<<

REVIEW J Am Soc Nephrol 14: 2402–2410, 2003

Memory T Cells: A Hurdle to Immunologic Tolerance

FADI G. LAKKIS* and MOHAMED H. SAYEGH† *Section of Nephrology, Department of Internal Medicine, and Section of Immunobiology, Yale University School of Medicine, New Haven, Connecticut; and †Renal Division, Brigham and Women’s Hospital, and Nephrology Division, Children’s Hospital, Harvard Medical School, Boston, Massachusetts.

The induction of immunologic tolerance is an important Immunobiology of Memory T Cells clinical goal in transplantation and autoimmunity. Immuno- Upon exposure to a foreign , antigen-specific T logic tolerance is traditionally defined as specific unrespon- cells proliferate (the expansion phase) and differentiate into siveness to a self- or foreign antigen while maintaining reac- effectors that eliminate the foreign intruder (Figure 1). The tivity to other (third party) (1,2). In the context of vast majority of effector T cells, however, undergo apopto- transplantation, a tolerant patient is someone who is capable of sis as the progresses (the death phase), mounting an effective immune response to vaccines and mi- and the few that survive become long-lived crobial pathogens but is incapable of rejecting the transplanted memory T cells (the memory phase) (8,9). Memory T cells organ. Similarly, induction of tolerance to an antigen that that recognize microbial antigens provide the organism with incites self-reactivity would ensure that the patient is free of long-lasting protection against potentially fatal . autoimmune manifestations in the absence of global and harm- Conversely, memory T cells that recognize donor alloanti- ful immunosuppression. Although several immunomodulatory gens jeopardize the survival of life-saving organ transplants strategies have been used successfully to induce immunologic by mediating rejection (10). Therefore, the central question in the pursuit of transplantation tolerance is how to coerce tolerance in rodents, the same strategies have failed in larger an immune response determined to generate memory animals and in humans. Examples of induced tolerance in into a state of antigen-specific unresponsiveness. To begin organ transplant recipients or in patients with autoimmune to answer this question, we first highlight the general char- disease have been rare and often unintentional (3–6). acteristics of memory T cells that distinguish them from Why, then, has clinical tolerance been such an elusive goal? naïve T cells. Second, we summarize what is known about The answer to this question most likely lies in the immunologic the generation, maintenance, and activation (recall) of mem- barriers that preclude the induction of antigen-specific unre- ory T cells. sponsiveness in the adult animal (1,2). These barriers include the limitations of peripheral (extrathymic) immunoregulatory Memory Advantage mechanisms that are commonly exploited to induce tolerance Memory T cells have over their naïve counterparts several (T cell deletion, suppression, deviation, and anergy), the large inherent advantages that endow them with the ability to clear a repertoire of alloreactive T cells in the case of transplantation, foreign antigen, whether a microbial pathogen or an allograft, the uncertain nature of the pathogenic antigen in many auto- in a vigorous manner (Table 1). The first advantage that immune diseases, and the unavoidable fact that the adaptive memory T cells have is that their response to a foreign antigen immune response, by virtue of evolutionary design, is destined (recall response) is greater in magnitude and faster than the to generate immunologic memory. It is the last barrier that is naïve T cell response. Memory T cells generate a considerable perhaps the most important obstacle to immunologic tolerance. number of effector T cells, capable of secretion and/or Here, we address this thesis by reviewing the immunobiology cytolytic activity, within hours of antigenic restimulation, of memory T cells and their impact on solid organ transplan- whereas naïve T cells generate a smaller number of effectors tation. The same concepts that apply to understanding and and at a much slower pace (days) (11–14). Second, memory T controlling memory T cells in the context of transplantation are cells have a survival advantage over their naïve counterparts also relevant to autoimmunity (7). (15,16). Antigen-specific populations persist for years to a lifetime in humans, and their survival seems to be antigen and MHC independent (17–19). Mature naïve T cell populations also persist for a relatively long period of time (months to a few years in humans), but their survival is Correspondence to Dr. Fadi G. Lakkis, Section of Nephrology, 333 Cedar Street, P.O. Box 208029, New Haven, CT 06520-8029. Phone: 203-737-2619; dependent on constant, low-grade stimulation with MHC–self- Fax: 203-737-1810; E-mail: [email protected] peptide complexes (20–22). Third, the circulation of naïve T 1046-6673/1409-2402 cells is restricted to secondary lymphoid tissues, the site where Journal of the American Society of Nephrology they are activated by foreign antigens presented by antigen- Copyright © 2003 by the American Society of Nephrology presenting cells (APC), whereas memory T cells circulate DOI: 10.1097/01.ASN.0000085020.78117.70 through both secondary lymphoid tissues and peripheral non- J Am Soc Nephrol 14: 2402Ð2410, 2003 Memory T Cells 2403

Figure 1. Foreign antigen triggers the proliferation of antigen-specific T cells (the expansion phase) and their differentiation into effectors. The vast majority of effector T cells undergo (the death phase) as the foreign antigen is eliminated, and the few lymphocytes that survive become long-lived memory T cells (the memory phase). Immunologic tolerance can perhaps be viewed as the demise of antigen-specific memory lymphocytes or their precursors (red arrows).

