Skip to main content

MINI REVIEW article

Front. Immunol., 14 September 2015
Sec. Alloimmunity and Transplantation
This article is part of the Research Topic Memory T cells: effectors, regulators and implications for transplant tolerance View all 6 articles

Role of memory T cells and perspectives for intervention in organ transplantation

\r\nKailin Lin&#x;Kailin Lin1†Song Chen,,&#x;Song Chen1,2,3†Gang Chen,,*\r\n   Gang Chen1,2,3*
  • 1Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
  • 2Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
  • 3Key Laboratory of Organ Transplantation, Ministry of Public Health, Wuhan, China

Memory T cells are necessary for protective immunity against invading pathogens, especially under conditions of immunosuppression. However, their presence also threatens transplant survival, making transplantation a great challenge. Significant progress has been achieved in recent years in advancing our understanding of the role that memory T cells play in transplantation. This review focuses on the latest advances in our understanding of the involvement of memory T cells in graft rejection and transplant tolerance and discusses potential strategies for targeting memory T cells in order to minimize allograft rejection and optimize clinical outcomes.

Introduction

Adaptive immune responses depend on the ability to recognize and eliminate recurrent pathogens, resulting in the generation of memory lymphocytes. The capacity of memory T cells to rapidly mobilize and initiate a potent recall response enhances protective immunity against previously encountered pathogens. However, the characteristics of memory T cells, with their lowered activation thresholds and lower susceptibility to conventional immunosuppressive agents, also makes them a significant obstacle to successful transplantation (1).

In the context of transplantation, three independent mechanisms for the generation of alloreactive memory T cells have been described. First, since memory T cells can be generated directly during primary immune responses, alloantigenic stimulation of naïve T cells serves as the most direct source of alloreactive memory T cells (2). Second, alloreactive memory T cells can also be generated through homeostatic proliferation, a spontaneous process that occurs in response to transient lymphopenia and induces the proliferation and differentiation of naïve T cells into memory cells; in transplantation, severe lymphopenia is often seen after T-cell depletion therapy (3). Third, memory T cells generated in response to infectious or environmental antigens have the potential to cross-react with donor allogeneic MHC molecules (4), creating a situation known as heterologous immunity. In recent years, a broader perspective has been established with regard to our understanding of the basic biology of memory T cells. Also, new approaches for modulating memory T cells have been designed and tested in various models, including transplantation. In this review, we mainly discuss the role of memory T cells in transplantation and explore the development of therapeutic strategies that can directly target memory T cells.

The Role of Memory T Cells in Transplantation

In contrast to naïve T cells, memory T cells have lower activation thresholds and are less dependent on costimulation signals (5), are more resistant to regulation by regulatory T cells (Treg) (6), and are less susceptible to conventional immunosuppressive agents (1). These features make memory T cells a significant obstacle to successful transplantation. However, the exact mechanisms involved remain unclear. In the pursuit of innovative and effective strategies to inhibit or deplete memory T cells and improve the clinical outcomes, a profound and comprehensive understanding of its role in transplantation is urgently required. Therefore, studies based upon animal models and clinical observations have therefore been established to directly or indirectly demonstrate the impact of memory T cells on transplantation and to elucidate the relevant mechanisms.

One Chief Culprit for Rejection

In addition to their role in protective immunity, memory T cells also take part in mediating transplant rejection (7). It is well understood that donor-specific memory T cells mediate the so-called second-set rejection that is rather difficult to block or inhibit (8, 9). In animal models, memory T cells alone are sufficient to trigger rejection, during which they are among the first cell types infiltrating the grafts (10). In recent observations from clinical practice, the expansion of memory CD8+ T cells and memory CD4+ T cells has been found to be associated with acute cellular and antibody-mediated rejection, respectively, in liver transplantation (11, 12), and these findings are consistent with reported experimental outcomes.

