Effector CD8 T Cell Development

2 downloads 0 Views 291KB Size Report
Foundation, the Cancer Research Institute, and the Richard K. Gershon Predoctoral. Fellowship. 2 Address correspondence and reprint requests to Dr. Susan M.
OF

THE

JOURNAL IMMUNOLOGY

BRIEF REVIEWS Effector CD8 T Cell Development: A Balancing Act between Memory Cell Potential and Terminal Differentiation1 Nikhil S. Joshi and Susan M. Kaech2 Immune responses to infection are optimally designed to generate large numbers of effector T cells while simultaneously minimizing the collateral damage of their potentially lethal actions and generating memory T cells to protect against subsequent encounter with pathogens. Much remains to be discovered about how these equally essential processes are balanced to enhance health and longevity and, more specifically, what factors control effector T cell expansion, differentiation, and memory cell formation. The innate immune system plays a prominent role in the delicate balance of these decisions. Insights into these questions from recent work in the area of effector CD8 T cell differentiation will be discussed. The Journal of Immunology, 2008, 180: 1309 –1315.

T

he generation of long-term immunity and efficacious vaccines against many viral and bacterial agents is dependent on the formation of large numbers of longlived memory CD8 T cells. Memory CD8 T cells have special features that make them well suited to respond quickly and effectively to reinfection. Compared with naive CD8 T cells, Agspecific memory CD8 T cells are present in greater number and have an increased ability to survey nonlymphoid (peripheral) sites for the presence of infection (1). Memory CD8 T cells appear poised for a rapid response to secondary infection because they persist in a “progrowth” state with low levels of p27kip and high levels of cyclin-dependent kinase 6 (CDK6)-cyclin D3 complexes (2, 3) and maintain mRNA expression of several cytotoxic proteins, antiviral cytokines, and chemokines (1, 3–7). These features allow the memory CD8 T cells to expand and develop effector functions faster than naive CD8 T cells. Memory CD8 T cells can also survive long term in the absence of Ag (⬎2 years in mice and ⬎50 years in humans) (8, 9) and are maintained through a process of self-renewal driven by IL-15 and IL-7 (also called homeostatic proliferation or turnover; Ref. 10). Given their long lifespan and ability to provide protection against recurrent infections, increasing the quantity and quality Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520 Received for publication November 6, 2007. Accepted for publication December 15, 2007. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1 This work was supported by the National Institutes of Health Grants AI066232 and T32 AI055403, the Burroughs-Wellcome Fund Grant 1004313, the Edward Mallinckrodt, Jr.

www.jimmunol.org

of memory CD8 T cells is paramount to improving the efficacy of most vaccines. Acquiring a terminally differentiated effector CD8 T cell state

In general, terminal differentiation of specialized cell types begins with immature precursor cells that progressively acquire (often in a stepwise manner) a distinguished gene expression profile, a specialized set of functions, and an altered morphology. As terminal differentiation proceeds, cells typically display a loss of multipotency (i.e., the ability to become another cell type), proliferative capacity, and telomerase activity (11). The lifespan of terminally differentiated cells can vary quite dramatically from relatively long-lived fates, such as neurons or Absecreting plasma cells that live for many years, to short-lived fates, such as neutrophils, intestinal villi, or epidermal skin cells that live for a few hours, days, or weeks, respectively. What properties distinguish terminally differentiated CD8 T cells and how this process influences memory CD8 T cell development are important questions to understand. In some ways, a mature CD8⫹ thymic emigrant might be considered a terminally differentiated cell type because it has acquired a distinct and stable naive T cell phenotype, can no longer differentiate into other types of lymphocytes, and does not express telomerase (12, 13). However, the naive stage is not necessarily a terminal endpoint, because when activated by Ag this cell proliferates profoundly and differentiates into an effector CD8 T cell that acquires a different pattern of gene expression, a more specialized set of functions (such as cytotoxicity and antiviral cytokine production), and telomerase activity (4, 14). However, after several rounds of cell division the proliferative potential of effector CD8 T cells declines and they become highly sensitive to cell death (15). For these latter reasons, effector CD8 T cell could be considered terminally differentiated, but if this is the case then how do longlived memory CD8 T cells arise from this population? Compared with effector T cells, memory CD8 T cells exhibit less terminally differentiated phenotypes because they are multipotent (they can remain resting memory cells or redifferentiate into cytotoxic effector cells), can self-renew, have a high proliferative potential and increased longevity (1, 16). In addition Foundation, the Cancer Research Institute, and the Richard K. Gershon Predoctoral Fellowship. 2 Address correspondence and reprint requests to Dr. Susan M. Kaech, 300 Cedar Street, The Anlyan Center S641B, Yale University School of Medicine, P. O. Box 208011, New Haven, CT 06520. E-mail address: [email protected]

Copyright © 2008 by The American Association of Immunologists, Inc. 0022-1767/08/$2.00

1310

BRIEF REVIEW: MECHANISMS FOR GENERATING EFFECTOR CD8 T CELL DIVERSITY

to these “stem cell-like” properties, there are many notable differences between the memory and effector CD8 T cell gene expression profiles (4, 5). Yet, the memory cell gene expression pattern also bears a strong resemblance to terminally differentiated effector cells (such as the sustained expression of many “effector-like” mRNAs such as IFN-␥, granzymes, perforin, chemokines, and chemokine receptors (3, 4, 17) and the continued expression of telomerase (14). Thus, memory CD8 T cells constitute a unique cell population whose members appear less differentiated than their terminally differentiated effector cell predecessors with regard to proliferative potential and multipotency, yet are imprinted with several key effector cell traits. This review will discuss potential mechanisms for how some effector T cells may be preserved in a less differentiated state during an immune response. The heterogeneity of effector and memory CD8 T cells

One prominent aspect that has come to light over the past few years is the heterogeneity of effector and memory T cells, and understanding the ontogeny of these different cell types will lend insight into memory T cell development. During many infections and immunizations, diverse effector CD8 T cell populations form that can sometimes be separated into cell subsets that differ in their anatomical locations, effector functions, proliferative capacity, and long-term fates. Several markers have become useful in this regard such as CD62L, CCR7, CD103, and ␣4␤7 (anatomical localization), IL-2/IFN-␥/TNF-␣, perforin, granzymes A/B/C/K, and programmed death-1 (effector functions), and Bcl-2, IL-7R, CD122, CD28, CD57, CD27, killer cell lectin-like receptor G1 (KLRG1),3 CXCR3, CD43, and CD62L (survival and/or proliferative capacity). Other markers such as Ly6C, NKG2A, and 2B4 can show heterogeneous expression among effector CD8 T cells but have not yet been attributed to a cellular process (3, 17–33). Various factors have been recognized to affect the expression of these markers such as the type or duration of infection, certain common ␥-chain and inflammatory cytokines, Ag specificity, naive T cell precursor frequency, and their location within the body (see Refs. 7, 19, 20 –22, 26 –31, and 34 – 46). With respect to memory CD8 T cell development, our ability to distinguish effector CD8 T cells that are destined to survive and become long-lived memory CD8 T cells (referred to here as memory precursor effector cells; MPECs) from those that are not (referred to as short-lived effector cells; SLECs) has markedly improved in several experimental systems using a combination of functional criteria and surface marker expression. Some studies from mice and nonhuman primates have noted that CD8 and CD4 T cells capable of producing IL-2 or simultaneously producing IFN-␥, TNF-␣, and IL-2 (triple producers) preferentially survive and provide greater protection against rechallenge compared with those that only produce one cytokine (21, 30, 47, 48). Other work has found that during acute lymphocytic choriomeningitis virus (LCMV) and Listeria monocytogenes infections in mice, the expression of the receptors IL-7R and KLRG1 are very valuable for detecting MPECs and SLECs (23, 30, 49). For the most part, IL-7R and KLRG1 are inversely expressed on MPECs and SLECs, with SLECs being 3 Abbreviations used in this paper: KLRG1, killer cell lectin-like receptor; LCMV, lymphocytic choriomeningitis virus; MPEC, memory precursor effector cell; SLEC, shortlived effector cell; TCM, central memory T cell; TEM, effector memory T cell.