Table 1. Advantages of memory T cells over naõ¬ve and effector T cellsa

Characteristic Naõ¬ve T Cell Effector T Cell Memory T Cell

Survival Months to years Hours to days Years to lifetime Dependent on TCR Ag persistence leads Not dependent on TCR interaction with MHC/Ag to AICD interaction with MHC/Ag Migration Secondary lymphoid tissues Nonlymphoid tissues Both lymphoid and nonlymphoid tissues Response and effector Requires stimulation with Immediate Requires restimulation with function Ag Ag but is faster and larger in magnitude than naõ¬ve T cells In vivo protection against Long-lived but relatively Short-lived but very Long-lived and efficient foreign Ag inefficient efficient

a AICD, activation-induced cell death; Ag, antigen; TCR, T cell receptor for antigen. lymphoid tissues (23–25). Unlike naïve T cells, memory T pated. An antigen-experienced animal will eliminate the for- cells can directly encounter foreign antigen and mount a pro- eign antigen, whether a virus or an allograft, more efficiently ductive immune response within nonlymphoid tissues (25). than a naïve animal. The migratory advantage of memory T cells, therefore, allows them to detect and eliminate a foreign intruder long before it Generation of Memory T Cells reaches secondary lymphoid tissues. In summary, the activa- Antigenic stimulation of naïve T cells is a prerequisite for tion, survival, and migration advantages of memory T cells memory generation (26,27). It is also established that the confer the organism with enhanced protection against foreign magnitude of the expansion phase (burst size) of the primary antigens long after the primary immune response has dissi- immune response determines the size of the memory T cell 2404 Journal of the American Society of Nephrology J Am Soc Nephrol 14: 2402Ð2410, 2003 pool (28,29). strategies that enhance T cell acti- naïve T cells that received suboptimal antigenic stimulation vation and proliferation by using strong adjuvants lead to larger and failed to proliferate. Importantly, it is not known how memory T cell populations. Because adjuvants directly influ- activated/effector T cells transition to the memory phenotype ence APC maturation and function, it is generally believed that (which is relatively quiescent and resembles that of a naïve T factors that stimulate the innate immune response, for example cell) and whether this transition is a stochastic process or an by inducing IFN-1 production or engaging Toll-like receptors instructional one (51). on APC (30,31), result in stronger adaptive immune responses Finally, recent studies propose the existence of two subsets and, subsequently, more memory T cells. Allografts are potent of CD4ϩ and CD8ϩ memory T cells based on the expression adjuvants as they contain their own APC and are subjected to of the homing receptors CC-chemokine receptor 7 (CCR7) and ischemia-reperfusion injury at the time of transplantation, L-selectin (CD62L) (56). The CCR7ϩ CD62Lhi “central” Ϫ lo which leads to activation of the innate (32). In memory T cells (TCM) and CCR7 CD62L “effector” mem- fact, signaling via Toll-like receptors on graft dendritic cells ory T cells (TEM) have distinct migratory and functional char- plays an important role in initiating the alloimmune response acteristics (56Ð58). TCM circulate through secondary lymphoid ␥ (33). Therefore, it should not come as a surprise that transplant tissues, produce IL-2 but little IFN- and no perforin. TEM recipients harbor significant numbers of alloreactive memory T circulate through nonlymphoid peripheral tissues and produce and B cells. IFN-␥, perforin, and IL-4 but little IL-2. It has been proposed

Because optimal T cell activation and proliferation require that the secondary lymphoid tissue resident TCM are responsi- the presence of adequate T cell co-stimulation, several co- ble for replenishing the memory T cell pool because of their stimulatory molecule pairs contribute to the generation of increased proliferative capacity, whereas the nonlymphoid tis- ϩ memory T cells. CD4 T cell proliferation and memory gen- sue resident TEM mediate effector functions and rapid elimi- eration are significantly impaired but not completely absent in nation of antigen (23,24,56). The origin of these memory mice deficient in CD28, CD40L (CD154), or OX40 (34Ð36). subpopulations and whether they are interrelated are not com- ϩ ϩ However, generation of CD8 memory T cells is reduced in pletely understood. One study suggested that CD8 TEM de- mice that lack 4-1BBL, CD40L, CD27 (CD70L), and, to a scend from fully differentiated effectors, whereas TCM arise lesser extent, CD28 (37Ð41). Therefore, redundancy in T cell from intermediates that have not yet acquired effector function co-stimulation ensures that memory generation is not abolished (54). In contrast, Ahmed and colleagues (59) showed in a in the absence of a single co-stimulatory pathway. Likewise, no murine viral model that both subsets in the CD8ϩ single cytokine deficiency blocks memory T cell generation. compartment arise from fully differentiated effector T cells and ϩ This is particularly true of the CD4 population (42), whereas that TEM slowly convert over time to the TCM phenotype. the absence of IL-2 or IL-15 reduces the number of CD8ϩ T cells generated during the primary immune response (43Ð45). Maintenance of Memory T Cells Recent studies provide evidence that generation of CD8ϩ Memory T cell populations persist for a very long period of memory T cells is dependent on signals provided by CD4ϩ T time, often for the lifetime of an individual, in the absence of cells at an early time point in the primary immune response repeated antigenic exposure. At least two mechanisms are (46Ð48). Although the exact nature of these signals is still responsible for the long-term maintenance of memory T cell unclear, it is possible that CD4ϩ T cells condition the APC via populations: constant turnover (antigen-independent homeo- CD40LÐCD40 interactions to provide optimal CD8ϩ T cell static proliferation) and the intrinsic ability of memory T cells triggering (49). to survive in a relatively resting state for an extended duration Which activated T cells constitute the precursors of memory (19,43,60Ð63). The homeostatic proliferation of memory T T cells remains a subject of controversy. Some investigators cells is much slower than antigen-driven prolifer- have provided evidence that memory T cells are direct descen- ation but is considerably faster than the homeostatic prolifer- dants of effector T cells, whereas others contend