In the absence of direct stimulation by alloantigen, memory T cells generated through either homeostatic proliferation or heterologous immunity seem to have the same ability to mediate transplant rejection. It has been demonstrated that the memory T cells generated via homeostatic proliferation act as potent effector cells in the rejection of heart and skin allografts (5). Likewise, heterologous immunity may also lead to the generation of alloreactive memory T cells (13). Functional heterogeneity of the human T-cell response induced by pathogens and vaccines has recently been described (14). Infection of B6 mice with parasitic or viral antigens, for instance, has been found to cause the generation of alloreactive memory T cells. As a result, heart grafts transplanted into these pathogen-recognizing mice demonstrate an accelerated rejection (15). It has also been reported that naïve untreated adult laboratory mice possess a repertoire of endogenous memory T cells that are naturally generated by environmental exposure (16). Endogenous memory T cells are not previously primed to donor antigens; however, a proportion of the endogenous memory T-cell repertoire in naïve mice is reactive with donor class I MHC molecules, and these T cells are rapidly detected and infiltrate into cardiac allografts within hours of reperfusion (17). In other selected models, memory T cells have been shown to be pivotal mediators of chronic allograft rejection, implying that memory T cells and chronic rejection may also be tightly associated (18, 19). Therefore, despite the different mechanisms by which they are generated, memory T cells are capable of mediating rejection.

The cognate interactions between memory helper T cells and B cells are also important for the development of allograft rejection. On the one hand, it has been reported that donor-reactive memory CD4+ T cells can provide CD40-independent help to B cells and induce high levels of alloantibodies that contribute to heart allograft rejection in mice (20). The CD40-independent help delivered by memory CD4+ T cells in alloreactive immune responses is mainly mediated by B cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL), as targeting the BAFF cytokine network has been reported to be able to inhibit both humoral and cellular immune responses induced by memory CD4+ T cells (21). Additionally, IFN-γ secreted by pre-existing memory helper cells has been found to be required for the CD40-independent alloantibody responses, which determines both isotype and specificity of donor-reactive alloantibodies and can thus affect allograft (22). On the other hand, B cells can also provide help for T cells via multiple mechanisms promoting T-cell proliferation, differentiation, and survival to generate memory T cells. Experimental evidence has shown that activated T cells cotransferred with B cells give rise to more memory T cells than those transferred without B cells and upon recall, mediated accelerated rejection of skin allografts in mice (23). Therefore, inhibiting memory T/B cells interactions could possibly prevent not only alloantibody production but also generation of long-lived memory T cells, which may improve allograft survival.

Turnoff: Tolerance-Inducing or Not?

Another important concern is the correlation between memory T cells and tolerance induction in transplantation. Memory T cells have been found to be highly resistant to tolerance induction, and tolerizing therapies that are effective in inducing allograft tolerance in naïve animals often fail to do so in memory-rich animals (24). A higher frequency of memory T cells in recipients is associated with a worse transplant outcome and poorer tolerance induction under conventional immunosuppression (25).

Memory T cells are distinguished by their reduced requirement for both TCR stimulation and costimulatory signals for recall responses. Showing differences in the expression of adhesion molecules (such as LFA-1, VLA-4, and CD44), cytokine receptors (such as CD122 and IL-15Rα), and apoptosis involving molecules of the Bcl-2 and caspase-3 families when compared to naïve T cells, donor-reactive memory T cells are relatively refractory to any blockade of conventional costimulatory pathways (2629). In a seminal study, although blockade of costimulation by CD28 and CD154 effectively inhibited graft rejection in naïve recipients, animals that had previously been infected with viruses were found to be refractory to the tolerance-inducing effects of this costimulation blockade; while this study focused primarily on the CD8+ memory T-cell barrier, it is clear that both CD4+ and CD8+ donor-specific memory cells can constitute a barrier to costimulation blockade-induced tolerance (30). A study examining the efficacy of an approach combining costimulatory blockade and bone marrow or donor-specific transfusion (DST) to induce tolerance in non-human primate renal transplantation showed that higher pre-transplant precursor frequencies of donor-reactive memory T cells were correlated with a failure of tolerance induction and acute rejection of the grafts, whereas low pre-transplant frequencies of donor-reactive memory T cells predicted successful tolerance induction and long-term renal survival (31).

Memory T cells have also been shown to exhibit increased resistance to regulation by Treg. In a murine model, transferred CD4+CD25+ cells effectively inhibited the rejection mediated by naïve but not by memory CD4+ T cells (32). Similarly, Treg were found to be unable to regulate CD8+ alloreactive T-cell responses, a finding that was true for both naive Treg and alloantigen-primed Treg, suggesting that strategies to enhance the frequency and/or activation of alloantigen-specific Treg are unlikely to be effective against donor-reactive memory T cells (33). However, another report has suggested that the stimulation of human T cells in vitro with TLR-stimulated plasmacytoid dendritic cells results in the generation of CD8+FoxP3+LAG-3+CTLA-4+ Treg, which can inhibit alloreactive memory T-cell responses (34, 35). It has also been reported that human CD45RAFoxP3hi memory-type Treg use a different T-cell receptor (TCR) repertoire from conventional T cells and play an important role in controlling early immune activation (36). Thus, such regulatory effects might be of clinical importance in the pursuit of a desirable level of tolerance.

