Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Aug 11.
Published in final edited form as: Curr Opin Immunol. 2007 Jul 20;19(5):516–521. doi: 10.1016/j.coi.2007.05.005

Apoptosis and the homeostatic control of immune responses

David Hildeman 1, Trine Jorgensen 2, John Kappler 2,3, Philippa Marrack 2,3
PMCID: PMC4127626  NIHMSID: NIHMS486102  PMID: 17644328

Introduction

During infections, antigen specific cells can increase to become a huge proportion of all the lymphocytes in the infected animals (> than 50% of all CD8 T cells can be specific for a single virus for example [1]). However, the antigen activated T and B cells die in large numbers as immune responses wane. This death allows the host to maintain its numbers of lymphocytes at reasonable levels in spite of the fact that the animal probably confronts hundreds of different infectious organisms in its lifetime.

Activated T cell death is readily observed in vivo and can only be subverted, to some extent, by life prolonging reagents such as adjuvants. In vitro, experiments have attempted to mimic the death of activated T cells through repetitive TCR stimulation. It is our view that these experiments have to be interpreted with caution, however. For example, early in vitro experiments suggested that Fas/Fas ligand interaction was a driving force behind activated T cell death [24]. However, this has turned out not to be the case in vivo, where other mechanisms may supercede the death receptors [5, 6]. Nevertheless, Fas clearly plays a role in control of lymphoproliferation and autoimmune disease. This may be due to non-lymphocyte effects of Fas signaling [7] since recent experiments suggest that Fas and other survival proteins may affect autoimmunity in part via the otherwise quite short lived dendritic cells (DCs). These and other issues are discussed in detail below.

Table 1 is a summary of the expression of mRNA for genes that might be involved in the death of lymphocytes and dendritic cells. Remarkably, the relative expression of these genes is quite similar among the T and B cells at various stages of maturation and activation. As expected, levels of Bcl-2 are decreased with activation [5], while Bim levels were increased slightly even though we have not observed increases in Bim protein levels after in vivo activation [5]. Strikingly, the two most highly induced genes following activation were A1 and survivin (Table 1). As the majority of the activated cells from which the RNA was taken are destined to die, it is unclear why high levels of these anti-apoptotic genes fail to rescue these cells. Perhaps these high levels were important for the survival of proliferating T cells, rather than survival of “post-peak” cells as has been suggested by recent studies [8, 9]. Of all the genes, only the death receptors, Fas and the TNFRs appear to be markedly differently expressed in T and B cells. Dendritic cell gene expression, by contrast, is quite different from that of lymphocytes, with greatly increased expression of the Bcl-2-related pro-apoptotic messangers, such as Puma, NOXA and BNIP3.

Table 1. Expression of Bcl-2 related genes in lymphocytes and dendritic cells.

C57BL/6 mice were either uninjected or injected with Staphylococcal enterotoxin B (SEB) for 48 hours prior to sacrifice. Single cell suspensions from pooled lymph nodes and spleens were stained with antibodies against TCRβ, CD4, CD8, IAb, CD69, and CD25 for na�ve cells (sorted for TCR+CD4+ or TCR+CD8+ and IAb-CD69−CD25−) and Vb8.x CD4, CD8, IAb for SEB activated cells (sorted for Vβ8+CD4+ or Vβ8+CD8+ and IAb-). RNA was extracted and sent to the Gene Microarray core at Cincinnati Children’s Hospital and run on Affymetrix mouse genome 430.2 arrays. Analysis was performed using Genespring software after normalization. Data shown reflect averages of multiple probe sets (whose values were above 50) or single probe sets depending upon the gene.

Naïve
CD4+
T cells
Activated
CD4+ T
cells
Memory
Phenotype
CD4+ T cells
Naïve
CD8+
T cells
Activated
CD8+ T
cells
Memory
Phenotype
CD8+ T
cells
(Follicular)
B cells
Germinal
Center B
cells
Memory
B cells
Plasma
Cells
Bone
marrow
derived
dendritic
cells
Spleen
CD4− CD8−
dendritic
cells
Bcl-2 +++++ ++++ ++ +++++ +++ +++++ ++ ++ +++ ++ +
Bcl-xL ++ ++ +++ + ++ ++++ + + + + +++ +
Mcl-1 +++++ +++++ +++++ +++++ +++++ +++++ +++++ +++++ +++++ +++++ +++++ +++
A1 +++ +++++ +++++ +++ +++++ +++++ +++ ++++ +++ + +++++ +++++
Bim +++ ++++ ++ ++ +++ +++ ++ ++ + ++ +++++ ++
Bad + ++ + + ++ + +
PUMA + + nd + + ++++ ++ nd
NOXA + + nd ++ +++ ++++ ++++ nd
Hrk + +
Bid + + ++ + ++ ++ ++ +++ +
Boo/DIVA +
Bik nd +
NIX +++++ +++++ nd +++++ +++++ +++++ ++++ ++ +++ ++ +++++ +
BNIP3 ++ ++ ++ + + +++ ++ + + +++++
Bax +++ +++ ++++ +++ +++ +++++ + + + + +++++ +++
Bak + ++ ++++ + ++ ++++ ++++ +
Bok ++ + ++ +
FAS +++ +++ ++ +++ +++ ++ ++ + +++
Tnfrsf1a +++ ++ +++ ++ ++ ++ + +++++ ++
Tnfrsf1b +++ ++++ +++++ +++ +++ ++ (+) + +++++ ++