that they arise ation of naïve T cells (43,60,62,63). Homeostatic proliferation from a second lineage of activated T cells, perhaps an “inter- of naïve T cells is what maintains the mature T cell pools in the mediate” cell population (8,50,51). Recent studies of CD8ϩ periphery even in the absence of thymic function (22). In a virus-specific memory T cells suggest a linear model of mem- viral infection mouse model, the CD8ϩ virus-specific memory ory T cell differentiation in which a naïve T cell is committed population turns over approximately once every 35 d to become an effector and, later, a memory T cell, shortly after (18,64,65). IL-15 is the central driver of memory T cell turn- antigenic stimulation (26,52,53). However, others have argued over in the CD8ϩ compartment (66,67), whereas IL-2 inhibits that effector differentiation is not required for the generation of homeostatic proliferation (43,63). Conversely, IL-7, seems to CD8ϩ memory T cells (54). Similarly, evidence has been be responsible for conferring a survival advantage to CD8ϩ presented to support either the linear differentiation of CD4ϩ memory T cells by upregulating antiapoptotic molecules such memory T cells from polarized effectors or their derivation as Bcl-2 and Bcl-XL (68). It is unclear what maintains CD4ϩ from a separate lineage of activated T cells with a nonpolar- T cell memory populations. Because long-lived CD4ϩ mem- ized, intermediate phenotype (27,55). Irrespective of which ory T cells can be generated from common cytokine receptor differentiation pathway is operational, memory T cells arise ␥-chain (␥c) knockout precursors, it seems that ␥c-binding from antigen-activated lymphocytes that proliferated during (IL-2, -4, -7, -9, and -15) are not required for either the expansion phase of the immune response and not from the generation or the maintenance of this memory T cell J Am Soc Nephrol 14: 2402Ð2410, 2003 Memory T Cells 2405 population (42). Finally, it is not known whether the rules that lose the ability to migrate to secondary lymphoid tissues as apply to the maintenance of the overall memory T cell popu- they shed the CCR7 and CD62L (also known as L-selectin) and lation also apply to the maintenance of the “effector” and acquire adhesion molecules and tissue-specific chemokine re- “central” memory subsets. ceptors that direct them to nonlymphoid tissue microenviron- ␣ ments (e.g., to skin: CCR8, CCR10, CLA; to gut mucosa: 4␤7 Recall of Memory T Cells ; and to sites of : CCR5) (23,24,81Ð83). When restimulated with antigen, memory T cells multiply Homing receptor expression depends on the route of antigen and differentiate into effector T cells much more rapidly than exposure during the primary immune response with preferen- their naïve counterparts. It is estimated that the average time tial migration of memory T cells back to the site of initial that it takes a virus-specific CD8ϩ memory T cell to differen- antigen entry (84,85). Both CD4ϩ and CD8ϩ memory T cells tiate into a cytotoxic T lymphocyte is 12 h as opposed to can be isolated from nonlymphoid tissues even in the absence several days for a naïve CD8ϩ T cell (Rafi Ahmed, Emory of inflammation (23,24), suggesting that memory T cells cir- University, personal communication, June 2001). In addition, culate through these tissues. More recently, it has been dem- the number of effector T cells generated during a recall re- onstrated that, unlike naïve T cells, memory T cells mount a sponse exceeds the number generated during a primary im- productive immune response independent of secondary lym- mune response by approximately fivefold (53). The vigor of phoid tissues. In a murine transplantation model, as little as a the recall response can be attributed to the increased frequency few thousand alloreactive memory T cells migrated directly to of antigen-specific CD4ϩ and CD8ϩ T cells in the memory the allograft and mediated its rejection in an immunodeficient population (28,69Ð71) and to the intrinsic antigen hyperre- host that lacks the spleen, lymph nodes, and mucosal lymphoid sponsiveness of memory T cells (11,12,71Ð73). The antigen structures (25). This observation applies to both CD4ϩ and hyperresponsiveness of memory T cells is best exemplified by CD8ϩ memory T cell recall. Therefore, the migratory advan- the short lag time to cytokine production, entering cell cycle, tage of memory T cells allows them to detect and eliminate a and acquisition of effector function after antigen stimulation. foreign antigen long before it reaches secondary lymphoid ϩ In fact, CD8 effector memory T cells (TEM) may have tissues. In contrast, naïve T cells are activated only after constitutive cytolytic function (56), effectively reducing lag antigen-bearing dendritic cells have reached the draining time between antigen encounter and effector function to zero. lymph nodes or the spleen, a process that could take at least The mechanisms responsible for the antigen-hyperrespon- 2d. siveness of memory T cells are not completely understood but may include enhanced signaling via the T cell receptor (TCR) Memory T Cells and Allograft Rejection complex. For example, plasma membrane-associated , a The same characteristics of memory T cells that make them protein kinase that phosphorylates CD3 and the CD3-associ- very efficient at eliminating microbial pathogens also enable ated ␨ chain leading to enhanced TCR signaling, has been them to rapidly reject foreign tissues. Unlike inbred mice, observed in memory T cells (74,75). Gene expression profiling outbred animals including humans harbor a significant number experiments confirmed that several gene products associated of memory T cells that are alloreactive. These memory T cells with TCR signaling are elevated in memory T cells compared generally arise if an individual is exposed to alloantigens via with naïve T cells (76). In addition, memory T cells may be pregnancy, transfusion, or a previous organ transplant. hyperresponsive to antigen because of affinity maturation, a However, alloreactive memory T cells also exist in individuals process in which T cells bearing TCR with high affinity to who have never been exposed to foreign tissues before (10). It antigen are selected during the immune response (9,77). Re- is proposed that such alloreactivity in the T cell memory pool duced activation threshold of memory T cells via the TCR is caused largely by cross-reactivity; in other words, exposure complex is consistent with mounting evidence that the activa- to viral and bacterial antigens over time leads to