplantation. Transplantation. 2010 Apr 27;89(8):928–36. doi: 10.1097/TP.0b013e3181d3c9d4. [DOI] [PubMed] [Google Scholar]
- 28.Poirier N, Azimzadeh AM, Zhang T, Dilek N, Mary C, Nguyen B, et al. Inducing CTLA-4-dependent immune regulation by selective CD28 blockade promotes regulatory T cells in organ transplantation. Sci Transl Med. 2010 Feb 3;2(17):17ra0. doi: 10.1126/scitranslmed.3000116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29**.Bestard O, Cassis L, Cruzado JM, Torras J, Franquesa M, Gil-Vernet S, et al. Costimulatory blockade with mTor inhibition abrogates effector T-cell responses allowing regulatory T-cell survival in renal transplantation. Transpl Int. 2011 May;24(5):451–60. doi: 10.1111/j.1432-2277.2011.01223.x. This study describes the immune profile of a cohort of renal transplant recipients 1 year following transplantation. The group receiving lymphocyte depletion with rATG, costimulatory blockade with belatacept and sirolimus maintenance therapy demonstrated an increased Treg/memory T cell ratio and potent anti-donor suppressive activity. [DOI] [PubMed] [Google Scholar]
- 30.Lopez M, Clarkson MR, Albin M, Sayegh MH, Najafian N. A novel mechanism of action for anti-thymocyte globulin: induction of CD4+CD25+Foxp3+ regulatory T cells. J Am Soc Nephrol. 2006 Oct;17(10):2844–53. doi: 10.1681/ASN.2006050422. [DOI] [PubMed] [Google Scholar]
- 31.Gurkan S, Luan Y, Dhillon N, Allam SR, Montague T, Bromberg JS, et al. Immune reconstitution following rabbit antithymocyte globulin. Am J Transplant. 2010 Sep;10(9):2132–41. doi: 10.1111/j.1600-6143.2010.03210.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Watanabe R, Fujimoto M, Ishiura N, Kuwano Y, Nakashima H, Yazawa N, et al. CD19 expression in B cells is important for suppression of contact hypersensitivity. Am J Pathol. 2007 Aug;171(2):560–70. doi: 10.2353/ajpath.2007.061279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yanaba K, Bouaziz JD, Haas KM, Poe JC, Fujimoto M, Tedder TF. A regulatory B cell subset with a unique CD1dhiCD5+ phenotype controls T cell-dependent inflammatory responses. Immunity. 2008 May;28(5):639–50. doi: 10.1016/j.immuni.2008.03.017. [DOI] [PubMed] [Google Scholar]
- 34.Matsushita T, Yanaba K, Bouaziz JD, Fujimoto M, Tedder TF. Regulatory B cells inhibit EAE initiation in mice while other B cells promote disease progression. J Clin Invest. 2008 Oct;118(10):3420–30. doi: 10.1172/JCI36030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Newell KA, Asare A, Kirk AD, Gisler TD, Bourcier K, Suthanthiran M, et al. Identification of a B cell signature associated with renal transplant tolerance in humans. J Clin Invest. 2010 Jun 1;120(6):1836–47. doi: 10.1172/JCI39933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36**.Carter NA, Vasconcellos R, Rosser EC, Tulone C, Munoz-Suano A, Kamanaka M, et al. Mice lacking endogenous IL-10-producing regulatory B cells develop exacerbated disease and present with an increased frequency of Th1/Th17 but a decrease in regulatory T cells. J Immunol. 2011 May 15;186(10):5569–79. doi: 10.4049/jimmunol.1100284. This study describes the functional importance of Bregs using an IL-10−/−Bcell mouse model. These mice developed exacerbated arthritis compared to wild type B cell mice with associated significant decrease in FoxP3+ Tregs and marked increase in Th1 and Th17 cells. [DOI] [PubMed] [Google Scholar]
- 37**.Ding Q, Yeung M, Camirand G, Zeng Q, Akiba H, Yagita H, et al. Regulatory B cells are identified by expression of TIM-1 and can be induced through TIM-1 ligation to promote tolerance in mice. J Clin Invest. 2011 Sep 1;121(9):3645–56. doi: 10.1172/JCI46274. This study reveals that TIM-1 is expressed on IL-10 secreting Bregs and can be used to identify this subset. The authors also use a TIM-1 specific antibody able to induce TIM-1+ Bregs in an IL-4 dependent manner. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Thomson AW. Tolerogenic dendritic cells: all present and correct? Am J Transplant. 2010 Feb;10(2):214–9. doi: 10.1111/j.1600-6143.2009.02955.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Boenisch O, Sayegh MH, Najafian N. Negative T-cell costimulatory pathways: their role in regulating alloimmune responses. Curr OpinOrgan Transplant. 2008 Aug;13(4):373–8. doi: 10.1097/MOT.0b013e328306117f. [DOI] [PubMed] [Google Scholar]
