当前位置: 首页 > 医学版 > 期刊论文 > 基础医学 > 感染与免疫杂志 > 2006年 > 第4期 > 正文
编号:11255510
Protective CD8+ T Cells Induced by Malaria Sporozoites Do Not Undergo Modulation of Interleukin-7 Receptor Expression
     Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205

    Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Campus Ciudad Universitaria Cantoblanco, 28049 Madrid, Spain

    ABSTRACT

    CD8+ T cells induced by Plasmodium yoelii sporozoites develop into protective memory cells without undergoing changes in interleukin-7 receptor (IL-7R) expression, differing from the development of memory CD8+ T cells against viruses, which is associated with enhanced IL-7R expression. This suggests a microbe-dependent diversity in the signals determining the development of memory populations.

    TEXT

    Memory CD8+ T cells develop following activation of nave CD8+ T cells induced by microbial infection and establish a pool of cells capable of responding in greater numbers and with greater efficacy than nave precursors (1). The formation of this stable pool of memory CD8+ T cells is vital to establish protective immunity upon reexposure to infectious intracellular pathogens. While significant advances have been made in our understanding of the induction of CD8+ T-cell responses, the molecular mechanisms controlling the development of memory cells and the markers that identify cells that may undergo memory differentiation still remain poorly characterized. A recent study using CD8+ T cells specific for lymphocytic choriomeningitis virus (LCMV) showed that expression of interleukin-7 receptor (IL-7R, CD127) among a small percentage of CD8+ T-cell effectors identified those cells that would become memory cells (4). Furthermore, nave mice that received IL-7Rhigh CD8+ T cells from immunized mice 11 days after immunization went on to develop into memory cells, whereas those that received IL-7Rlow CD8+ T cells did not. These observations led the authors to conclude that the selective expression of IL-7R identified memory cell precursors, thus suggesting that this marker may be useful in predicting the number of memory T cells generated after infection or immunization (4). Given this finding, we characterized the expression of IL-7R among CD8+ T cells in a rodent malaria model. Numerous studies have demonstrated that the induction of effector and memory CD8+ T cells specific for the pre-erythrocytic stage of Plasmodium is critical to establish protective immunity against malaria parasite infection (5, 7, 9, 10). The use of a malaria T-cell receptor (TCR) transgenic system, in which most CD8+ T cells express TCR specific for the H2Kd-restricted epitope SYVPSAEQI from the circumsporozoite protein of Plasmodium yoelii, has provided new insights into the development and tissue distribution of memory cells. We have recently shown that IL-4-IL-4R interactions are critical to the development and maintenance of protective memory CD8+ T cells in nonlymphoid tissue (3, 6).

    Using this P. yoelii-specific TCR transgenic model, we studied the modulation of IL-7R expression after immunization with either P. yoelii sporozoites or recombinant vaccinia virus expressing the parasite epitope (rec-VV) (8). In this system, effector and memory CD8+ T cells express the SYVPSAEQI-specific TCR, which will have the same affinity and strength for the recognition of peptide-major histocompatibility complex complexes expressed after infection with the different microbes.

    To examine the expression of activated IL-7R on T cells, we used an in vivo system in which TCR-transgenic CD8+ T cells were adoptively transferred to normal mice, which were then immunized with either irradiated P. yoelii sporozoites or rec-VV. In mice immunized with rec-VV, antigen-specific CD8+ T cells expressed lower levels of IL-7R at 8 days postimmunization compared to nave cells (Fig. 1A). As the transition from effector to memory cells evolved, the expression of IL-7R increased on the antigen-specific CD8+ T cells, and by day 15, the majority of the cells became IL-7Rhigh (Fig. 1A). Thus, the response elicited by rec-VV regarding the expression of IL-7R on effector and memory CD8+ T cells follows the pattern described for CD8+ T cells after infection with LCMV. In contrast to the shifting pattern of IL-7R expression after infection with this virus, CD8+ T cells from mice immunized with irradiated sporozoites exhibited no modulation of IL-7R expression throughout the development of this T-cell response (Fig. 1A). Indeed, the expression of IL-7R on these cells remained unchanged throughout the effector phase and continued through the memory transition phase at levels similar to those found in nave cells. Importantly, this lack of modulation in the surface expression of IL-7R in parasite-immunized mice was not due to a lack of activation of these cells, as both immunogens were equally efficient at driving CD8+ T-cell activation, because essentially all SYVPSAEQI-specific CD8+ T cells induced by either sporozoites or rec-VV expressed identical, high levels of CD44, the most reliable surface marker of cell activation (Fig. 1B).