Cells were purified and analyzed on Affymetrix chips. All cells were from mice. Germinal center B cells, Memory B cells and plasma cells were from the spleens of mice immunized with NP-CGG 11-14 days, 10–12 weeks and 7 days previously respectively. Data on T cells are unpublished from our laboratories or from Swanson et al. Immunity 17:605, 2002. Data on B cells are unpublished from our laboratories or from Weissman, I. depositied in the Geo data base, 1/30/06. Data on bone marrow derived dendritic cells are from D.Stetson depositied in the Geo data base 3/1/05. Data on spleen CD4− CD8− dendritic cells are from Chaussabel, deposited in the Geo Data base 6/13/03. To help readers, values are expressed as: −, 0–50; + 51–200; ++, 201–500; +++, 501–1000; ++++, 1001–2000; +++++, >2000.

Life and death of antigen presenting dendritic cells

Dendritic cells, regardless of their subtype, have relatively short half-lives [10, 11]. Nevertheless these are long enough under most circumstances to achieve successful antigen presentation to T cells. The normal life expectancy of DCs is thought to be controlled by balances between Bcl-2-related proteins and not via the action of death receptor proteins such as Fas [1113]. Even so, DCs that express the baculovirus caspase inhibitor, p35 (which primarily blocks death receptors) are resistant to death in vivo and increase in numbers, albeit only after one year of age [14]. Mice containing such DCs have increased numbers of activated T cells and anti-nuclear-antigen antibody production [14]. In contrast, mice whose DCs overexpress a hBcl-2 transgene or lack Bim have increased steady-state levels of DCs in relatively young mice, which can elicit increased T cell responses and autoantibody production [11, 15]. Thus, both death receptors and Bcl-2 family members contribute to the demise of DCs following their activation, suggesting that DCs with extended life times can enhance T and B cell responses.

It is not obvious why longer life expectancies for DCs would promote autoimmunity. The DCs are always present, after all, and are always presenting self peptides, so why should their life spans affect the outcome? One obvious possibility is that activation of DCs or their interactions with activated T cells significantly shortens their lifespan, which in turn effectively removes T cell substrates. Indeed, it has long been known that survival of DCs is curtailed by cytotoxic T cells [16], which probably act both via perforin [17] and Fas [14, 18]. This killing of DCs may also explain the profound immunosuppression observed after certain viral infections such as measles [19]. Conversely, perhaps Fas signaling has an additional effect on DCs, as indicated by induction of chemokine production by DCs stimulated with anti-Fas [20]. If this is so, autoimmunity induced by the DCs in these experiments may be caused by increased DC activity induced by the very signals which are normally dampened by their ability to also drive DC death. Overall, these experiments suggest that the death of DCs (especially activated DCs) may be an important contributor to the resolution of immune responses and to the development of autoimmunity. Certainly this is a subject which we expect to see more of in the next few years.

Life and death of activated B cells

Research over many years has suggested that activated B cells die via Fas (reviewed in [21]). However, the death of some types of activated B cells is affected by the balance of pro and anti-apoptotic members of the Bcl-2 family, for example, Bcl-2 itself (of course!), Bax and Bak and Bim [2227]. BAFF is the principle survival factor for activated B cells and helps to break the unresponsiveness of autoimmune B cells [25, 26]. In this regard, the discussion of the effects of Bcl-2 related proteins on the activities of DCs is quite interesting since DCs are themselves are a potential source of BAFF (reviewed in [28]). Thus, breakage of B cell tolerance in autoimmunity could be due to effects on DCs which, in turn via BAFF, may be manifested on B cells.

The BH3-only molecule, Bim, is critical for apoptotic contraction of T cell responses

The pro-apoptotic BH3-only molecule, Bim, plays a critical role in culling activated effector T cells [5, 6, 2931]. A major factor controlling Bim activation resides not with Bim but with its major antagonist, Bcl-2. At the peak of response, levels of Bcl-2 are significantly decreased within antigen-specific T cells [5, 32, 33]. In unstimulated T cells, Bim is complexed to Bcl-2 [34], the downregulation of Bcl-2 that occurs during T cell activation may result in increased amounts of free Bim. Bim that is unopposed by Bcl-2 could then either directly or indirectly lead to the activation of Bax and/or Bak. Indeed, Bax/Bak conformational changes are not observed in activated Bim−/− T cells [31]. Other factors also may contribute to Bim activation as T cells lacking an adjuvant-induced survival factor, Bcl-3 [35], accumulate cytosolic Bim which may then drive cell death [36]. However, additional processes such as release from microtubules, variation in expression of isoforms or phosophorylation may also affect Bim activity [37, 38]. Further work is required to understand how these mechanisms are integrated following T cell activation in vivo.