the develop- tion of memory T cells is less dependent on co-stimulation than ment of memory T cells that also recognize alloantigens. This naïve T cells (74,78Ð80). Normal cytokine production is ob- phenomenon, commonly referred to as heterologous , served when ovalbumin-specific TCR-transgenic CD4ϩ mem- has been demonstrated in murine infection models (86,87) and ory T cells are stimulated with antigen-loaded APC that lack is supported by experimental findings in humans (88). either B7 or CD40 (80). Moreover, the proliferation of these Several investigators have provided evidence that alloreac- CD4ϩ memory T cells is normal in the absence of CD40 and tive memory T cells indeed contribute to both acute and is only slightly impaired in the absence of B7 (80). Antigen- chronic allograft rejection. Heeger et al. (10) found that the specific CD8ϩ memory T cells confer normal protective im- pretransplant frequency of donor-specific memory T cells, munity in the absence of CD28 (41). Similarly, CD40ÐCD40L identified by their ability to produce IFN-␥ within 24 h of co-stimulation is dispensable for the recall of alloreactive allostimulation, correlates with the risk of rejection after trans- CD8ϩ memory T cells (35). Recent work suggests that CD27Ð plantation. Others have observed that the presence of memory- CD70 interaction may be important for the recall of CD8ϩ phenotype T cells (CD45ROϩ) in heart and kidney allograft memory T cells (39). biopsies and in the peripheral blood of transplant recipients Another important advantage of memory T cells over naïve correlates with the incidence and severity of rejection (89Ð92). T cells is their ability to seek antigen in nonlymphoid tissues. This was also true in patients experiencing acute rejection Memory T cells, particularly those of the “effector” phenotype, episodes 2 to 18 yr after transplantation (93). In addition, the 2406 Journal of the American Society of Nephrology J Am Soc Nephrol 14: 2402Ð2410, 2003 frequency of IFN-␥Ðproducing peripheral blood lymphocytes, of cardiac allograft survival induced through a combination of possibly memory T cells, responding to donor-derived peptides anti-CD40L monoclonal treatment and donor spleen via the indirect antigen presentation pathway are more fre- cell transfusion. The same therapeutic strategy also failed to quently found in renal transplant recipients who previously prolong allograft survival or to induce tolerance in mice har- experienced acute rejection episodes than in those without boring cross-reactive memory T cells (e.g., mice previously rejection (94). Although these studies do not establish a causeÐ infected with lymphocytic choriomeningitis virus or Leishma- effect relationship between T cell memory and rejection, they nia major) (86,87). A recent manuscript provided direct evi- suggest that the persistence of memory T cells hinders long- dence that virally induced alloreactive memory T cells interfere term allograft survival in humans. Moreover, donor-reactive T with the most robust form of transplantation tolerance, that cells with memory phenotype have been detected in organ induced by mixed allogeneic chimerism (104). In this study transplant recipients receiving high levels of immunosuppres- (104), CD8ϩ “central” memory T cells were responsible for sion (10,94), indicating that current immunosuppressive ther- preventing tolerance, and they did so in a dose-dependent apies do not inhibit the generation or maintenance of T cell manner. Therefore, alloreactive T cells, whether generated by memory. previous encounter to alloantigens or via heterologous immu- The existence of allospecific memory T cells in experimen- nity, constitute a formidable barrier to achieving transplanta- tal models of alloimmunity was first suggested by the finding tion tolerance. Alloreactive memory T cells generated as a that spleen cells from allosensitized mice mount a stronger result of heterologous immunity may very well offer an expla- mixed leukocyte response and generate more cytotoxic effec- nation for why transplantation tolerance has been such an tors than naïve splenocytes (95). More recently, TCR-trans- elusive goal in human and nonhuman primates that have a rich genic models have been used to identify allospecific CD8ϩ immunologic history of exposure to environmental and micro- memory T cells generated after cardiac allograft rejection in bial antigens. mice (96). Memory T cells mediate an accelerated form of rejection as indicated by earlier demonstration that mice that Repressing Undesirable Memories have previously rejected an allograft reject a second graft from The success of experimental tolerance strategies has been the same donor with accelerated kinetics, a phenomenon re- largely restricted to naïve hosts that do not harbor significant ferred to as second set rejection (97). Furthermore, purified numbers of memory T cells, for example, mice housed in a memory T cell populations, unlike their naïve counterparts, pathogen-free environment (1). These strategies induce immu- cause allograft rejection without the need to home first to nologic tolerance to a foreign antigen by exploiting the same secondary lymphoid tissues (25), and the rejection response is principles that underlie tolerance to self-antigens: deletion, not inhibited by agents that block CD28 or CD40L (CD154) T anergy, suppression, and immune deviation (105). Deletion is cell co-stimulation (Sayegh et al., unpublished data). These mediated by the apoptosis of antigen-specific naïve or recently findings underscore the formidable challenge to prevent suc- activated T cells leading to antigen-specific unresponsiveness. cessfully the recall of alloreactive memory T cells. Alternatively, a tolerance-inducing strategy could inactivate T cells without causing their death (anergy), generate regulatory Memory T Cells and Transplantation Tolerance cells that block T cell activation and function (suppression), or Memory T cells not only endanger allograft survival by induce the differentiation of naïve T cells into a nonharmful causing both acute and chronic rejection but also impede the phenotype (immune deviation). Therefore, for tolerance to induction of transplantation tolerance. Several rodent studies work in immunologically experienced hosts, such as humans, provide indirect evidence in support of this observation. Re- one first has to ask whether memory T cells can be “tolerized.” sistance to neonatal tolerance in certain mouse strains is asso- In other words, are memory T cells subject to deletion, anergy, ciated