KLRG1highIL-7Rlow and MPECs being KLRG1lowIL-7Rhigh (30). However, it is important to note that the combination of KLRG1 and IL-7R alone are still not exact indicators of SLECs and MPECs because not all IL-7Rhigh effector CD8 T cells become memory cells, some KLRG1high IL-7Rlow cells persist for some time following primary and/or secondary infections, and some effector and memory CD8 T cells express both KLRG1 and IL-7R (23, 30). Moreover, noninfectious methods of T cell activation (e.g., dendritic cell vaccination) can generate nearly uniform IL-7Rhigh MPEC-like CD8 T cell populations, but many of these cells do not become long-lived memory CD8 T cells (41, 42, 50, 51). In this review, the IL-6 receptor may also help to distinguish memory precursor cells (51). These results suggest that the effector CD8 T cells may reside in a range of differentiated states spanning the SLEC and MPEC states, and more attributes are necessary to refine the identity of these important cell types. The diversity in effector CD8 T cells is likely to be related to that found in the memory CD8 T cell population, but it has been difficult to track the developmental lineage of CD8 T cells between these two stages because the memory T cell population appears to continuously evolve over time following infections (19, 29, 30, 52, 53). Multiple subsets of memory T cells have been characterized, most notably, the central (TCM) and effector (TEM) memory T cells (based on CD62L and CCR7 expression) (18). Compared with TCM cells, TEM cells have an decreased ability to traffic to lymphoid tissues and appear more terminally differentiated because, in many cases but not all, they have a lower capacity to proliferate to Ag and homeostatic cytokines (IL-15 and IL-7) and shortened telomere length (18, 19, 54). However, even within the TCM and TEM categories there seems to be additional heterogeneity. For example, studies from Sendai virus infection found that the TCM cells persisting a year after infection had a higher proliferative capacity than TCM cells found one month after infection (55). This data is somewhat akin to that found following LCMV infection (4). When these Sendai-specific memory CD8 T cells were interrogated further, the cells expressing heightened levels of CD27 and CXCR3 demonstrated the greatest proliferative potential, yet they did not fall into the traditional TCM or TEM subsets (29). Understanding the derivation of the memory CD8 T cell subsets is complicated because although several cell surface phenotypes are stable, some are not. For example, following acute LCMV infection the expression of KLRG1 and IL-7R appears quite stable and little conversion is observed between the SLEC and MPEC subsets (23, 30). However, the expression of CD62L and CD27 can vary according to time and/or tissue residence (19, 36, 44, 45). LCMV-specific KLRG1highIL-7Rlow SLECs that persist into “memory” time points retain a TEM cell phenotype (CD62Llow, CD27low, IL-2low, bcl-2low; low proliferative capacity), but the memory cells generated from MPECs are often a mixture of TEM and TCM (23, 30, 32, 33, 41, 45) (N.S.J. and S.M.K., unpublished data). Furthermore, it has been noted by several groups that following some acute infections the memory CD8 T cell population evolves progressively over time into one that is enriched for cells of a higher proliferative capacity (to Ag and homeostatic cytokines) and greater longevity that can be characterized as IL-7Rhigh, KLRG1low, CD27high, CD62Lhigh, CXCR3high, IL-2high, and Bcl-2high (19, 22, 23, 25, 29, 30, 52). This trend can be altered in latent or chronic infections, and the stability of

The Journal of Immunology

1311

certain memory CD8 T cell subsets may be tailored by the type of viral infection (1, 46, 53). In addition to better understanding the origin of different memory T cell subsets, it is also important to determine their relevance and roles in providing long-term protection. Developmental lineages of memory CD8 T cells

How is effector CD8 T cell differentiation balanced to permit the formation of effector cell properties in the MPECs and yet prevent them from acquiring a terminal SLEC state? Data from many murine model systems of infection indicate that the precursors to memory CD8 T cells acquire a wide range of potent effector cell qualities and therefore do not “bypass” the effector cell stage. Evidence for this point is illustrated by the memory CD8 T cell gene expression patterns discussed above and several lineage-tracing experiments that showed that at some point memory CD8 T cells expressed effector molecules (4, 5, 56 – 58). Moreover, LCMV-specific KLRG1lowIL-7Rhigh MPECs perform most effector functions as well as SLECs (23, 30). This point should be kept in mind when discussing activated T cell fate decisions, because other frequently used terms such as “memory” or “effector” fates, may be misleading as they imply that the “memory” fated CD8 T cells do not acquire effector T cell properties. Indeed, other scenarios may exist during some types of noninfectious immunizations (with weaker inflammatory or antigenic stimulations) where the memory CD8 T cell precursor population does not develop a full-fledged set of effector cell properties. Under these conditions though, factors that enhance the differentiation of effector functions often promote memory CD8 T cell formation (15, 58 – 60). Nonetheless, based on the studies made during infection it is clear that the expression of effector molecules does not preclude memory CD8 T cell formation. Certain developmental models may help to explain how some effector CD8 T cells gain or maintain memory cell potential whereas others do not (Fig. 1). One model may be that all CD8 T cells reach a terminally differentiated effector stage but then some are capable of dedifferentiating into cells that gain longevity and a high proliferative potential (Fig. 1A). Experimental evidence supports a dedifferentiation model to a certain degree because the surviving IL-7Rhigh MPECs “functionally mature” during the effector to memory (E3 M) transition period and gradually acquire an increased capacity to proliferate and produce IL-2, Bcl-2, and CD62L (4, 19, 23, 25, 61). What instructs this functional maturation is unclear, but IL-2 exposure during infection, the presence of CD4 T cells, and maintaining lower expression of T-bet appear to be important for establishing a memory CD8 T cell population with a higher proliferative capacity (61– 63). A second model for generating CD8 T cell diversity is that an activated CD8 T cell differentiates in a stepwise manner and, in so doing, progressively acquires a more terminally differentiated phenotype; the cells that become MPECs do not progress as far as the SLECs (Fig. 1B). In this model, often referred to as the “decreasing potential model,” the progression of differentiation may be controlled by successive stimulations with Ag or other signals (64). Thus, the differentiation state of an effector CD8 T cell is reflective of the cumulative history of signals that were encountered during infection. Such a linear model provides a nice mechanism for generating a spectrum of different types of effector and memory CD8 T cells (36, 64 – 66). One requisite