- 40.Li XC, Rothstein DM, Sayegh MH. Costimulatory pathways in transplantation: challenges and new developments. Immunol Rev. 2009 May;229(1):271–93. doi: 10.1111/j.1600-065X.2009.00781.x. [DOI] [PubMed] [Google Scholar]
- 41.Strom TB, Koulmanda M. Recently discovered T cell subsets cannot keep their commitments. J Am Soc Nephrol. 2009 Aug;20(8):1677–80. doi: 10.1681/ASN.2008101027. [DOI] [PubMed] [Google Scholar]
- 42**.Park J, Gao W, Whiston R, Strom TB, Metcalfe S, Fahmy TM. Modulation of CD4+ T lymphocyte lineage outcomes with targeted, nanoparticle-mediated cytokine delivery. Mol Pharm. 2011 Feb 7;8(1):143–52. doi: 10.1021/mp100203a. This study uses nanoparticles loaded with either Luekemia inhibitory factor (LIF) or IL-6 targeted towards CD4+ T cells to drive their differentiation towards FoxP3+ T cells or Th17 cells respectively, demonstrating a novel method to exploit T cell plasticity for therapeutic outcomes. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43*.De Serres SA, Vadivel N, Mfarrej BG, Grafals M, DeJoseph M, Dyer C, et al. Monocyte-secreted inflammatory cytokines are associated with transplant glomerulopathy in renal allograft recipients. Transplantation. 2011 Mar 15;91(5):552–9. doi: 10.1097/TP.0b013e318205b3c1. This study shows that monocytes secrete inflammatory cytokines, namely IL-1β, IL-6 and TNF-α in renal transplant recipients with transplant glomerulopathy and that these monocytes seem to be under the regulation of functional regulatory T cells, demonstrating the reciprocal interaction between the innate and adaptive immune system. [DOI] [PubMed] [Google Scholar]
- 44.Diaz JA, Booth AJ, Lu G, Wood SC, Pinsky DJ, Bishop DK. Critical role for IL-6 in hypertrophy and fibrosis in chronic cardiac allograft rejection. Am J Transplant. 2009 Aug;9(8):1773–83. doi: 10.1111/j.1600-6143.2009.02706.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45**.Zhao X, Boenisch O, Yeung M, Mfarrej B, Yang S, Turka LA, et al. Critical Role of Proinflammatory Cytokine IL-6 in Allograft Rejection and Tolerance. Am J Transplant. 2011 Oct 12; doi: 10.1111/j.1600-6143.2011.03770.x. This study utilizes IL-6 −/− mice in a cardiac allograft experiment and shows allograft acceptance when treated with CTLA4Ig compared to CTLA4Ig treated wild type mice that reject the graft. This suggests that targeting the IL-6 signalling pathway could be a valuable adjunct in promoting allograft tolerance. [DOI] [PubMed] [Google Scholar]
- 46.Burmester GR, Feist E, Kellner H, Braun J, Iking-Konert C, Rubbert-Roth A. Effectiveness and safety of the interleukin 6-receptor antagonist tocilizumab after 4 and 24 weeks in patients with active rheumatoid arthritis: the first phase IIIb real-life study (TAMARA) Ann Rheum Dis. 2011 May;70(5):755–9. doi: 10.1136/ard.2010.139725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Chen Y, Wood KJ. Interleukin-23 and TH17 cells in transplantation immunity: does 23+17 equal rejection? Transplantation. 2007 Nov 15;84(9):1071–4. doi: 10.1097/01.tp.0000287126.12083.48. [DOI] [PubMed] [Google Scholar]
- 48.Xie A, Wang S, Zhang K, Wang G, Ye P, Li J, et al. Treatment with interleukin-12/23p40 antibody attenuates acute cardiac allograft rejection. Transplantation. 2011 Jan 15;91(1):27–34. doi: 10.1097/tp.0b013e3181fdd948. [DOI] [PubMed] [Google Scholar]
- 49.Vanaudenaerde BM, De Vleeschauwer SI, Vos R, Meyts I, Bullens DM, Reynders V, et al. The role of the IL23/IL17 axis in bronchiolitis obliterans syndrome after lung transplantation. Am J Transplant. 2008 Sep;8(9):1911–20. doi: 10.1111/j.1600-6143.2008.02321.x. [DOI] [PubMed] [Google Scholar]
- 50*.Boenisch O, D’Addio F, Watanabe T, Elyaman W, Magee CN, Yeung MY, et al. TIM-3: a novel regulatory molecule of alloimmune activation. J Immunol. 2010 Nov 15;185(10):5806–19. doi: 10.4049/jimmunol.0903435. This study describes a novel role of TIM-3 in a murine model of vascularized cardiac transplantation using a blocking anti-TIM-3 mAb. Treatment led to accelerated rejection but only in the presence of host CD4+T cells and was associated with skewing towards an effector T cell phenotype. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ueno T, Habicht A, Clarkson MR, Albin MJ, Yamaura K, Boenisch O, et al. The emerging role of T cell Ig mucin 1 in alloimmune responses in an experimental mouse transplant model. J Clin Invest. 2008 Feb;118(2):742–51. doi: 10.1172/JCI32451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kuchroo VK, Dardalhon V, Xiao S, Anderson AC. New roles for TIM family members in immune regulation. Nat Rev Immunol. 2008 Aug;8(8):577–80. doi: 10.1038/nri2366. [DOI] [PubMed] [Google Scholar]