Current and Potential Memory T Cell-Directed Intervention Therapies

If the negative impact of memory T cells on transplantation and their distinct hindrance of tolerance induction are taken into account, strategies to target memory T cells may offer a solution to improve transplant outcomes. The pursuit of memory T cell-directed therapies in transplantation has arisen from the observation that standard immunosuppressive agents often have undesirable effects on memory T cells. Nevertheless, studies in several experimental models have suggested that targeting the infiltration, proliferation, activation, and the intrinsic apoptosis pathway of memory T cells and the blocking of memory T/B cells interactions may be promising therapeutic approaches.

Blocking the Infiltration of Memory T Cells

After organ transplantation, the alloreactive memory T cells first infiltrate the grafts, then proliferate and mediate significant injury. Therefore, inhibition of their initial entry into the graft may improve allograft survival. There have been numerous attempts to control memory T-cell infiltration in transplant models. For example, the administration of FTY720, the sphingosine-1 phosphate receptor agonist, has been found to lead to a quarantine of donor-specific memory CD4+ T cells in the peripheral lymph nodes and to postpone heart allograft rejection in mice (37). In this situation, the administration of FTY720 prevents the migration of lymphocytes from thymus and peripheral lymphoid tissues, sequesters T cells in the lymph nodes, and inhibits them from infiltrating the grafts. However, this sequestration does not affect the ability of donor antigen-reactive CD4+ T cells to facilitate the helper signals needed to stimulate a donor-specific antibody response, which plays a vital role in the graft loss in these FTY720-treated recipients.

Other studies have also demonstrated that the disruption of adhesion molecules, e.g., leukocyte integrins such as LFA-1 and VLA-4, is effective in preventing the infiltration of memory T cells into grafts (38, 39). Anti-LFA-1 or anti-VLA-4 monoclonal antibodies can attenuate donor-reactive memory recall responses and reduce T-cell trafficking into allografts in mouse models, resulting in a prolongation of allograft survival (40). However, a preclinical test of the anti-LFA-1 agent Efalizumab in primate renal transplantation suggested that this agent evokes a higher rate of EBV-associated malignancy despite the promising outcomes produced for the grafts (40, 41). Taken together, these successful treatments in animal models, though not currently available in the clinic, suggest that targeting trafficking molecules on memory T cells may be a valid approach, but further investigation is urgently needed to provide a full evaluation of its clinical validity and potential side effects.

Suppressing the Proliferation of Memory T Cells

Another potential therapeutic strategy is targeting the proliferation of memory T cells induced by cytokine and TCR signaling. Janus kinase-3 (JAK-3), the downstream receptor of the common γ chain, binds a magnitude of cytokines, including IL-2, IL-7, IL-9, IL-15, and IL-21 (42). These cytokines have been shown to play pivotal roles in the generation, maintenance, and proliferation of memory T cells. Tofacitinib (CP-690550), a highly selective and potent JAK-3 inhibitor, has been shown to prevent allograft rejection in both rodent and non-human primate models and may offer a novel means of targeting memory T cells without TCR inhibition (43).

Cell proliferation and survival signaling may also be induced through the TCR pathway. Nuclear factor-κB (NF-κB), a protein complex that regulates DNA transcription, plays an important role in the TCR pathway (44). The NF-κB blocker 15-deoxyspergualin can block the activation of donor-specific memory CD8+ T cells and has been shown to induce skin allograft survival in a mouse model in combination with costimulatory blockade (45). The inhibition of NF-κB also suppresses the proliferation of rapamycin-resistant memory T cells in non-human primates (46).

Inhibiting the Activation of Memory T Cells

An increasing amount of evidence has shown that conventional blockade of costimulation has only a minimal effect on memory T cells. Several studies have suggested that memory T cells use alternative, unique costimulatory pathways for activation and effector activity. On CD8+ T cells, it has been shown that the engagement of 4-1BB (CD137) by its ligand provides both CD28-dependent and CD28-independent signals that lead to cytokine production, cell proliferation, augmented cytotoxic effector activity, and enhanced cell survival (47). In mouse models, blockade of the 4-1BB costimulatory pathway has been shown to be valid in prolonging the survival of intestinal, skin, and heart allografts (48, 49). Another costimulatory pathway that might account for the recall of memory T cells is the OX40/OX40L pathway (50). A deficiency in or blockade of OX40 has been shown to lead to an impairment of memory CD4+ T cell formation and to prolong the survival of heart and skin allografts in recipients. Anti-OX40L monoclonal antibody prolongs the secondary cardiac allograft survival on the basis of CD40/CD40L and LFA-1/ICAM-1 blockade, with the anti-OX40L mAb impairing the generation of memory T cells and up-regulating IL-10-producing Tregs, thereby inhibiting T-cell function (51).