FIGURE 1. Models for generating diverse differentiated states of effector and memory CD8 T cells. A, Dedifferentiation model. After activation by Ag, naive CD8 T cells become terminally differentiated (dark blue cells), fully functional, cytotoxic effector CD8 T cells. Following infection, the majority of effector cells die (indicated by cross), but a minority progressively dedifferentiate into long-lived memory CD8 T cells (purple-shaded cells). If cells are activated by Ag in the absence of inflammation/costimulation (dashed arrow), this will lead to tolerance and/or deletion of T cells. B, Decreasing potential model. The degree of effector cell differentiation is regulated by the duration of exposure to extrinsic factors such as Ag and inflammatory cytokines (indicated by black shading in triangle). Cumulative encounters with these signals drive the cells toward a terminally differentiated state (as indicated by intensifying blue shading). The majority of terminally differentiated effector cells die, but those that do not reach this end stage develop into memory CD8 T cells (purple cells). The memory CD8 T cell phenotypes vary according to the differentiation state of the effector cells from which they descended; curved arrows with bold, thin, or dashed lines indicate a high, medium, or low degree of proliferative potential and longevity. C, Divergent lineage model. As described in B, except that the degree of effector cell differentiation is controlled by the strength of the signal to which naive CD8 T cells are exposed early during T cell activation.

of the linear model is that all activated CD8 T cells transition through an MPEC stage as they terminally differentiate; if true, then controlling the progression of terminal differentiation might enable the generation of more memory CD8 T cells. Some evidence for this model may be found in studies in which the duration of infection is truncated and activated CD8 T cells develop into protective memory CD8 T cells more efficiently and rapidly, but to date these approaches have not resulted in a larger number of memory CD8 T cells (30, 41– 43, 67– 69). This is likely because a major regulator of clonal expansion is

1312

BRIEF REVIEW: MECHANISMS FOR GENERATING EFFECTOR CD8 T CELL DIVERSITY

the load or duration of antigenic stimulation (70 –72). Therefore, to generate more memory CD8 T cells one may need to find a way to confine terminal differentiation without minimizing clonal expansion. A third working model for generating CD8 T cell diversity incorporates divergent developmental pathways and proposes that soon after activation daughter cells are instructed to generate SLEC or MPEC fates (Fig. 1C). The divergent model need not be completely asymmetric to only produce two cell types but could also incorporate a range of differentiated states according to the overall strength of signal a T cells sees at or near the time of priming (43, 73, 74). Experimental support for this model may be found in a recent study examining the first cell division of an activated CD8 T cell where it appeared to divide in an asymmetric manner producing daughter cells that have increased or decreased memory CD8 T cell potential (75). Other recent work tracking the formation of MPECs and SLECs during LCMV and Listeria infections shows that these two lineages begin to diverge from a common pool of KLRG1low effector cells after ⬎7–10 cell divisions and 4 days of infection (30, 33). In line with this model, several studies indicate that early and brief exposure to Ag and inflammation are sufficient to direct the formation of long-lived and short-lived effector CD8 T cell fates (30, 42, 43, 70). Clarifying the precise mechanism(s) at play will require elucidation of the numerous genetic pathways that control the balance between terminal differentiation and memory CD8 T cell developmental potential, and in so doing, it is likely that some aspects of all of the above models will be incorporated in this process depending on the cellular phenotype being analyzed. Moreover, the inductive signals that control these cell fate decisions are likely to differ and/or be produced with varying kinetics depending on the infectious pathogen and its tropism. A case in point is that the proportion of IL-7Rlow Ag-specific effector CD8 T cells formed varies across multiple types of infections (LCMV, Listeria, influenza, and vaccinia virus) and between tissues within a given infection (15, 29, 30, 43, 76 –78). Cytokine control of effector and memory CD8 T cell development

Perhaps our knowledge of effector CD8 T cell differentiation can be enhanced by that of their kindred lymphocytes, CD4 T cells. In many ways one can consider CD4 T cell differentiation as divergent, because under different priming conditions the same naive CD4 T cell can adopt one of several effector cell lineages according to the type of lineage-determining cytokines it was exposed to during activation (e.g., Th1, Th2, Th17, and regulatory T cell specifications are directed by IFN-␥/IL-12, IL-4, IL-6/TGF␤/IL-1␤, and TGF␤ exposure, respectively) (79). Although the repertoire of effector CD8 T cells may not be as diverse as that of CD4 T cells, a handful of inflammatory cytokines have been found to influence CD8 T cell differentiation (80). As with Th1 cells, the inflammatory cytokines IL-12, IFN-␥, and IFN␣␤ potently enhance effector CD8 T cell expansion, cytotoxicity, and production of antiviral cytokines, particularly when CD8 T cells are activated by a weak stimuli or cross-presented Ag (15, 79, 81– 86). Interestingly, the reliance of CD8 T cell expansion on particular inflammatory cytokines (IFN␣␤, IFN-␥, and IL-12) or IL-2 may depend on the type of infectious pathogen or the tissues they reside in, respectively

(83, 85, 87, 88). Additionally, inflammatory cytokines, particularly IL-12, induce the expression of Bcl-3, which can enhance activated CD8 T cell expansion (89, 90). Some inflammatory cytokines can also modulate the expression of key transcription factors that regulate effector T cell differentiation. T-bet is a major regulator of Th1 effector cell differentiation, whereas Tbet and eomesodermin (eomes), another T-box family member, appear to coordinately regulate the formation of effector CD8 T cell functions (91, 92). IFN-␥ is critical for T-bet induction and IL-12 acts to augment its expression in CD4 T cells (79); but in CD8 T cells, IL-12 augments T-bet expression and diminishes eomes expression (30, 86). The above data emphasize the beneficial role of inflammatory cytokines on maximizing effector T cell expansion and differentiation, but these cytokines may also act as a double-edged sword; on one hand they stimulate effector CD8 T cell function and expansion, but on the other they appear to drive terminal maturation and limit memory cell potential. Recent work has shown that inflammation, most notably by IFN-␥, promotes effector CD8 T cell contraction and down-regulation of IL-7R (84). However, another recent report showed that IFN-␥R was needed in a CD8 T cell-autonomous manner for normal memory CD8 T cell formation, so the mechanism of IFN-␥ action in different types of immune responses remains to be clarified (93). In Il12⫺/⫺ mice, a higher frequency of IL-7Rhigh MPECs and memory CD8 T cells form following Listeria infection (85, 86), suggesting that IL-12 plays a critical role in the effector CD8 T cell fate decisions. Our recent work has shown that IL-12 can induce T-bet expression in a dose-dependent manner in activated CD8 T cells and that T-bet may act like a rheostat to determine SLEC vs MPEC fate decisions, with high T-bet expression favoring the formation of KLRG1highIL-7Rlow SLECs and lower expression favoring KLRG1lowIL-7Rhigh MPECs (30). A similar pathway using IL-12 (and IL-18) to drive T-bet expression may be required for the terminal differentiation of short-lived KLRG1high NK cells during murine CMV infection (94 –97). These data demonstrate multiple modes of T-bet function. In both CD4 and CD8 T cells T-bet acts in a divergent manner to specify different cell fates, but in NK cells it may promote terminal maturation in a linear manner. Moreover, in CD4 T cells T-bet operates fairly asymmetrically to specify Th1 vs Th2, Th17, and regulatory T cell fates, whereas in CD8 T cells it operates according to an expression gradient to specify SLEC vs MPEC fates. Linking the inflammatory cytokines and lineage-determining transcription factors (such as T-bet) to both the development of effector functions and longevity provides a unique way for the innate immune response to regulate effector T cell homeostasis. Additional support for this model may be found in recent studies where specific TLR agonists differentially influenced the generation of long-lived IFN-␥/TNF-␣/IL-2 tripleproducer CD4 and CD8 T cells. Because T-bet repress IL-2, it will be interesting to determine whether the “memory-enhancing” adjuvants modulated T-bet expression levels in a manner that promoted both IL-2 production and memory cell generation (21, 48, 91). It is most certain that other cytokines aside from IL-12 and IFN-␥ will similarly regulate memory T cell potential in activated CD8 T cells, because not all infections and adjuvants elicit the same cytokine profiles. Also, the role of tolerogenic cytokines, such as IL-10, which can regulate IL-12 production, needs to be considered in this process. For example,