- 53.Degauque N, Mariat C, Kenny J, Zhang D, Gao W, Vu MD, et al. Immunostimulatory Tim-1-specific antibody deprograms Tregs and prevents transplant tolerance in mice. J Clin Invest. 2008 Feb;118(2):735–41. doi: 10.1172/JCI32562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hastings WD, Anderson DE, Kassam N, Koguchi K, Greenfield EA, Kent SC, et al. TIM-3 is expressed on activated human CD4+ T cells and regulates Th1 and Th17 cytokines. Eur J Immunol. 2009 Sep;39(9):2492–501. doi: 10.1002/eji.200939274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.He W, Fang Z, Wang F, Wu K, Xu Y, Zhou H, et al. Galectin-9 significantly prolongs the survival of fully mismatched cardiac allografts in mice. Transplantation. 2009 Sep 27;88(6):782–90. doi: 10.1097/TP.0b013e3181b47f25. [DOI] [PubMed] [Google Scholar]
- 56.Wang F, He W, Yuan J, Wu K, Zhou H, Zhang W, et al. Activation of Tim-3-Galectin-9 pathway improves survival of fully allogeneic skin grafts. Transpl Immunol. 2008 Apr;19(1):12–9. doi: 10.1016/j.trim.2008.01.008. [DOI] [PubMed] [Google Scholar]
- 57.Wang F, He W, Zhou H, Yuan J, Wu K, Xu L, et al. The Tim-3 ligand galectin-9 negatively regulates CD8+ alloreactive T cell and prolongs survival of skin graft. Cell Immunol. 2007 Nov–Dec;250(1–2):68–74. doi: 10.1016/j.cellimm.2008.01.006. [DOI] [PubMed] [Google Scholar]
- 58.Wang F, Wan L, Zhang C, Zheng X, Li J, Chen ZK. Tim-3-Galectin-9 pathway involves the suppression induced by CD4+CD25+ regulatory T cells. Immunobiology. 2009;214(5):342–9. doi: 10.1016/j.imbio.2008.10.007. [DOI] [PubMed] [Google Scholar]
- 59.Anderson AC, Anderson DE, Bregoli L, Hastings WD, Kassam N, Lei C, et al. Promotion of tissue inflammation by the immune receptor Tim-3expressed on innate immune cells. Science. 2007 Nov 16;318(5853):1141–3. doi: 10.1126/science.1148536. [DOI] [PubMed] [Google Scholar]
- 60.Yeung MY, McGrath M, Najafian N. The Emerging Role of the TIM Molecules in Transplantation. Am J Transplant. 2011 Oct;11(10):2012–9. doi: 10.1111/j.1600-6143.2011.03727.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hu J, Wan Y. Tolerogenic dendritic cells and their potential applications. Immunology. 2011 Mar;132(3):307–14. doi: 10.1111/j.1365-2567.2010.03396.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hanvesakul R, Kubal C, Moore J, Neil D, Cook M, Ball S, et al. KIR and HLA-C Interactions Promote Differential Dendritic Cell Maturation and Is a Major Determinant of Graft Failure following Kidney Transplantation. PLoS One. 2011;6(8):e23631. doi: 10.1371/journal.pone.0023631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63*.Lan Z, Ge W, Arp J, Jiang J, Liu W, Gordon D, et al. Induction of kidney allograft tolerance by soluble CD83 associated with prevalence of tolerogenic dendritic cells and indoleamine 2,3-dioxygenase. Transplantation. 2010 Dec 27;90(12):1286–93. doi: 10.1097/TP.0b013e3182007bbf. The authors successfully used human soluble CD83 treatment to induce kidney allograft tolerance in a murine model. DCs from the recipients demosntrated a “tolerant” phenotype with decreased surface expression of MHC Class II, CD80 and CD40. [DOI] [PubMed] [Google Scholar]
- 64*.Ge W, Arp J, Lian D, Liu W, Baroja ML, Jiang J, et al. Immunosuppression involving soluble CD83 induces tolerogenic dendritic cells that prevent cardiac allograft rejection. Transplantation. 2010 Dec 15;90(11):1145–56. doi: 10.1097/TP.0b013e3181f95718. This study provides another model for using soluble CD83 to tolerize DCs and achieve cardiac allograft tolerance when used in combination with other therapies. [DOI] [PubMed] [Google Scholar]
- 65.Peng W, Ran B, Ma Y, Huang X, Chang Q, Wang X. Dendritic cells transfected with PD-L1 recombinant adenovirus induces T cell suppression and long-term acceptance of allograft transplantation. Cell Immunol. 2011;271(1):73–7. doi: 10.1016/j.cellimm.2011.06.007. [DOI] [PubMed] [Google Scholar]