Depleting Memory T Cells

In addition to the strategies mentioned above, the most common therapeutic option for induction in transplantation is the administration of polyclonal antithymocyte globulin (ATG), which is produced in response to target thymocytes or T-cell lines (52, 53). Although ATG can effectively deplete alloreactive T cells, lymphopenia will induce compensatory proliferation that paradoxically supports memory T cells, favoring graft rejection and making it difficult to achieve or sustain operational tolerance of the graft (54, 55). However, several studies have shown that ATG may also play an important role in human Treg survival and expansion both in vitro and in vivo (56, 57). An increased ratio of Treg/T effector cells during ATG-induced homeostatic proliferation has been observed in rat kidney transplantation (58). Although memory T cells are much more resistant to depletion than are naïve T cells, Tregs proliferate to a significantly higher extent than do effector T cells, which suggests a biological preference of ATG for regulation rather than for promoting an effector immune function. In mice, it has been reported that pre-transplant administration of ATG results in increased efficacy in controlling donor-reactive memory T cells when compared to its peri-transplant administration (59). The application of ATG pre-transplantation results in a greater inhibition of pre-existing donor-reactive memory T-cell responses and a slower recovery of memory T cell counts than does peri-transplant treatment, making ATG-mediated depletion more efficient in prolonging allograft survival (59).

Some fundamental studies of the role of apoptosis in memory T cells have prompted us to target the apoptotic pathway to achieve functionally relevant depletion of memory T cells (58, 60). Regulation of the intrinsic apoptotic pathway by both pro- and antiapoptotic factors of the Bcl-2 family is critically important for the selection of T-cell clones for memory generation and for the maintenance of memory T cells. ABT-737, a small-molecule inhibitor of Bcl-2/Bcl-XL, efficiently induces apoptosis in alloreactive memory T cells in vitro and in vivo and prolongs skin graft survival in sensitized mouse recipients (26). Memory T-cell reduction produced by Bcl-2 inhibition seems to represent an important advance in the field of transplantation, which may benefit HLA-presensitized transplantation as well as tolerance induction.

Blocking Memory T/B Cells Interactions

As mentioned above, blocking interactions between memory T and B cells in transplantation may prevent not only alloantibody formation but also generation of long-lived memory T cells improving allograft survival. The short-term neutralization of BAFF alone or BAFF plus APRIL synergized with anti-CD154 monoclonal antibody was reported to prolong heart allograft survival in recipient mice containing donor-reactive memory CD4+ T cells, indicating that reagents neutralizing BAFF and APRIL might be used to enhance the efficacy of CD40/CD154 costimulatory blockade and improve allograft survival in T-cell-sensitized recipients (21). Another recent study reported that IFN-γ neutralization via blocking anti-IFN-γ antibody could prevent memory CD4+ T cells from providing CD40-independent help to B cells and thus improve allograft survival, which might be valuable for identifying transplant patients at risk for generation of de novo alloantibodies and for preventing alloantibody production in T-cell-sensitized recipients (22).

Conclusion

Despite their role in protective immunity against invading pathogens, the presence of memory T cells also threatens transplant survival. The unique characteristics of memory T cells in protective immunity against recurrent pathogens also make them formidable barriers to desired tolerance and successful transplantation. Studies in experimental models have suggested that targeting the infiltration, proliferation, activation, and intrinsic apoptotic pathway of memory T cells may be promising therapeutic approaches. However, with any therapy designed to inhibit memory T-cell recall responses, there is an inherent risk of impairing pathogen-specific protective immune responses. Therefore, the benefits in terms of graft survival obtained by attenuating memory T-cell responses must be carefully weighed against the cost of compromised protective immunity, the risk of infection of the host, and the potential for the development of malignancy. Recent evidence, however, has shown fundamental differences in the recall responses to pathogens and allografts of memory T cells that may offer a therapeutic window in which “detrimental” graft-specific recall responses can be attenuated, while “beneficial” pathogen-derived responses remain unaffected.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

We thank Dr. Deborah McClellan for editorial assistance.