The Journal of Immunology

in Il10⫺/⫺ mice fewer memory CD8 T cells form following Listeria infection, but this is likely not due to direct effects of IL-10 on the CD8 T cells (98, 99). These data suggest that the balance between IL-12 and IL-10 expression may represent a potential regulatory axis in SLEC and MPEC development during certain types of infections. The role of other cell intrinsic factors in effector CD8 T cell terminal differentiation

Many other important cell-intrinsic factors regulate the memory potential of developing effector CD8 T cells. Similar to T-bet, the transcriptional regulator ID2 (inhibitor of differentiation 2) plays a critical role in the development of NK and CD8 T cells (100), and ID2-deficient CD8 T cells have a more IL-7RhighCD27high“MPEC-like” phenotype (101). Another transcription factor, Blimp-1 (prdm1), critical for the terminal differentiation of B cells into plasma cells, is expressed at high levels by IL-7Rlow SLECs (63, 102). CD8 T cells in Blimp1-deficient mice have an activated and highly proliferative phenotype, suggesting that Blimp1 expression is antiproliferative (103, 104). Moreover, potential antagonists to Blimp-1, Bcl-6, and its homologue, Bcl-6b, promote memory CD8 T cell development and increase proliferative responses (105, 106). Further evidence suggests that the reduced proliferative potential of KLRG1high CD8 T cells is regulated by their increased expression of the cell cycle inhibitor p27kip (52) and their reduced ability to express Bmi-1, a transcriptional repressor that promotes T cell proliferation (107). Undoubtedly, the list of factors involved in this process will continue to grow as more genes are examined, and gene expression profiling between LCMV-specific MPECs and SLECs has revealed a handful of interesting candidates (30). It will be interesting to discover in the future whether more extensive overlap exists in the genes that regulate differentiation of effector CD8, CD4, NK T cells, and NK cells. Regulating CD8 T cell longevity and maintenance

One aspect associated with effector CD8 T cell terminal differentiation is its shortened lifespan, but it is not clear how this process is regulated. IL-7 and IL-15 are critical for the longterm survival and homeostatic turnover of memory CD8 T cells (10), and prolonged deprivation of these cytokines has considerable consequences on the formation and or maintenance of memory CD8 T cells (22, 23, 108 –110). Our work indicates that KLRG1highIL-7Rlow SLECs depend on IL-15, but this alone is not sufficient to maintain these cells long term (30). Surprisingly, IL-7R overexpression was unable to save these cells from death after acute infection (52, 111), suggesting that the down-regulation of IL-7R is symptomatic of, but not causal to, their death. Moreover, increasing the expression of Bcl-2 and Bcl-XL or blocking their actions in effector CD8 T cells does not greatly affect the normal rate of effector cell death following infection (10, 22, 52, 109, 112, 113). Currently, key molecules that have been found to promote and prevent effector CD8 T cell contraction are the proapoptotic molecule Bim and the serine protease inhibitor 2A (Spi2A), respectively (114 –118). Together these data suggest that MPECs need to see IL-7 and IL-15 to become long-lived memory CD8 T cells, but SLECs die for reasons other than deprivation of these cytokines.

1313 Implications of effector CD8 T cell terminal differentiation on vaccine design

Understanding the factors that regulate the terminal differentiation of effector CD8 T cells is critical for designing vaccines that generate long-lasting CD8 T cell immunity. Much of the recent work in the vaccine field has focused on generating larger expansions of CD8 T cell subsets because of the direct relationship between effector cell expansion and memory formation (119). A number of adjuvants, being potent inducers of effector CD8 T cell expansion and effector function, have logically been included in a variety of vaccine formulations. However, in light of the additional role that some adjuvants may play in restricting memory CD8 T cell potential, it will be critical to find the proper adjuvants and their respective doses that balance the effector T cell expansion and terminal differentiation best for vaccines and other immunotherapies. Moreover, it is important to remember that studying the phenotype of effector CD8 T cells generated in response to vaccines may be as important as studying the number of CD8 T cells generated in response to vaccination, as demonstrated already by some experimental and clinical data (21, 53, 65).

Disclosures The authors have no financial conflict of interest.

References 1. Kaech, S. M., and E. J. Wherry. 2007. Heterogeneity and cell-fate decisions in effector and memory CD8⫹ T cell differentiation during viral infection. Immunity 27: 393– 405. 2. Veiga-Fernandes, H., and B. Rocha. 2004. High expression of active CDK6 in the cytoplasm of CD8 memory cells favors rapid division. Nat. Immunol. 5: 31–37. 3. Grayson, J. M., K. Murali-Krishna, J. D. Altman, and R. Ahmed. 2001. Gene expression in antigen-specific CD8⫹ T cells during viral infection. J. Immunol. 166: 795–799. 4. Kaech, S. M., S. Hemby, E. Kersh, and R. Ahmed. 2002. Molecular and functional profiling of memory CD8 T cell differentiation. Cell. 111: 837– 851. 5. Goldrath, A. W., C. J. Luckey, R. Park, C. Benoist, and D. Mathis. 2004. The molecular program induced in T cells undergoing homeostatic proliferation. Proc. Natl. Acad. Sci. USA 101: 16885–16890. 6. Luckey, C. J., D. Bhattacharya, A. W. Goldrath, I. L. Weissman, C. Benoist, and D. Mathis. 2006. Memory T and memory B cells share a transcriptional program of self-renewal with long-term hematopoietic stem cells. Proc. Natl. Acad. Sci. USA 103: 3304 –3309. 7. Bachmann, M. F., R. R. Beerli, P. Agnellini, P. Wolint, K. Schwarz, and A. Oxenius. 2006. Long-lived memory CD8⫹ T cells are programmed by prolonged antigen exposure and low levels of cellular activation. Eur. J. Immunol. 36: 842– 854. 8. Hammarlund, E., M. W. Lewis, S. G. Hansen, L. I. Strelow, J. A. Nelson, G. J. Sexton, J. M. Hanifin, and M. K. Slifka. 2003. Duration of antiviral immunity after smallpox vaccination. Nat. Med. 9: 1131–1137. 9. Lau, L. L., B. D. Jamieson, T. Somasundaram, and R. Ahmed. 1994. Cytotoxic Tcell memory without antigen. Nature 369: 648 – 652. 10. Boyman, O., J. F. Purton, C. D. Surh, and J. Sprent. 2007. Cytokines and T-cell homeostasis. Curr. Opin. Immunol. 19: 320 –326. 11. Weissman, I. L. 2000. Stem cells: units of development, units of regeneration, and units in evolution. Cell 100: 157–168. 12. Bhandoola, A., H. von Boehmer, H. T. Petrie, and J. C. Zuniga-Pflucker. 2007. Commitment and developmental potential of extrathymic and intrathymic T cell precursors: plenty to choose from. Immunity 26: 678 – 689. 13. Weng, N. P., B. L. Levine, C. H. June, and R. J. Hodes. 1996. Regulated expression of telomerase activity in human T lymphocyte development and activation. J. Exp. Med. 183: 2471–2479. 14. Hathcock, K. S., S. M. Kaech, R. Ahmed, and R. J. Hodes. 2003. Induction of telomerase activity and maintenance of telomere length in virus-specific effector and memory CD8⫹ T cells. J. Immunol. 170: 147–152. 15. Mescher, M. F., J. M. Curtsinger, P. Agarwal, K. A. Casey, M. Gerner, C. D. Hammerbeck, F. Popescu, and Z. Xiao. 2006. Signals required for programming effector and memory development by CD8⫹ T cells. Immunol. Rev. 211: 81–92. 16. Tough, D. F., and J. Sprent. 1994. Turnover of naive- and memory-phenotype T cells. J. Exp. Med. 179: 1127–1135. 17. Peixoto, A., C. Evaristo, I. Munitic, M. Monteiro, A. Charbit, B. Rocha, and H. Veiga-Fernandes. 2007. CD8 single-cell gene coexpression reveals three different effector types present at distinct phases of the immune response. J. Exp. Med. 204: 1193–1205.