with the persistence of memory-type T cells (98), suppression, or immune deviation in the same way that naïve T whereas successfully tolerized mice have much lower frequen- cells are? cies of these cells (99,100). Moreover, immunomodulatory The answers to these questions are beginning to emerge, and agents that do not consistently induce donor-specific tolerance the initial data are encouraging. First, CD8ϩ memory T cells invariably fail to suppress immunologic memory in experimen- seem to be as susceptible to tolerance as their naïve counter- tal animals (100,101). parts. Kreuwell et al. (106) demonstrated that influenza hem- Direct evidence that memory T cells impede tolerance in- agglutinin A (HA)-specific CD8ϩ memory T cells undergo duction derives from studies using co-stimulation blockade peripheral tolerance after either exogenous administration of and/or mixed allogeneic chimerism strategies. Zhai et al. (102) soluble HA peptide or cross-presentation of the HA antigen by found that anti-CD40L (CD154) monoclonal antibody therapy, dendritic cells in transgenic mice that produce HA in the which induces transplantation tolerance in some rodent strain pancreatic ␤ cells (the latter phenomenon is referred to as combinations, fails to do so in mice presensitized with skin cross-tolerance). Tolerance of CD8ϩ memory T cells to the grafts from the same donor. They further demonstrated that, HA antigen was mediated by peripheral clonal deletion. Sec- unlike naïve T cells, T cells from presensitized mice can be ond, although memory T cells are less susceptible to apoptosis activated in vitro in the absence of CD40ÐCD40L co-stimula- than naïve T cells, they can still be coerced to die. Memory tion. Valusjkikh et al. (103) found that the adoptive transfer of CD8ϩ T cells that migrate to immune-privileged sites undergo primed donor-reactive T cells interferes with the prolongation apoptosis mediated by the TNF receptor (TNFR) family (Dai, J Am Soc Nephrol 14: 2402Ð2410, 2003 Memory T Cells 2407

Lakkis, et al., unpublished observations). Third, memory T immunologic tolerance would perhaps become a reality if we cells are subject to suppression by regulatory T cells. were to master the art of repressing undesirable memories CD4ϩCD25ϩ T cells suppress in vivo CD8ϩ memory T cell while keeping the sweet and desirable ones alive. responses to Listeria monocytogenes (107), and, in an adoptive transfer model, they significantly delay allograft rejection me- diated by CD8ϩ memory T cells (Dai and Lakkis, unpublished References observations). Fourth, memory T cells are not rigidly differen- 1. Salama A, Remuzzi G, Harmon W, Sayegh M: Challenges to achieving clinical transplantation tolerance. J Clin Invest 108: tiated and may be amenable to immune deviation. Ahmadza- ϩ 943Ð948, 2001 deh and Farber (108) observed that antigen-specific CD4 2. Chalasani G, Lakkis, FG: Immunologic ignorance of organ al- memory T cells produce either T helper 1 or T helper 2 effector lografts. Curr Opin Organ Transplant 6: 83Ð88, 2001 cytokines depending on the nature of the recall stimulus, indi- 3. Strober S, Dhillon M, Schubert M, Holm B, Engleman E, Benike cating that the memory T cells, like naïve T cells, retain C, Hoppe R, Sibley R, Myburgh J, Collins G, et al.: Acquired functional plasticity. Therefore, memory T cells can be poten- to cadaveric renal allografts. A study of three tially controlled via the same mechanisms that mediate naïve T patients treated with total lymphoid irradiation. N Engl J Med cell tolerance. 321: 28Ð33, 1989 An alternative approach to targeting memory is to block 4. Sayegh M, Fine N, Smith J, Rennke H, Milford E, Tilney N: activation pathways unique to the recall of memory T cells. Immunologic tolerance to renal allografts after bone marrow Recent studies have identified co-stimulatory pathways that are transplants from the same donors. Ann Intern Med 114: 954Ð955, 1991 involved in the activation of antigen-experienced but not naïve 5. Spitzer T, Delmonico F, Tolkoff-Rubin N, McAfee S, Sackstein T cells. One such pathway is the interaction of inducible R, Saidman S, Colby C, Sykes M, Sachs D, Cosimi A: Combined costimulator (ICOS) on T cells with B7h (B7RP-1) on antigen- histocompatibility leukocyte-antigen matched donor bone mar- presenting cells (109). Co-stimulation through ICOS is crucial row and renal transplantation for multiple myeloma with end for the activation of previously primed T cell populations that stage renal disease: The induction of allograft tolerance through include memory T cells. Blocking the B7h-ICOS costimulation mixed lymphohematopoietic chimerism. Transplantation 68: pathway prolongs allograft survival in rodents, particularly if 480Ð484, 1999 the blockade is administered at a delayed time point after 6. VanBuskirk A, Burlingham W, Jankowska-Gan E, Chin T, transplantation, suggesting that it interferes with the recall of Kusaka S, Geisller F, Pelletier R, Orosz C: Human allograft primed T cells (110). Agents that target the interaction of ICOS acceptance is associated with immune regulation. J Clin Invest with B7h have also been shown to prevent chronic rejection 106: 145Ð155, 2000 7. Kuchroo V, Anderson A, Waldner H, Munder M, Bettelli E, and to facilitate tolerance induction when combined with other Nicholson L: T cell response in experimental autoimmune en- immunosuppressive modalities (111). Another co-stimulatory cephalomyelitis (EAE): Role of self and cross-reactive antigens pathway that may be important for the recall of memory T in shaping, tuning, and regulating the autopathogenic T cell cells, particularly the CD4ϩ population, is the OX40 repertoire. Ann Rev Immunol 20: 101Ð123, 2002 (CD134)ÐOX40L (CD134L) pathway. In transplantation mod- 8. Kaech SM, Wherry EJ, Ahmed R: Effector and memory T cell els, simultaneous blockade of OX40L and B7 prolongs cardiac differentiation: Implications for vaccine development. Nat Rev allograft survival in recipients that had received primed T cells Immunol 2: 251Ð262, 2002 (112). Finally, preliminary data suggest that blocking the 9. Sprent J, Surh C: T cell memory. Annu Rev Immunol 20: 551Ð CD70ÐCD27 (CD70L) pathway inhibits allograft rejection 579, 2002 mediated by CD8ϩ memory T cells (Sayegh et al., unpub- 10. Heeger PS, Greenspan NS, Kuhlenschmidt S, Dejelo C, Hricik lished observations). This finding is consistent with the role of DE, Schulak JA, Tary-Lehmann M: Pretransplant frequency of donor-specific, IFN-␥-producing lymphocytes is a manifestation CD70ÐCD27 co-stimulation in the activation of