References

1. Su CA, Fairchild RL. Memory T cells in transplantation. Curr Transplant Rep (2014) 1:137–46. doi:10.1007/s40472-014-0018-5

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Kaech SM, Cui W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat Rev Immunol (2012) 12:749–61. doi:10.1038/nri3307

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Sprent J, Tough DF. Lymphocyte life-span and memory. Science (1994) 265:1395–400. doi:10.1126/science.8073282

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Tchao NK, Turka LA. Lymphodepletion and homeostatic proliferation: implications for transplantation. Am J Transplant (2012) 12:1079–90. doi:10.1111/j.1600-6143.2012.04008.x

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Vu MD, Clarkson MR, Yagita H, Turka LA, Sayegh MH, Li XC. Critical, but conditional, role of OX40 in memory T cell-mediated rejection. J Immunol (2006) 176:1394–401. doi:10.4049/jimmunol.176.3.1394

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Yang J, Brook MO, Carvalho-Gaspar M, Zhang J, Ramon HE, Sayegh MH, et al. Allograft rejection mediated by memory T cells is resistant to regulation. Proc Natl Acad Sci U S A (2007) 104:19954–9. doi:10.1073/pnas.0704397104

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Sprent J, Surh CD. Generation and maintenance of memory T cells. Curr Opin Immunol (2001) 13:248–54. doi:10.1016/S0952-7915(00)00211-9

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Bingaman AW, Farber DL. Memory T cells in transplantation: generation, function, and potential role in rejection. Am J Transplant (2004) 4:846–52. doi:10.1111/j.1600-6143.2004.00453.x

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Lakkis FG, Sayegh MH. Memory T cells: a hurdle to immunologic tolerance. J Am Soc Nephrol (2003) 14:2402–10. doi:10.1097/01.ASN.0000085020.78117.70

CrossRef Full Text | Google Scholar

10. Chalasani G, Dai Z, Konieczny BT, Baddoura FK, Lakkis FG. Recall and propagation of allospecific memory T cells independent of secondary lymphoid organs. Proc Natl Acad Sci U S A (2002) 99:6175–80. doi:10.1073/pnas.092596999

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Donckier V, Craciun L, Miqueu P, Troisi RI, Lucidi V, Rogiers X, et al. Expansion of memory-type CD8+ T cells correlates with the failure of early immunosuppression withdrawal after cadaver liver transplantation using high-dose ATG induction and rapamycin. Transplantation (2013) 96:306–15. doi:10.1097/TP.0b013e3182985414

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Gerlach UA, Vogt K, Schlickeiser S, Meisel C, Streitz M, Kunkel D, et al. Elevation of CD4+ differentiated memory T cells is associated with acute cellular and antibody-mediated rejection after liver transplantation. Transplantation (2013) 95:1512–20. doi:10.1097/TP.0b013e318290de18

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Wu Z, Bensinger SJ, Zhang J, Chen C, Yuan X, Huang X, et al. Homeostatic proliferation is a barrier to transplantation tolerance. Nat Med (2004) 10:87–92. doi:10.1038/nm965

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Becattini S, Latorre D, Mele F, Foglierini M, De Gregorio C, Cassotta A, et al. T cell immunity. Functional heterogeneity of human memory CD4(+) T cell clones primed by pathogens or vaccines. Science (2015) 347:400–6. doi:10.1126/science.1260668

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Pantenburg B, Heinzel F, Das L, Heeger PS, Valujskikh A. T cells primed by Leishmania major infection cross-react with alloantigens and alter the course of allograft rejection. J Immunol (2002) 169:3686–93. doi:10.4049/jimmunol.169.7.3686

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Schenk AD, Nozaki T, Rabant M, Valujskikh A, Fairchild RL. Donor-reactive CD8 memory T cells infiltrate cardiac allografts within 24-h posttransplant in naive recipients. Am J Transplant (2008) 8:1652–61. doi:10.1111/j.1600-6143.2008.02302.x

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Su CA, Iida S, Abe T, Fairchild RL. Endogenous memory CD8 T cells directly mediate cardiac allograft rejection. Am J Transplant (2014) 14:568–79. doi:10.1111/ajt.12605

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Richards DM, Dalheimer SL, Hertz MI, Mueller DL. Trachea allograft class I molecules directly activate and retain CD8+ T cells that cause obliterative airways disease. J Immunol (2003) 171:6919–28. doi:10.4049/jimmunol.171.12.6919