1314

BRIEF REVIEW: MECHANISMS FOR GENERATING EFFECTOR CD8 T CELL DIVERSITY

18. Sallusto, F., D. Lenig, R. Forster, M. Lipp, and A. Lanzavecchia. 1999. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401: 708 –712. 19. Wherry, E. J., V. Teichgraber, T. C. Becker, D. Masopust, S. M. Kaech, R. Antia, U. H. von Andrian, and R. Ahmed. 2003. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nat. Immunol. 4: 225–234. 20. Masopust, D., V. Vezys, E. J. Wherry, D. L. Barber, and R. Ahmed. 2006. Cutting edge: gut microenvironment promotes differentiation of a unique memory CD8 T cell population. J. Immunol. 176: 2079 –2083. 21. Wille-Reece, U., B. J. Flynn, K. Lore, R. A. Koup, A. P. Miles, A. Saul, R. M. Kedl, J. J. Mattapallil, W. R. Weiss, M. Roederer, and R. A. Seder. 2006. Toll-like receptor agonists influence the magnitude and quality of memory T cell responses after primeboost immunization in nonhuman primates. J. Exp. Med. 203: 1249 –1258. 22. Schluns, K. S., W. C. Kieper, S. C. Jameson, and L. Lefrancois. 2000. Interleukin-7 mediates the homeostasis of naive and memory CD8 T cells in vivo. Nat. Immunol. 1: 426 – 432. 23. Kaech, S. M., J. T. Tan, E. J. Wherry, B. T. Konieczny, C. D. Surh, and R. Ahmed. 2003. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat. Immunol. 4: 1191–1198. 24. Barber, D. L., E. J. Wherry, D. Masopust, B. Zhu, J. P. Allison, A. H. Sharpe, G. J. Freeman, and R. Ahmed. 2006. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439: 682– 687. 25. Grayson, J. M., A. J. Zajac, J. D. Altman, and R. Ahmed. 2000. Cutting edge: increased expression of Bcl-2 in antigen-specific memory CD8⫹ T cells. J. Immunol. 164: 3950 –3954. 26. de Bree, G. J., E. M. van Leeuwen, T. A. Out, H. M. Jansen, R. E. Jonkers, and R. A. van Lier. 2005. Selective accumulation of differentiated CD8⫹ T cells specific for respiratory viruses in the human lung. J. Exp. Med. 202: 1433–1442. 27. Ibegbu, C. C., Y. X. Xu, W. Harris, D. Maggio, J. D. Miller, and A. P. Kourtis. 2005. Expression of killer cell lectin-like receptor G1 on antigen-specific human CD8⫹ T lymphocytes during active, latent, and resolved infection and its relation with CD57. J. Immunol. 174: 6088 – 6094. 28. McMahon, C. W., A. J. Zajac, A. M. Jamieson, L. Corral, G. E. Hammer, R. Ahmed, and D. H. Raulet. 2002. Viral and bacterial infections induce expression of multiple NK cell receptors in responding CD8⫹ T cells. J. Immunol. 169: 1444 –1452. 29. Hikono, H., J. E. Kohlmeier, S. Takamura, S. T. Wittmer, A. D. Roberts, and D. L. Woodland. 2007. Activation phenotype, rather than central- or effector-memory phenotype, predicts the recall efficacy of memory CD8⫹ T cells. J. Exp. Med. 204: 1625–1636. 30. Joshi, N. S., W. Cui, A. Chandele, H. K. Lee, D. R. Urso, J. Hagman, L. Gapin, and S. M. Kaech. 2007. Inflammation directs memory precursor and short-lived effector CD8⫹ T cell fates via the graded expression of T-bet transcription factor. Immunity 27: 281–295. 31. Laouar, A., M. Manocha, M. Wan, H. Yagita, R. A. van Lier, and N. Manjunath. 2007. Cutting Edge: distinct NK receptor profiles are imprinted on CD8 T cells in the mucosa and periphery during the same antigen challenge: role of tissue-specific factors. J. Immunol. 178: 652– 656. 32. Voehringer, D., M. Koschella, and H. Pircher. 2002. Lack of proliferative capacity of human effector and memory T cells expressing killer cell lectinlike receptor G1 (KLRG1). Blood 100: 3698 –3702. 33. Voehringer, D., C. Blaser, P. Brawand, D. H. Raulet, T. Hanke, and H. Pircher. 2001. Viral infections induce abundant numbers of senescent CD8 T cells. J. Immunol. 167: 4838 – 4843. 34. Masopust, D., V. Vezys, A. L. Marzo, and L. Lefrancois. 2001. Preferential localization of effector memory cells in nonlymphoid tissue. Science 291: 2413–2417. 35. Jenkins, M. R., K. Kedzierska, P. C. Doherty, and S. J. Turner. 2007. Heterogeneity of effector phenotype for acute phase and memory influenza A virus-specific CTL. J. Immunol. 179: 64 –70. 36. Marzo, A. L., K. D. Klonowski, A. Le Bon, P. Borrow, D. F. Tough, and L. Lefrancois. 2005. Initial T cell frequency dictates memory CD8⫹ T cell lineage commitment. Nat Immunol. 6: 793–799. 37. Badovinac, V. P., J. S. Haring, and J. T. Harty. 2007. Initial T cell receptor transgenic cell precursor frequency dictates critical aspects of the CD8⫹ T cell response to infection. Immunity 26: 827– 841. 38. Lang, K. S., M. Recher, A. A. Navarini, N. L. Harris, M. Lohning, T. Junt, H. C. Probst, H. Hengartner, and R. M. Zinkernagel. 2005. Inverse correlation between IL-7 receptor expression and CD8 T cell exhaustion during persistent antigen stimulation. Eur. J. Immunol. 35: 738 –745. 39. Manjunath, N., P. Shankar, J. Wan, W. Weninger, M. A. Crowley, K. Hieshima, T. A. Springer, X. Fan, H. Shen, J. Lieberman, and U. H. von Andrian. 2001. Effector differentiation is not prerequisite for generation of memory cytotoxic T lymphocytes. J. Clin. Invest. 108: 871– 878. 40. Vezys, V., D. Masopust, C. C. Kemball, D. L. Barber, L. A. O’Mara, C. P. Larsen, T. C. Pearson, R. Ahmed, and A. E. Lukacher. 2006. Continuous recruitment of naive T cells contributes to heterogeneity of antiviral CD8 T cells during persistent infection. J. Exp. Med. 203: 2263–2269. 41. Badovinac, V. P., and J. T. Harty. 2007. Manipulating the rate of memory CD8⫹ T cell generation after acute infection. J. Immunol. 179: 53– 63. 42. Badovinac, V. P., K. A. Messingham, A. Jabbari, J. S. Haring, and J. T. Harty. 2005. Accelerated CD8⫹ T-cell memory and prime-boost response after dendritic-cell vaccination. Nat. Med. 11: 748 –756. 43. Badovinac, V. P., B. B. Porter, and J. T. Harty. 2004. CD8⫹ T cell contraction is controlled by early inflammation. Nat. Immunol. 5: 809 – 817. 44. Marzo, A. L., H. Yagita, and L. Lefrancois. 2007. Cutting edge: migration to nonlymphoid tissues results in functional conversion of central to effector memory CD8 T cells. J. Immunol. 179: 36 – 40.