antigen-expe- ϩ of immunologic memory and correlates with the risk of post- rienced CD8 T cells (39). transplant rejection episodes. J Immunol 163: 2267Ð2275, 1999 Finally, controlling T cell memory responses does not only 11. Rogers PR, Dubey C, Swain SL: Qualitative changes accompany entail suppressing existing memory cells but also inhibiting the memory T cell generation: Faster, more effective responses at generation of new ones. Immunosuppressive strategies that lower doses of antigen. J Immunol 164: 2338Ð2346, 2000 decrease the T cell clonal burst during the expansion phase of 12. Garcia S, DiSanto J, Stockinger B: Following the development of the primary alloimmune response, increase T cell apoptosis a CD4 T cell response in vivo: From activation to memory during the death phase, or both are expected to reduce the formation. Immunity 11: 163Ð171, 1999 number of newly generated memory T cells after transplanta- 13. Bachmann M, Barner M, Viola A, Kopf M: Distinct kinetics of tion. Alternatively, a better understanding of what determines cytokine production and cytolysis in effector and memory T cells the transition of an activated/effector lymphocyte into a mem- after viral infection. Eur J Immunol 29: 291Ð299, 1999 14. Veiga-Fernandes H, Walter U, Bourgeois C, McLean A, Rocha ory T cell could shed light on how to sway the immune B: Response of naive and memory CD8ϩ T cells to antigen response toward tolerance (Figure 1). Of course, the greatest stimulation in vivo. Nat Immunol 1: 47Ð53, 2000 challenge that remains is how to achieve all of this in an 15. Goldrath AW, Bevan MJ: Selecting and maintaining a diverse antigen-specific manner: selective deletion or suppression of T-cell repertoire. Nature 402: 255Ð262, 1999 alloreactive or self-reactive memory T cells without globally 16. Freitas AA, Rocha B: Peripheral T cell survival. Curr Opin compromising the host’s immune system. In other words, Immunol 11: 152Ð156, 1999 2408 Journal of the American Society of Nephrology J Am Soc Nephrol 14: 2402Ð2410, 2003

17. Mullbacher A: The long-term maintenance of 36. Gramaglia I, Jember A, Pippig S, Weinberg A, Killeen N, Croft memory does not require persistence of antigen. J Exp Med 179: M: The OX40 costimulatory receptor determines the develop- 317Ð321, 1994 ment of CD4 memory by regulating primary clonal expansion. 18. Murali-Krishna K, Lau LL, Sambhara S, Lemonnier F, Altman J, J Immunol 165: 3043Ð3050, 2000 Ahmed R: Persistence of memory CD8 T cells in MHC class 37. Tan JT, Whitmire JK, Murali-Krishna K, Ahmed R, Altman JD, I-deficient mice. Science 286: 1377Ð1381, 1999 Mittler RS, Sette A, Pearson TC, Larsen CP: 4Ð1BB costimula- 19. Swain SL, Hu H, Huston G: Class II-independent generation of tion is required for protective anti-viral immunity after peptide CD4 memory T cells from effectors. Science 286: 1381Ð1383, vaccination. J Immunol 164: 2320Ð2325, 2000 1999 38. Borrow P, Tishon A, Lee S, Xu J, Grewal IS, Oldstone MBA, 20. Takeda S, Rodewald HR, Arakawa H, Bluethmann H, Shimizu Flavell RA: CD40L-deficient mice show deficits in antiviral T: MHC class II molecules are not required for survival of newly immunity and have an impaired memory CD8ϩ CTL response. generated CD4ϩ T cells, but affect their long-term life span. J Exp Med 183: 2129Ð2142, 1996 Immunity 5: 217Ð228, 1996 39. Hendriks J, Gravestein LA, Tesselaar K, van Lier RAW, Shu- 21. Tanchot C, Lemonnier FA, Perarnau B, Freitas AA, Rocha B: macher TNM, Borst J: CD27 is required for generation and Differential requirements for survival and proliferation of CD8 long-term maintenance of T cell immunity. Nat Immunol 1: naive or memory T cells. Science 276: 2057Ð2062, 1997 433Ð440, 2000 22. Dai Z, Lakkis FG: Cutting edge: Secondary lymphoid organs are 40. Liu Y, Wenger RH, Zhao M, Nielsen PJ: Distinct costimulatory necessary for maintaining the CD4, but not CD8, molecules are required for the induction of effector and memory pool. J Immunol 167: 6711Ð6715, 2001 cytotoxic T lymphocytes. J Exp Med 185: 252Ð262, 1997 23. Reinhardt RL, Khoruts A, Merica R, Zell T, Jenkins MK: Visu- 41. Suresh M, Whitmire J, Harrington L, Larsen C, Pearson T, alizing the generation of memory CD4 T cells in the whole body. Altman J, Ahmed R: Role of CD28-B7 interactions in generation Nature 410: 101Ð105, 2001 and maintenance of CD8 T cell memory. J Immunol 167: 5565Ð 24. Masopust D, Vezys V, Marzo AL, Lefrancois L: Preferential 5573, 2001 localization of effector memory cells in nonlymphoid tissue. 42. Lantz O, Grandjean I, Matzinger P, Di Santo J: ␥ chain is Science 291: 2413Ð2417, 2001 required for naive CD4ϩ T cell survival but not for antigen 25. Chalasani G, Dai Z, Konieczny BT, Baddoura FK, Lakkis FG: proliferation. Nat Immunol 1: 54Ð58, 2000 Recall and propagation of allospecific memory T cells indepen- 43. Dai Z, Konieczny BT, Lakkis FG: The dual role of -2 dent of secondary lymphoid organs. Proc Natl Acad SciUSA in the generation and maintenance of CD8ϩ memory T cells. 99: 6175Ð6180, 2002 J Immunol 165: 3031Ð3036, 2000 26. Kaech SM, Ahmed R: Memory CD8ϩ T cell differentiation: 44. Blattman J, Grayson J, Wherry E, Kaech S, Smith K, Ahmed R: Initial antigen encounter triggers a developmental program in Therapeutic use of IL-2 to enhance antiviral T-cell responses. naive cells. Nat Immunol 2: 415Ð422, 2001 Nat Med 9: 540Ð547, 2003 27. Hu H, Huston G, Duso D, Lepak N, Roman E, Swain SL: CD4ϩ 45. Schluns KS, Williams K, Ma A, Zheng XX, Lefrancois L: T cell effectors can become memory cells with high efficiency Cutting edge: Requirement for IL-15 in the generation of primary and without further division. Nat Immunol 2: 705Ð710, 2001 and memory antigen-specific CD8 T cells. J Immunol 168: 28. Hou S, Hyland L, Ryan K, Portner A, Doherty P: Virus-specific 4827Ð4831, 2002 CD8ϩ T cell memory determined by clonal burst size. Nature 369: 652Ð654, 1994 46. Janssen E, Lemmens E, Wolfe T, Christen U, von Herrath M, 29. Murali-Krishna K, Altman JD, Suresh M, Sourdive DJD, Zajac Schoenberger S: CD4 T cells are required for secondary expan- AJ, Miller JD, Slansky J, Ahmed R: Counting antigen-specific sion and memory in CD8 T lymphocytes. Nature 421: 852Ð854, CD8 T cells: A re-evaluation of bystander activation during viral 2003 infections. Immunity 8: 177Ð187, 1998 47. Shedlock D, Shen H: Requirement for CD4 T cell help in 30. Biron C: Interferons alpha and beta as immune regulators—A generating functional CD8 T cell memory. Science 300: 337Ð new look. Immunity 14: 661Ð664, 2001 339, 2003 31. Akira S, Takeda K, Kaisho T: Toll-like receptors: Critical pro- 48. Sun J, Bevan M: Defective CD8 T cell memory following acute teins linking innate and acquired immunity. Nat Immunol 2: infection without CD4 T cell help. Science 300: 339Ð342, 2003 675Ð680, 2001 49. Bourgeois C, Rocha B, Tanchot C: A role for CD40 expression ϩ ϩ 32. Christopher K, Mueller T, Ma C, Liang Y, Perkins D: Analysis on CD8 T cells in the generation of CD8 T cell memory. of innate and adaptive phases of allograft rejection by cluster Science 297: 2060Ð2063, 2002 analysis of transcriptional profiles. J Immunol 169: 522Ð530, 50. Lanzavecchia A, Sallusto F: Dynamics of T lymphocyte re- 2002 sponses: Intermediates, effectors, and memory cells. Science 33. Goldstein D, Tesar B, Akira S, Lakkis F: Critical role of the 290: 92Ð97, 2000 Toll-like receptor signal adaptor protein Myd88 in acute allograft 51. Sprent J, Tough DF: T cell death and memory. Science 293: rejection. J Clin Invest 111: 15771Ð1578, 2003 245Ð248, 2001 34. Pape KA, Merica R, Mondino A, Khoruts A, Jenkins MK: Direct 52. Jacob J, Baltimore D: Modelling T-cell memory by genetic evidence that functionally impaired CD4ϩ T cells persist in vivo marking of memory T cells in vivo. Nature 399: 593Ð597, 1999 following induction of peripheral tolerance. J Immunol 160: 53. Opferman JT, Ober BT, Ashton-Rickardt PG: Linear differenti- 4719Ð4729, 1998 ation of cytotoxic effectors into memory T lymphocytes. Science 35. Whitmire JK, Flavell RA, Grewal IS, Larsen CP, Pearson TC, 283: 1745Ð1748, 1999 Ahmed R: CD40-CD40 ligand costimulation is required for 54. Manjunath N, Shankar P, Wan J, Weninger W, Crowley MA, generating antiviral CD4 T cell responses but is dispensable for Hieshima K, Springer TA, Fan X, Shen H, Lieberman J, von CD8 T cell responses. J Immunol 163: 3194Ð3201, 1999 Andrian UH: Effector differentiation is not prerequisite for gen- J Am Soc Nephrol 14: 2402Ð2410, 2003 Memory T Cells 2409

eration of memory cytotoxic T lymphocytes. J Clin Invest 108: (CD44low, LY-6CϪ) to TCR/CD8 signaling in response to anti- 871Ð878, 2001 gen. J Immunol 160: 3236Ð3242, 1998 55. Geginat J, Sallusto F, Lanzavecchia A: Cytokine-driven prolif- 74. Bachmann MF, Gallimore A, Linkert S, Cerundolo V, Lanza- eration and differentiation of human naive, central memory, and vecchia A, Kopf M, Viola A: Developmental regulation of Lck effector memory CD4ϩ T cells. J Exp Med 194: 1711Ð1719, targeting to the CD8 coreceptor controls signaling in naive and 2001 memory T cells. J Exp Med 189: 1521Ð1530, 1999 56. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A: Two 75. Lanzavecchia A, Sallusto F: From synapses to immunological subsets of memory T lymphocytes with distinct homing poten- memory: The role of sustained T cell stimulation. Curr Opin tials and effector functions. Nature 401: 708Ð712, 1999 Immunol 12: 92Ð98, 2000 57. Weninger W, Crowley MA, Manjunath N, von Andrian UH: 76. Kaech S, Hemby S, Kersh E, Ahmed R: Molecular and func- Migratory properties of naive, effector, and memory CD8ϩ T tional profiling of memory CD8 T cell differentiation. Cell 111: cells. J Exp Med 194: 953Ð966, 2001 837Ð851, 2002 58. Iezzi G, Scheidegger D, Lanzavecchia A: Migration and function 77. Busch D, Pamer E: T cell affinity maturation by selective ex- of antigen-primed nonpolarized T lymphocytes in vivo. J Exp pansion during infection. J Exp Med 189: 701Ð710, 1999 ϩ Med 193: 987Ð993, 2001 78. Croft M, Bradley LM, Swain, S: Naive versus memory CD4 59. Wherry E, Teichgraber V, Becker T, Masopust D, Kaech S, responses to antigen: Memory cells are less dependent on acces- Antia R, von Andrian U, Ahmed R: Lineage relationship and sory cell costimulation and respond to many antigen-presenting protective immunity of memory CD8 T cell subsets. Nat Immu- cell types including resting B cells. J Immunol 152: 2675Ð2685, nol 4: 225Ð234, 2003 1994 60. Tough DF, Sprent J: Turnover of naive- and memory-phenotype 79. Iezzi G, Karjalainen K, Lanzavecchia A: The duration of anti- T cells. J Exp Med 179: 1127Ð1135, 1994 genic stimulation determines the fate of naive and effector T 61. Bachmann MF, Odermatt B, Hengartner H, Zinkernagel RM: cells. Immunity 8: 89Ð95, 1998 Induction of long-lived germinal centers associated with persist- 80. London CA, Lodge MP, Abbas AK: Functional responses and ϩ ing antigen after viral infection. J Exp Med 183: 2259Ð2269, costimulator dependence of memory CD4 T cells. J Immunol 1996 164: 265Ð272, 2000 81. Johnston B, Butcher E: Chemokines in rapid leukocyte adhesion 62. Zhang X, Sun S, Hwang I, Tough DF, Sprent J: Potent and triggering and migration. Semin Immunol 14: 83Ð92, 2002 selective stimulation of memory-phenotype CD8ϩ T cells in 82. Koelle D, Liu Z, McClurkan C, Topp M, Riddell S, Pamer E, vivo by IL-15. Immunity 8: 591Ð599, 1998 Johnson A, Wald A, Corey L: Expression of cutaneous lympho- 63. Ku CC, Murakami M, Sakamoto A, Kappler J, Marrack P: cyte-associated antigen by CD8ϩ T cells specific for skin-tropic Control of homeostasis of CD8ϩ memory T cells by opposing virus. J Clin Invest 110: 537Ð548, 2002 cytokines. Science 288: 675Ð678, 2000 83. Hudak S, Hagen M, Liu Y, Catron D, Oldham E, McEvoy L, 64. Lau LL, Jamieson BD, Somasundaram T, Ahmed R: Cytotoxic Bowman E: Immune surveillance and effector functions of T-cell memory without antigen. Nature 369: 648Ð652, 1994 CCR10ϩ skin homing T cells. J Immunol 169: 1189Ð1196, 2002 65. Ahmed R, Gray D: and protective im- 84. Campbell D, Butcher E: Rapid acquisition of tissue-specific munity: Understanding their relation. Science 272: 54Ð60, 1996 homing phenotypes by CD4ϩ T cells activated in cutaneous or 66. Becker T, Wherry E, Boone D, Murali-Krishna, K, Antia R, Ma mucosal lymphoid tissues. J Exp Med 195: 135Ð141, 2002 A, Ahmed R: is required for proliferative renewal 85. Kim S-K, Schluns K, Lefrancois L: Induction and visualization of virus-specific memory CD8 T cells. J Exp Med 195: 1541Ð of mucosal memory CD8ϩ T cells following systemic virus 1548, 2002 infection. J Immunol 163: 4125Ð4132, 1999 67. Judge A, Zhang X, Fujii H, Surh C, Sprent J: Interleukin 15 86. Pantenburg B, Heinzel F, Das L, Heeger P, Valujskikh A: T cells controls both proliferation and survival of a subset of memory- primed by leishmania major infection cross-react with alloanti- ϩ phenotype CD8 T cells. J Exp Med 196: 935Ð946, 2002 gens and alter the course of allograft rejection. J Immunol 169: 68. Schluns KS, Kieper WC, Jameson SC, LeFrancois L: Interleu- 3686Ð3693, 2002 kin-7 mediates the homeostasis of naive and memory CD8 T 87. Brehm M, Markees T, Daniels K, Greiner D, Rossini A, Welsh