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Kalache S, Lakhani P, Heeger PS. Effects of preexisting autoimmunity on heart graft prolongation after donor-specific transfusion and anti-CD154. Transplantation (2014) 97:12–9. doi:10.1097/TP.0b013e3182a77eba

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Rabant M, Gorbacheva V, Fan R, YU H, Valujskikh A. CD40-independent help by memory CD4 T cells induces pathogenic alloantibody but does not lead to long-lasting humoral immunity. Am J Transplant (2013) 13:2831–41. doi:10.1111/ajt.12432

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Gorbacheva V, Ayasoufi K, Fan R, Baldwin WM III, Valujskikh A. B cell activating factor (BAFF) and a proliferation inducing ligand (APRIL) mediate CD40-independent help by memory CD4 T cells. Am J Transplant (2015) 15:346–57. doi:10.1111/ajt.12984

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Gorbacheva V, Fan R, Wang X, Baldwin WM III, Fairchild RL, Valujskikh A. IFN-gamma production by memory helper T cells is required for CD40-independent alloantibody responses. J Immunol (2015) 194:1347–56. doi:10.4049/jimmunol.1401573

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Ng YH, Oberbarnscheidt MH, Chandramoorthy HC, Hoffman R, Chalasani G. B cells help alloreactive T cells differentiate into memory T cells. Am J Transplant (2010) 10:1970–80. doi:10.1111/j.1600-6143.2010.03223.x

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Sener A, Tang AL, Farber DL. Memory T-cell predominance following T-cell depletional therapy derives from homeostatic expansion of naive T cells. Am J Transplant (2009) 9:2615–23. doi:10.1111/j.1600-6143.2009.02820.x

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Valujskikh A. Targeting T-cell memory: where do we stand? Curr Opin Organ Transplant (2008) 13:344–9. doi:10.1097/MOT.0b013e3283061126

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Cippa PE, Gabriel SS, Kraus AK, Chen J, Wekerle T, Guimezanes A, et al. Bcl-2 inhibition to overcome memory cell barriers in transplantation. Am J Transplant (2014) 14:333–42. doi:10.1111/ajt.12554

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Ashokkumar C, Sun Q, Ningappa M, Higgs BW, Mazariegos G, Zeevi A, et al. Antithymocyte globulin facilitates alloreactive T-cell apoptosis by means of caspase-3: potential implications for monitoring rejection-free outcomes. Transplantation (2015) 99:164–70. doi:10.1097/TP.0000000000000289

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Kern F, Docke WD, Reinke P, Volk HD. Discordant expression of LFA-1, VLA-4alpha, VLA-beta 1, CD45RO and CD28 on T-cell subsets: evidence for multiple subsets of ‘memory’ T cells. Int Arch Allergy Immunol (1994) 104:17–26. doi:10.1159/000236704

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Mobley JL, Rigby SM, Dailey MO. Regulation of adhesion molecule expression by CD8 T cells in vivo. II. Expression of L-selectin (CD62L) by memory cytolytic T cells responding to minor histocompatibility antigens. J Immunol (1994) 153:5443–52.

PubMed Abstract | Google Scholar

30. Setoguchi K, Kishimoto H, Kobayashi S, Shimmura H, Ishida H, Toki D, et al. Potential role of host effector memory CD8+ T cells in marrow rejection after mixed chimerism induction in cynomolgus monkeys. Transpl Immunol (2010) 23:194–203. doi:10.1016/j.trim.2010.06.010

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Zhai Y, Meng L, Gao F, Busuttil RW, Kupiec-Weglinski JW. Allograft rejection by primed/memory CD8+ T cells is CD154 blockade resistant: therapeutic implications for sensitized transplant recipients. J Immunol (2002) 169:4667–73. doi:10.4049/jimmunol.169.8.4667

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Xia CQ, Chernatynskaya AV, Wasserfall CH, Wan S, Looney BM, Eisenbeis S, et al. Anti-thymocyte globulin (ATG) differentially depletes naive and memory T cells and permits memory-type regulatory T cells in nonobese diabetic mice. BMC Immunol (2012) 13:70. doi:10.1186/1471-2172-13-70

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Zheng J, Liu Y, Qin G, Chan PL, Mao H, Lam KT, et al. Efficient induction and expansion of human alloantigen-specific CD8 regulatory T cells from naive precursors by CD40-activated B cells. J Immunol (2009) 183:3742–50. doi:10.4049/jimmunol.0901329