45. Sarkar, S., V. Teichgraber, V. Kalia, A. Polley, D. Masopust, L. E. Harrington, R. Ahmed, and E. J. Wherry. 2007. Strength of stimulus and clonal competition impact the rate of memory CD8 T cell differentiation. J. Immunol. 179: 6704 – 6714. 46. Sierro, S., R. Rothkopf, and P. Klenerman. 2005. Evolution of diverse antiviral CD8⫹ T cell populations after murine cytomegalovirus infection. Eur. J. Immunol. 35: 1113–1123. 47. Saparov, A., F. H. Wagner, R. Zheng, J. R. Oliver, H. Maeda, R. D. Hockett, and C. T. Weaver. 1999. Interleukin-2 expression by a subpopulation of primary T cells is linked to enhanced memory/effector function. Immunity 11: 271–280. 48. Precopio, M. L., M. R. Betts, J. Parrino, D. A. Price, E. Gostick, D. R. Ambrozak, T. E. Asher, D. C. Douek, A. Harari, G. Pantaleo, et al. 2007. Immunization with vaccinia virus induces polyfunctional and phenotypically distinctive CD8⫹ T cell responses. J. Exp. Med. 204: 1405–1416. 49. Huster, K. M., V. Busch, M. Schiemann, K. Linkemann, K. M. Kerksiek, H. Wagner, and D. H. Busch. 2004. Selective expression of IL-7 receptor on memory T cells identifies early CD40L-dependent generation of distinct CD8⫹ memory T cell subsets. Proc. Natl. Acad. Sci. USA 101: 5610 –5615. 50. Lacombe, M. H., M. P. Hardy, J. Rooney, and N. Labrecque. 2005. IL-7 receptor expression levels do not identify CD8⫹ memory T lymphocyte precursors following peptide immunization. J. Immunol. 175: 4400 – 4407. 51. Castellino, F., and R. N. Germain. 2007. Chemokine-guided CD4⫹ T cell help enhances generation of IL-6RalphahighIL-7R␣ high prememory CD8⫹ T cells. J. Immunol. 178: 778 –787. 52. Hand, T. W., M. Morre, and S. M. Kaech. 2007. Expression of IL-7 receptor ␣ is necessary but not sufficient for the formation of memory CD8 T cells during viral infection. Proc. Natl. Acad. Sci. USA 104: 11730 –11735. 53. van Leeuwen, E. M., G. J. de Bree, I. J. ten Berge, and R. A. van Lier. 2006. Human virus-specific CD8⫹ T cells: diversity specialists. Immunol. Rev. 211: 225–235. 54. Akbar, A. N., and J. M. Fletcher. 2005. Memory T cell homeostasis and senescence during aging. Curr. Opin. Immunol. 17: 480 – 485. 55. Roberts, A. D., K. H. Ely, and D. L. Woodland. 2005. Differential contributions of central and effector memory T cells to recall responses. J. Exp. Med. 202: 123–133. 56. Jacob, J., and D. Baltimore. 1999. Modelling T-cell memory by genetic marking of memory T cells in vivo. Nature 399: 593–597. 57. Opferman, J. T., B. T. Ober, and P. G. Ashton-Rickardt. 1999. Linear differentiation of cytotoxic effectors into memory T lymphocytes. Science 283: 1745–1748. 58. Lefrancois, L., A. Marzo, and K. Williams. 2003. Sustained response initiation is required for T cell clonal expansion but not for effector or memory development in vivo. J. Immunol. 171: 2832–2839. 59. Curtsinger, J. M., D. C. Lins, and M. F. Mescher. 2003. Signal 3 determines tolerance versus full activation of naive CD8 T cells: dissociating proliferation and development of effector function. J. Exp. Med. 197: 1141–1151. 60. Lauvau, G., S. Vijh, P. Kong, T. Horng, K. Kerksiek, N. Serbina, R. A. Tuma, and E. G. Pamer. 2001. Priming of memory but not effector CD8 T cells by a killed bacterial vaccine. Science 294: 1735–1739. 61. Bachmann, M. F., P. Wolint, K. Schwarz, P. Jager, and A. Oxenius. 2005. Functional properties and lineage relationship of CD8⫹ T cell subsets identified by expression of IL-7 receptor ␣ and CD62L. J. Immunol. 175: 4686 – 4696. 62. Williams, M. A., A. J. Tyznik, and M. J. Bevan. 2006. Interleukin-2 signals during priming are required for secondary expansion of CD8⫹ memory T cells. Nature 441: 890 – 893. 63. Intlekofer, A. M., N. Takemoto, C. Kao, A. Banerjee, F. Schambach, J. K. Northrop, H. Shen, E. J. Wherry, and S. L. Reiner. 2007. Requirement for T-bet in the aberrant differentiation of unhelped memory CD8⫹ T cells. J. Exp. Med. 204: 2015–2021. 64. Ahmed, R., and D. Gray. 1996. Immunological memory and protective immunity: understanding their relation. Science 272: 54 – 60. 65. Gattinoni, L., D. J. Powell, Jr., S. A. Rosenberg, and N. P. Restifo. 2006. Adoptive immunotherapy for cancer: building on success. Nat. Rev. Immunol. 6: 383–393. 66. Sallusto, F., J. Geginat, and A. Lanzavecchia. 2004. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu. Rev. Immunol. 22: 745–763. 67. D’Souza, W. N., and S. M. Hedrick. 2006. Cutting edge: latecomer CD8 T cells are imprinted with a unique differentiation program. J. Immunol. 177: 777–781. 68. Sarkar, S., V. Kalia, W. N. Haining, B. T. Konieczny, S. Subramaniam, and R. Ahmed. Functional and genomic profiling of effector CD8 T cell subsets with distinct fates. J. Exp. Med. In press. 69. Williams, M. A., and M. J. Bevan. 2004. Shortening the infectious period does not alter expansion of CD8 T cells but diminishes their capacity to differentiate into memory cells. J. Immunol. 173: 6694 – 6702. 70. Prlic, M., G. Hernandez-Hoyos, and M. J. Bevan. 2006. Duration of the initial TCR stimulus controls the magnitude but not functionality of the CD8⫹ T cell response. J. Exp. Med. 203: 2135–2143. 71. Wherry, E. J., K. A. Puorro, A. Porgador, and L. C. Eisenlohr. 1999. The induction of virus-specific CTL as a function of increasing epitope expression: responses rise steadily until excessively high levels of epitope are attained. J. Immunol. 163: 3735–3745. 72. Kaech, S. M., and R. Ahmed. 2001. Memory CD8⫹ T cell differentiation: initial antigen encounter triggers a developmental program in naive cells. Nat. Immunol. 2: 415– 422. 73. Gett, A. V., F. Sallusto, A. Lanzavecchia, and J. Geginat. 2003. T cell fitness determined by signal strength. Nat. Immunol. 4: 355–360. 74. Lanzavecchia, A., and F. Sallusto. 2002. Progressive differentiation and selection of the fittest in the immune response. Nat. Rev. Immunol. 2: 982–987. 75. Chang, J. T., V. R. Palanivel, I. Kinjyo, F. Schambach, A. M. Intlekofer, A. Banerjee, S. A. Longworth, K. E. Vinup, P. Mrass, J. Oliaro, et al. 2007. Asymmetric T lymphocyte division in the initiation of adaptive immune responses. Science 315: 1687–1691.