    We wished to confirm that the cells present 20 days after irradiated-sporozoite immunization represent a bona fide memory population despite their lack of increased IL-7R expression. Our previous work has shown that irradiated sporozoites induce a classical pattern of activation, expansion, and contraction in the CD8 T-cell response, which leaves a residual population of memory cells. These cells express a surface phenotype typical of antigen-experienced memory cells with respect to CD11a, CD11b, CD11c, CD25, CD27, CD31, CD45RB, CD49d, CD62L, CD69, CD71, CD122, CCR5, CCR7, P-selectin, and granzyme B (6). For all these markers, the phenotypes of parasite-induced memory cells and rec-VV-induced cells are identical (Fig. 2). The notable exception to this is IL-7R. We wanted to know if this difference in IL-7R expression marked some wider defect in the function of these cells. To address this question, we performed adoptive transfer experiments in which nave mice received equal numbers of memory CD8+ T cells obtained from mice immunized with either irradiated sporozoites or rec-VV. Memory CD8+ T cells used in these experiments were obtained from spleens 25 days after immunization and were purified by depleting CD4+ T and B cells by magnetic sorting, as described previously (6). After adoptive transfer into nave mice, the protective capacity of these cells was assessed by determining their ability to inhibit the development of the parasites in the liver, measured 34 h postchallenge using real-time PCR (2, 6). The results from these protection experiments showed that CD8+ T cells primed by either rec-VV or irradiated sporozoites inhibited parasite development with equivalent efficacies and followed a similar dependence on effector T-cell frequency (Fig. 3). Thus, the differential expression of IL-7R on memory antigen-specific CD8+ T cells did not affect their protective qualities.

    Taken together, our data and previous work suggest that memory cells can develop by following different pathways of differentiation. Despite the fact that CD8+ T cells responding to sporozoites and vaccinia virus may differ in their requirements for IL-7 during differentiation, the cells induced by both microbes were able to protect against live sporozoite challenge. Thus, while it is likely that the expression of IL-7R is important for memory development in certain situations, IL-7R cannot be considered a universal marker for protective immunity. It is perhaps not unexpected that CD8+ T-cell responses to the same epitope may follow different activation pathways. The innate immune responses to sporozoites and vaccinia virus may vary widely in terms of the stimulation of Toll-like receptors, the role of dendritic cells and NK cells, production of cytokines, and the strength of costimulation. All these things may alter the development of the acquired immune response. The present results indicate that multiple mechanisms may exist that lead to the development of memory CD8+ T cells and provide insights into the relationships between pathogens and their ability to generate lymphocyte memory.

    ACKNOWLEDGMENTS

    This work was supported by National Institutes of Health grant AI44375.

    Flow cytometry was performed in the Becton Dickinson Immune Function Laboratory at the Johns Hopkins Bloomberg School of Public Health.

    We have no conflicting financial interests.

    REFERENCES

    1. Ahmed, R., and D. Gray. 1996. Immunological memory and protective immunity: understanding their relation. Science 272:54-60.

    2. Bruna-Romero, O., J. C. Hafalla, G. Gonzalez-Aseguinolaza, G. Sano, M. Tsuji, and F. Zavala. 2001. Detection of malaria liver-stages in mice infected through the bite of a single Anopheles mosquito using a highly sensitive real-time PCR. Int. J. Parasitol. 31:1499-1502.

    3. Carvalho, L. H., G. Sano, J. C. Haffala, A. Morrot, M. A. Curotto de Lafaille, and F. Zavala. 2002. IL-4-secreting CD4+ T cells are crucial to the development of CD8+ T-cell responses against malaria liver stages. Nat. Med. 8:166-170.

    4. 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 CD8 T cells that give rise to long-lived memory cells. Nat. Immunol. 4:1191-1198.

    5. Morrot, A., and F. Zavala. 2002. Effector and memory CD8+ T cells as seen in immunity to malaria. Immunol. Rev. 201:291-303.

    6. Morrot, A., J. C. Hafalla, I. A. Cockburn, L. H. Carvalho, and F. Zavala. 2005. IL-4 receptor expression on CD8+ T cells is required for the development of protective memory responses against liver stages of malaria parasites. J. Exp. Med. 202:551-560.

    7. Rodrigues, M. M., A. S. Cordey, G. Arreaza, G. Corradin, P. Romero, J. L. Maryanski, R. S. Nussenzweig, and F. Zavala. 1991. CD8+ cytolytic T cell clones derived against the Plasmodium yoelii circumsporozoite protein protect against malaria. Int. Immunol. 3:982-987.

    8. Rodrigues, M., S. Li, K. Murata, D. Rodriguez, J. R. Rodriguez, I. Bacik, J. R. Bennink, J. W. Yewdell, A. Garcia-Sastre, R. S. Nussenzweig, M. Esteban, P. Palese, and F. Zavala. 1994. Influenza and vaccinia viruses expressing malaria CD8+ T and B cell epitopes. Comparison of their immunogenicity and capacity to induce protective immunity. J. Immunol. 153:4636-4648.

    9. Schofield, L., J. Villaquiran, A. Ferreira, H. Schellekens, R. Nussenzweig, and V. Nussenzweig. 1987. Gamma interferon, CD8+ T cells and antibodies required for immunity to malaria sporozoites. Nature 330:664-666.

    10. Weiss, W. R., M. Sedegah, R. L. Beaudoin, L. H. Miller, and M. F. Good. 1988. CD8+ T cells (cytotoxic/supressors) are required for protection in mice immunized with malaria sporozoites. Proc. Natl. Acad. Sci. USA 85:573-576.(Alexandre Morrot, Ian A. )