cells in vivo. Nat Immunol 1: 426Ð432, 2000 R: Direct visualization of cross-reactive effector and memory 69. Bradley LM, Duncan DD, Tonkonogy SL, Swain SL: Charac- allo-specific CD8 T cells generated in response to viral infection. ϩ terization of antigen-specific CD4 effector T cells in vivo: J Immunol 170: 4077Ð4086, 2003 results in transient population of MEL-14-, 88. Burrows S, Khanna R, Burrows J, Moss D: An alloresponse in CD45RB- helper cells that secretes IL-2, IL-3, and IL-4. J Exp humans is dominated by cytotoxic T lymphocytes (CTL) cross- Med 174: 547Ð559, 1991 reactive with a single Epstein-Barr virus CTL epitope: Implica- 70. Dutton RW, Bradley LM, Swain SL: T cell memory. Annu Rev tions for graft-versus-host disease. J Exp Med 179: 1155Ð1161, Immunol 16: 201Ð223, 1998 1994 71. Kedl RM, Mescher MF: Qualitative differences between naive 89. Azzawi M, Hasleton PS, Geraghty PJ, Yonan N, Krysiak P, and memory T cells make a major contribution to the more rapid El-Gammal A, Deiraniya AK, Hutchinson IV: RANTES chemo- and efficient memory CD8ϩ T cell response. J Immunol 161: kine expression is related to acute cardiac cellular rejection and 674Ð683, 1998 infiltration by CD45RO T-lymphocytes and macrophages. 72. Pihlgren M, Dubois PM, Tomkowiak M, Sjogren T, Marvel J: J Heart Lung Transplant 17: 881Ð887, 1998 Resting memory CD8ϩ T cells are hyperreactive to antigenic 90. Salom RN, Maguire JA, Esmore D, Hancock WW: Analysis of challenge in vitro. J Exp Med 184: 2141Ð2151, 1996 proliferating cell nuclear antigen expression aids histological 73. Curtsinger J, Lins D, Mescher M: CD8ϩ: memory T cells diagnosis and is predictive of progression of human cardiac (CD44high, Ly-6Cϩ) are more sensitive than naive cells allograft rejection. Am J Pathol 145: 876Ð882, 1994 2410 Journal of the American Society of Nephrology J Am Soc Nephrol 14: 2402Ð2410, 2003

91. Akbar AN, Amlot PL, Timms A, Lombardi G, Lechler R, 102. Zhai Y, Meng L, Gao F, Busuttil R, Kupiec-Weglinski J: Allo- Janossy G: The development of primed/memory CD8ϩ lympho- graft rejection by primed/memory CD8ϩ T cells is CD154 cytes in vitro and in rejecting kidneys after transplantation. Clin blockade resistant: Therapeutic implications for sensitized trans- Exp Immunol 81: 225Ð231, 1990 plant recipients. J Immunol 169: 4667Ð4673, 2002 92. Ibrahim S, Dawson DV, Sanfilippo F: Predominant infiltration of 103. Valujskikh A, Pantenburg B, Heeger P: Primed allospecific T rejecting human renal allografts with T cells expressing CD8 and cells prevent the effects of costimulatory blockade on prolonged CD45RO. Transplantation 59: 724Ð728, 1995 cardiac allograft survival in mice. Am J Transplant 2: 501Ð509, 93. Reinke P, Fietze E, Docke WD, Kern F, Ewert R, Volk HD: Late 2002 acute rejection in long-term renal allograft recipients. Diagnostic 104. Adams A, Williams A, Jones T, Shirasugi N, Durham M, and predictive value of circulating activated T cells. Transplan- Kaech S, Wherry E, Omani T, Lanier J, Kokko K, Pearson T, tation 58: 35Ð41, 1994 Ahmed R, Larsen C: Heterologous immunity provides a po- 94. Najafian N, Salama A, Fedoseyeva E, Benichou G, Sayegh M: tent barrier to transplantation tolerance. J Clin Invest 111: Enzyme-linked immunosorbent spot assay analysis of peripheral 1887Ð1895, 2003 blood lymphocyte reactivity to donor HLA-DR peptides: Poten- 105. Charlton B, Hugh Auchincloss J, Fathman CG: Mechanisms tial novel assay for prediction of outcomes for renal transplant of transplantation tolerance. Annu Rev Immunol 12: 707Ð734, recipients. J Am Soc Nephrol 13: 252Ð259, 2002 1994 95. Cerottini JC, Engers HD, Macdonald HR, Brunner T: Generation 106. Kreuwel H, Aung S, Silao C, Sherman L: Memory CD8ϩ T cells of cytotoxic T lymphocytes in vitro. I. Response of normal and undergo peripheral tolerance. Immunity 17: 73Ð81, 2002 immune spleen cells in mixed leukocyte cultures. J Exp Med 140: 107. Kursar M, Bonhagen K, Fensterle J, Kohler A, Hurwitz R, 703Ð717, 1974 Kamradt T, Kaufmann S, Mittrucker H-W: Regulatory 96. Jones ND, Van Maurik A, Hara M, Gilot BJ, Morris PJ, Wood CD4ϩCD25ϩ T cells restrict memory CD8ϩ T cell responses. KJ: T-cell activation, proliferation, and memory after cardiac J Exp Med 196: 1585Ð1592, 2002 transplantation in vivo. Ann Surg 229: 570Ð578, 1999 108. Ahmadzadeh M, Farber D: Functional plasticity of an antigen- 97. Gordon R, Mathieson B, Samelson L, Boyse E, Simpson E: The specific memory CD4 T cell population. Proc Natl Acad Sci U S effect of allogeneic presensitization on H-Y graft survival and in A 99: 11802Ð11807, 2002 vitro cell-mediated responses to H-Y antigen. J Exp Med 144: 810Ð820, 1976 109. Yoshinaga S, Whoriskey J, Khare S, Sarmiento U, Guo J, Horan 98. Ruiz P, Nassiri M, Viciana AL, Padmanabhan J, Streilein JW: T, Shih G, Zhang M, Coccia M, Kohno T, Tafuri-Bladt A, Characterization of donor chimerism, alloreactive host T cells and Brankow D, Campbell P, Chang D, Chiu L, Dai T, Duncan C, memory cell development in thymi from mice resistant to neonatal Elliot G, Hui A, McCabe S, Scully S, Shahinian A, Shaklee C, transplantation tolerance. J Immunol 154: 633Ð643, 1995 Van G, Mak T, Senaldi G: T-cell co-stimulation through B7RP-1 99. Ruiz P, Nassiri M, Gregorian S, Viciana AL, Streilein JW: and ICOS. Nature 402: 827Ð832, 1999 Neonatal transplantation tolerance is associated with a systemic 110. Harada H, Salama A, Sho M, Izawa A, Sandner S, Ito T, Akiba reduction in memory cells, altered chimeric cell phenotype, and H, Yagita H, Sharpe A, Freeman G, Sayegh M: The role of the modified eicosanoid and cytokine production. Transplantation ICOS-B7h T cell costimulatory pathway in transplantation im- 61: 1198Ð1205, 1996 munity. J Clin Invest 2003, in press 100. Chace JH, Cowdery JS, Field EH: Effect of anti-CD4 on CD4 111. Ozkaynak E, Gao W, Shemmeri N, Wnag C, Gutierrex-Ramos subsets. I. Anti-CD4 preferentially deletes resting, naive CD4 J-C, Amaral J, Qin S, Rottman JB, Coyle AJ, Hancock WW: cells and spares activated CD4 cells. J Immunol 152: 405Ð412, Importance of ICOS-B7RP-1 costimulation in acute and chronic 1994 allograft rejection. Nat Immunol 2: 591Ð596, 2001 101. Propper DJ, Woo J, Macleod AM, Catto GR, Thomson AW: The 112. Yuan X, Salama A, Dong V, Schmitt I, Najafian N, Chandraker effects of rapamycin on in vivo. Suppression of A, Sayegh M: The role of the CD134-CD134 ligand costimula- primary responses but not of ongoing alloantibody synthesis or tory pathway in alloimmune responses in vivo. J Immunol 170: memory responses. Transplantation 54: 1058Ð1063, 1992 2949Ð2955, 2003