The Journal of Immunology 76. Xiao, Z., J. M. Curtsinger, M. Prlic, S. C. Jameson, and M. F. Mescher. 2007. The CD8 T cell response to vaccinia virus exhibits site-dependent heterogeneity of functional responses. Int. Immunol. 19: 733–743. 77. Kedzierska, K., N. L. La Gruta, S. J. Turner, and P. C. Doherty. 2006. Establishment and recall of CD8⫹ T-cell memory in a model of localized transient infection. Immunol. Rev. 211: 133–145. 78. Bengsch, B., H. C. Spangenberg, N. Kersting, C. Neumann-Haefelin, E. Panther, F. von Weizsacker, H. E. Blum, H. Pircher, and R. Thimme. 2007. Analysis of CD127 and KLRG1 expression on hepatitis C virus-specific CD8⫹ T cells reveals the existence of different memory T-cell subsets in the peripheral blood and liver. J. Virol. 81: 945–953. 79. Weaver, C. T., R. D. Hatton, P. R. Mangan, and L. E. Harrington. 2007. IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu. Rev. Immunol. 25: 821– 852. 80. Sad, S., R. Marcotte, and T. R. Mosmann. 1995. Cytokine-induced differentiation of precursor mouse CD8⫹ T cells into cytotoxic CD8⫹ T cells secreting Th1 or Th2 cytokines. Immunity 2: 271–279. 81. Whitmire, J. K., J. T. Tan, and J. L. Whitton. 2005. Interferon-␥ acts directly on CD8⫹ T cells to increase their abundance during virus infection. J. Exp. Med. 201: 1053–1059. 82. Hernandez, J., S. Aung, K. Marquardt, and L. A. Sherman. 2002. Uncoupling of proliferative potential and gain of effector function by CD8⫹ T cells responding to self-antigens. J. Exp. Med. 196: 323–333. 83. Kolumam, G. A., S. Thomas, L. J. Thompson, J. Sprent, and K. Murali-Krishna. 2005. Type I interferons act directly on CD8 T cells to allow clonal expansion and memory formation in response to viral infection. J. Exp. Med. 202: 637– 650. 84. Badovinac, V. P., A. R. Tvinnereim, and J. T. Harty. 2000. Regulation of antigenspecific CD8⫹ T cell homeostasis by perforin and interferon-␥. Science 290: 1354 –1358. 85. Pearce, E. L., and H. Shen. 2007. Generation of CD8 T cell memory is regulated by IL-12. J. Immunol. 179: 2074 –2081. 86. Takemoto, N., A. M. Intlekofer, J. T. Northrup, E. J. Wherry, and S. L. Reiner. 2006. Cutting Edge: IL-12 inversely regulates T-bet and eomesodermin expression during pathogen-induced CD8⫹ T cell differentiation. J. Immunol. 177: 7515–7519. 87. Thompson, L. J., G. A. Kolumam, S. Thomas, and K. Murali-Krishna. 2006. Innate inflammatory signals induced by various pathogens differentially dictate the IFN-I dependence of CD8 T cells for clonal expansion and memory formation. J. Immunol. 177: 1746 –1754. 88. D’Souza, W. N., K. S. Schluns, D. Masopust, and L. Lefrancois. 2002. Essential role for IL-2 in the regulation of antiviral extralymphoid CD8 T cell responses. J. Immunol. 168: 5566 –5572. 89. Mitchell, T. C., D. Hildeman, R. M. Kedl, T. K. Teague, B. C. Schaefer, J. White, Y. Zhu, J. Kappler, and P. Marrack. 2001. Immunological adjuvants promote activated T cell survival via induction of Bcl-3. Nat. Immunol. 2: 397– 402. 90. Valenzuela, J. O., C. D. Hammerbeck, and M. F. Mescher. 2005. Cutting edge: Bcl-3 up-regulation by signal 3 cytokine (IL-12) prolongs survival of antigen-activated CD8 T cells. J. Immunol. 174: 600 – 604. 91. Szabo, S. J., B. M. Sullivan, C. Stemmann, A. R. Satoskar, B. P. Sleckman, and L. H. Glimcher. 2002. Distinct effects of T-bet in TH1 lineage commitment and IFN-␥ production in CD4 and CD8 T cells. Science 295: 338 –342. 92. Pearce, E. L., A. C. Mullen, G. A. Martins, C. M. Krawczyk, A. S. Hutchins, V. P. Zediak, M. Banica, C. B. DiCioccio, D. A. Gross, C. A. Mao, et al. 2003. Control of effector CD8⫹ T cell function by the transcription factor Eomesodermin. Science 302: 1041–1043. 93. Whitmire, J. K., B. Eam, N. Benning, and J. L. Whitton. 2007. Direct interferon-␥ signaling dramatically enhances CD4⫹ and CD8⫹ T cell memory. J. Immunol. 179: 1190 –1197. 94. Huntington, N. D., H. Tabarias, K. Fairfax, J. Brady, Y. Hayakawa, M. A. Degli-Esposti, M. J. Smyth, D. M. Tarlinton, and S. L. Nutt. 2007. NK cell maturation and peripheral homeostasis is associated with KLRG1 up-regulation. J. Immunol. 178: 4764 – 4770. 95. Robbins, S. H., M. S. Tessmer, T. Mikayama, and L. Brossay. 2004. Expansion and contraction of the NK cell compartment in response to murine cytomegalovirus infection. J. Immunol. 173: 259 –266. 96. Robbins, S. H., M. S. Tessmer, L. Van Kaer, and L. Brossay. 2005. Direct effects of T-bet and MHC class I expression, but not STAT1, on peripheral NK cell maturation. Eur. J. Immunol. 35: 757–765. 97. Townsend, M. J., A. S. Weinmann, J. L. Matsuda, R. Salomon, P. J. Farnham, C. A. Biron, L. Gapin, and L. H. Glimcher. 2004. T-bet regulates the terminal maturation and homeostasis of NK and Valpha14i NKT cells. Immunity 20: 477– 494.