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Riise RE, Bernson E, Aurelius J, Martner A, Pesce S, Della Chiesa M, et al. TLR-stimulated neutrophils instruct NK cells to trigger dendritic cell maturation and promote adaptive T cell responses. J Immunol (2015) 195:1121–8. doi:10.4049/jimmunol.1500709

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Boor PP, Metselaar HJ, Jonge S, Mancham S, Van Der Laan LJ, Kwekkeboom J. Human plasmacytoid dendritic cells induce CD8(+) LAG-3(+) Foxp3(+) CTLA-4(+) regulatory T cells that suppress allo-reactive memory T cells. Eur J Immunol (2011) 41:1663–74. doi:10.1002/eji.201041229

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Lei H, Kuchenbecker L, Streitz M, Sawitzki B, Vogt K, Landwehr-Kenzel S, et al. Human CD45RA FoxP3 memory-type regulatory T cells show distinct TCR repertoires with conventional T cells and play an important role in controlling early immune activation. Am J Transplant (2015). doi:10.1111/ajt.13315

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Zhang Q, Chen Y, Fairchild RL, Heeger PS, Valujskikh A. Lymphoid sequestration of alloreactive memory CD4 T cells promotes cardiac allograft survival. J Immunol (2006) 176:770–7. doi:10.4049/jimmunol.176.2.770

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Xia J, Chen J, Shao W, Lan T, Wang Y, Xie B, et al. Suppressing memory T cell activation induces islet allograft tolerance in alloantigen-primed mice. Transpl Int (2010) 23:1154–63. doi:10.1111/j.1432-2277.2010.01106.x

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Carvalho-Gaspar M, Jones ND, Luo S, Martin L, Brook MO, Wood KJ. Location and time-dependent control of rejection by regulatory T cells culminates in a failure to generate memory T cells. J Immunol (2008) 180:6640–8. doi:10.4049/jimmunol.180.10.6640

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Kwun J, Farris AB, Song H, Mahle WT, Burlingham WJ, Knechtle SJ. Impact of leukocyte function-associated antigen-1 blockade on endogenous allospecific T cells to multiple minor histocompatibility antigen mismatched cardiac allograft. Transplantation (2015). doi:10.1097/TP.0000000000000805

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Yabu JM, Vincenti F. Novel immunosuppression: small molecules and biologics. Semin Nephrol (2007) 27:479–86. doi:10.1016/j.semnephrol.2007.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Wang T, Alam R, Langley KE, Klimpel GR. Stem cell factor and IL-2 act synergistically in inducing intraepithelial lymphocyte proliferation and cytokine production: upregulation of the IL-2 receptor gamma-chain and signaling via JAK-3. Cell Immunol (2000) 205:62–71. doi:10.1006/cimm.2000.1707

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Paniagua R, SI MS, Flores MG, Rousvoal G, Zhang S, Aalami O, et al. Effects of JAK3 inhibition with CP-690,550 on immune cell populations and their functions in nonhuman primate recipients of kidney allografts. Transplantation (2005) 80:1283–92. doi:10.1097/01.tp.0000177643.05739.cd

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Cismasiu VB, Duque J, Paskaleva E, Califano D, Ghanta S, Young HA, et al. BCL11B enhances TCR/CD28-triggered NF-kappaB activation through up-regulation of Cot kinase gene expression in T-lymphocytes. Biochem J (2009) 417:457–66. doi:10.1042/BJ20080925

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Molinero LL, Zhou P, Wang Y, Harlin H, Kee B, Abraham C, et al. Epidermal Langerhans cells promote skin allograft rejection in mice with NF-kappa B-impaired T cells. Am J Transplant (2008) 8:21–31. doi:10.1111/j.1600-6143.2007.02038.x

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Kim JS, Lee JI, Shin JY, Kim SY, Shin JS, Lim JH, et al. Bortezomib can suppress activation of rapamycin-resistant memory T cells without affecting regulatory T-cell viability in non-human primates. Transplantation (2009) 88:1349–59. doi:10.1097/TP.0b013e3181bd7b3a

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Habib-Agahi M, Jaberipour M, Searle PF. 4-1BBL costimulation retrieves CD28 expression in activated T cells. Cell Immunol (2009) 256:39–46. doi:10.1016/j.cellimm.2009.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Cho HR, Kwon B, Yagita H, LA S, Lee EA, Kim JE, et al. Blockade of 4-1BB (CD137)/4-1BB ligand interactions increases allograft survival. Transpl Int (2004) 17:351–61. doi:10.1007/s00147-004-0726-3