1315 98. Foulds, K. E., M. J. Rotte, and R. A. Seder. 2006. IL-10 is required for optimal CD8 T cell memory following Listeria monocytogenes infection. J. Immunol. 177: 2565–2574. 99. Biswas, P. S., V. Pedicord, A. Ploss, E. Menet, I. Leiner, and E. G. Pamer. 2007. Pathogen-specific CD8 T cell responses are directly inhibited by IL-10. J. Immunol. 179: 4520 – 4528. 100. Yokota, Y., A. Mansouri, S. Mori, S. Sugawara, S. Adachi, S. Nishikawa, and P. Gruss. 1999. Development of peripheral lymphoid organs and natural killer cells depends on the helix-loop-helix inhibitor Id2. Nature 397: 702–706. 101. Cannarile, M. A., N. A. Lind, R. Rivera, A. D. Sheridan, K. A. Camfield, B. B. Wu, K. P. Cheung, Z. Ding, and A. W. Goldrath. 2006. Transcriptional regulator Id2 mediates CD8⫹ T cell immunity. Nat. Immunol. 7: 1317–1325. 102. Shapiro-Shelef, M., K. I. Lin, L. J. McHeyzer-Williams, J. Liao, M. G. McHeyzer-Williams, and K. Calame. 2003. Blimp-1 is required for the formation of immunoglobulin secreting plasma cells and pre-plasma memory B cells. Immunity 19: 607– 620. 103. Kallies, A., E. D. Hawkins, G. T. Belz, D. Metcalf, M. Hommel, L. M. Corcoran, P. D. Hodgkin, and S. L. Nutt. 2006. Transcriptional repressor Blimp-1 is essential for T cell homeostasis and self-tolerance. Nat. Immunol. 7: 466 – 474. 104. Martins, G. A., L. Cimmino, M. Shapiro-Shelef, M. Szabolcs, A. Herron, E. Magnusdottir, and K. Calame. 2006. Transcriptional repressor Blimp-1 regulates T cell homeostasis and function. Nat. Immunol. 7: 457– 465. 105. Manders, P. M., P. J. Hunter, A. I. Telaranta, J. M. Carr, J. L. Marshall, M. Carrasco, Y. Murakami, M. J. Palmowski, V. Cerundolo, S. M. Kaech, et al. 2005. BCL6b mediates the enhanced magnitude of the secondary response of memory CD8⫹ T lymphocytes. Proc. Natl. Acad. Sci. USA 102: 7418 –7425. 106. Ichii, H., A. Sakamoto, Y. Kuroda, and T. Tokuhisa. 2004. Bcl6 acts as an amplifier for the generation and proliferative capacity of central memory CD8⫹ T cells. J. Immunol. 173: 883– 891. 107. Heffner, M., and D. T. Fearon. 2007. Loss of T cell receptor-induced Bmi-1 in the KLRG1⫹ senescent CD8⫹ T lymphocyte. Proc. Natl. Acad. Sci. USA 104: 13414 –13419. 108. Becker, T. C., E. J. Wherry, D. Boone, K. Murali-Krishna, R. Antia, A. Ma, and R. Ahmed. 2002. Interleukin 15 is required for proliferative renewal of virus-specific memory CD8 T cells. J. Exp. Med. 195: 1541–1548. 109. Osborne, L. C., S. Dhanji, J. W. Snow, J. J. Priatel, M. C. Ma, M. J. Miners, H. S. Teh, M. A. Goldsmith, and N. Abraham. 2007. Impaired CD8 T cell memory and CD4 T cell primary responses in IL-7R ␣ mutant mice. J. Exp. Med. 204: 619 – 631. 110. Goldrath, A. W., P. V. Sivakumar, M. Glaccum, M. K. Kennedy, M. J. Bevan, C. Benoist, D. Mathis, and E. A. Butz. 2002. Cytokine requirements for acute and Basal homeostatic proliferation of naive and memory CD8⫹ T cells. J. Exp. Med. 195: 1515–1522. 111. Sun, J. C., S. M. Lehar, and M. J. Bevan. 2006. Augmented IL-7 signaling during viral infection drives greater expansion of effector T cells but does not enhance memory. J. Immunol. 177: 4458 – 4463. 112. Tripathi, P., T. C. Mitchell, F. Finkelman, and D. A. Hildeman. 2007. Cutting Edge: Limiting amounts of IL-7 do not control contraction of CD4⫹ T cell responses. J. Immunol. 178: 4027– 4031. 113. Petschner, F., C. Zimmerman, A. Strasser, D. Grillot, G. Nunez, and H. Pircher. 1998. Constitutive expression of Bcl-xL or Bcl-2 prevents peptide antigen-induced T cell deletion but does not influence T cell homeostasis after a viral infection. Eur. J. Immunol. 28: 560 –569. 114. Grayson, J. M., A. E. Weant, B. C. Holbrook, and D. Hildeman. 2006. Role of Bim in regulating CD8⫹ T-cell responses during chronic viral infection. J. Virol. 80: 8627– 8638. 115. Hildeman, D. A., Y. Zhu, T. C. Mitchell, P. Bouillet, A. Strasser, J. Kappler, and P. Marrack. 2002. Activated T cell death in vivo mediated by proapoptotic Bcl-2 family member Bim. Immunity 16: 759 –767. 116. Pellegrini, M., G. Belz, P. Bouillet, and A. Strasser. 2003. Shutdown of an acute T cell immune response to viral infection is mediated by the proapoptotic Bcl-2 homology 3-only protein Bim. Proc. Natl. Acad. Sci. USA 100: 14175–14180. 117. Wojciechowski, S., M. B. Jordan, Y. Zhu, J. White, A. J. Zajac, and D. A. Hildeman. 2006. Bim mediates apoptosis of CD127lo effector T cells and limits T cell memory. Eur. J. Immunol. 36: 1694 –1706. 118. Liu, N., T. Phillips, M. Zhang, Y. Wang, J. T. Opferman, R. Shah, and P. G. Ashton-Rickardt. 2004. Serine protease inhibitor 2A is a protective factor for memory T cell development. Nat. Immunol. 5: 919 –926. 119. Hou, S., L. Hyland, K. W. Ryan, A. Portner, and P. C. Doherty. 1994. Virus-specific CD8⫹ T-cell memory determined by clonal burst size. Nature 369: 652– 654.