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Qin L, Guan HG, Zhou XJ, Yin J, Lan J, Qian HX. Blockade of 4-1BB/4-1BB ligand interactions prevents acute rejection in rat liver transplantation. Chin Med J (Engl) (2010) 123:212–5. doi:10.3760/cma.j.issn.0366-6999.2010.02.017

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Wang GM, Yang Y, Jin YZ, LI AL, Hao J, Gao X, et al. Blockade of both CD28/B7 and OX40/OX40L co-stimulatory signal pathways prolongs the survival of islet xenografts. Transplant Proc (2005) 37:4449–51. doi:10.1016/j.transproceed.2005.10.117

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Dai H, Peng F, Lin M, Xia J, YU S, Lan G, et al. Anti-OX40L monoclonal antibody prolongs secondary heart allograft survival based on CD40/CD40L and LFA-1/ICAM-1 blockade. Transpl Immunol (2015) 32:84–91. doi:10.1016/j.trim.2015.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Mohty M. Mechanisms of action of antithymocyte globulin: T-cell depletion and beyond. Leukemia (2007) 21:1387–94. doi:10.1038/sj.leu.2404683

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Gaber AO, Monaco AP, Russell JA, Lebranchu Y, Mohty M. Rabbit antithymocyte globulin (thymoglobulin): 25 years and new frontiers in solid organ transplantation and haematology. Drugs (2010) 70:691–732. doi:10.2165/11315940-000000000-00000

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Moxham VF, Karegli J, Phillips RE, Brown KL, Tapmeier TT, Hangartner R, et al. Homeostatic proliferation of lymphocytes results in augmented memory-like function and accelerated allograft rejection. J Immunol (2008) 180:3910–8. doi:10.4049/jimmunol.180.6.3910

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Neujahr DC, Chen C, Huang X, Markmann JF, Cobbold S, Waldmann H, et al. Accelerated memory cell homeostasis during T cell depletion and approaches to overcome it. J Immunol (2006) 176:4632–9. doi:10.4049/jimmunol.176.8.4632

PubMed Abstract | CrossRef Full Text | Google Scholar

56. LaCorcia G, Swistak M, Lawendowski C, Duan S, Weeden T, Nahill S, et al. Polyclonal rabbit antithymocyte globulin exhibits consistent immunosuppressive capabilities beyond cell depletion. Transplantation (2009) 87:966–74. doi:10.1097/TP.0b013e31819c84b8

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Broady R, YU J, Levings MK. ATG-induced expression of FOXP3 in human CD4(+) T cells in vitro is associated with T-cell activation and not the induction of FOXP3(+) T regulatory cells. Blood (2009) 114:5003–6. doi:10.1182/blood-2009-04-214437

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Valdez-Ortiz R, Bestard O, Llaudo I, Franquesa M, Cerezo G, Torras J, et al. Induction of suppressive allogeneic regulatory T cells via rabbit antithymocyte polyclonal globulin during homeostatic proliferation in rat kidney transplantation. Transpl Int (2015) 28:108–19. doi:10.1111/tri.12448

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Ayasoufi K, YU H, Fan R, Wang X, Williams J, Valujskikh A. Pretransplant antithymocyte globulin has increased efficacy in controlling donor-reactive memory T cells in mice. Am J Transplant (2013) 13:589–99. doi:10.1111/ajt.12068

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Neff KS, Richards SM, Williams JM, Garman RD, Ruzek MC. Murine antithymocyte globulin T-cell depletion is mediated predominantly by macrophages, but the Fas/FasL pathway selectively targets regulatory T cells. Transplantation (2011) 92:523–8. doi:10.1097/TP.0b013e31822923f7

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: memory T cells, transplantation, allograft rejection, tolerance, therapy

Citation: Lin K, Chen S and Chen G (2015) Role of memory T cells and perspectives for intervention in organ transplantation. Front. Immunol. 6:473. doi: 10.3389/fimmu.2015.00473

Received: 11 July 2015; Accepted: 31 August 2015;
Published: 14 September 2015

Edited by:

Zhenhua Dai, Guangdong Provincial Academy of Chinese Medical Sciences, China

Reviewed by:

Alain Le Moine, Université Libre de Bruxelles, Belgium
Stanislaw Stepkowski, University of Toledo College of Medicine, USA

Copyright: © 2015 Lin, Chen and Chen. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Gang Chen, Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China, gchen@tjh.tjmu.edu.cn

Kailin Lin and Song Chen have contributed equally to this work.

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.