Other BH-3 proteins substitute poorly for Bim

If T cells express multiple BH3-only molecules (see Table 1), why does Bim play such a dominant role in T cell death and what is the function of other BH-3-only molecules in T cells? It is possible that other BH3-only molecules may act as “messengers” of apoptotic signals, then transmitted by Bim. Alternatively, as suggested by others, different BH3-only molecules may act as sensors of divergent pro-apoptotic stimuli [39]. For example, Bim appears to protect cells from “growth factor withdrawal” induced death [40], while Puma and Noxa are direct transcriptional targets of the tumor suppressor p53 and are critical for DNA-damage-induced apoptosis [41]. Puma is particularly intriguing because its overall structure, with respect to its arrangement of BH and transmembrane domains, is similar to that of Bim. However, only minor effects, if any, of Puma on activated T cell death have been observed, even in activated T cells that lack Bim [42]. Absence of Noxa likewise has no effect on activated T cell death (in spite of the fact that it is well expressed in these cells, Table 1) indicating that termination of T cell responses is not driven by a DNA-damage response [36]. Overall, Bim appears to be the dominant and largely non-redundant BH3-only molecule critical for apoptotic contraction of T cell responses in vivo.

How do some T cells survive and become memory cells?

Since Bim is critical for activated T cell death, the cells that survive apoptosis to become memory cells must somehow avoid Bim-driven death. One possibility is that the phenomenon is stochastic, and depends on random over-expression of death-avoiding proteins in the activated cells. Alternatively, cells that survive may be those which receive or inherit different signals or proteins after TCR engagement. Such an idea is supported by the increased conversion to memory of daughter cells which are distal rather than proximal to the contact point between T cells and antigen presenting cells [43]. As distal daughter cells express more IL-7Rα, they may be more able to receive the life giving signals (such as Bcl-2 induction) of IL-7 [4345]. Thus, IL-7Rαhi effector T cells may compete for limiting amounts of IL-7, and may be selected to survive and become memory T cells. These data suggest that early differentiation decisions result in the formation of “memory precursors” and a pool of terminally differentiated effectors. Given this scenario, it would be expected that interference with contraction of the response at the level of apoptosis would not increase memory T cell numbers, assuming that apoptosis is downstream of differentiation decisions. But, data from studies using Bim−/− mice argue against this possibility because effector T cells from these mice die poorly and give rise to significantly increased numbers of memory T cells [29]. These data suggest that effector T cells are somewhat flexible in their ability to form memory cells.

Further, other pieces of data suggest that contraction of the response is not solely dictated by limiting amounts of IL-7. First, significant numbers of viral-specific IL-7Rαlo T cells survive contraction of the response [29]. Second, following peptide immunization, substantial numbers of antigen-specific IL-7Rαhi T cells die [46]. Moreover, during chronic infections, IL-7Rαlo T cells persist and these cells can re-express IL-7Rα once the virus is cleared [47]. Finally, neutralization of IL-7 (or Bcl-2) during viral infection does not exacerbate contraction of the response, suggesting that IL-7 is not required for T cells to survive contraction of the response [48]. It is possible that IL-15 plays a redundant role with IL-7 because IL-15-deficient mice have a slightly exacerbated contraction of CD8+ T cell responses [49, 50]. Thus, while decreased IL-7Rα expression may mark some effector T cells destined to die via Bim, expression of IL-7Rα is not the only determinant of death decisions within the effector population.

Role of Bcl-2 family members in controlling long-term memory T cell survival

Following the contraction phase of the T cell response, in general, a long-lived memory population persists. Memory CD8+ T cells are maintained after the contraction phase by the pro-survival and proliferative actions of IL-7 and IL-15, respectively, and do not appear to require interactions with class I MHC [5052]. Memory CD4+ T cells are largely maintained by IL-7 [53] and possibly by interactions with class II MHC, although this is controversial [54, 55]. Nonetheless, because memory T cells divide regularly, but generally do not accumulate in number, apoptosis is critical to maintain memory T cell homeostasis.

Memory T cells express higher levels of Bcl-2 compared to naïve T cells [32] (Table 1). This may help memory T cells combat Bim. In addition, TNF receptor-associated factor-1 (TRAF-1) signals may suppress Bim expression to allow memory cell survival [56]. Also, in human memory T cells, lowered levels of Bim, perhaps because of Fox03a expression, may allow better survival of central versus effector memory cells [57]. Consistent with these observations, viral-specific, effector memory T cells accumulate more in Bim−/− mice compared to controls (D.H. personal observations). However, the overall numbers of memory T cells in Bim−/− mice slowly wane over time and approach levels observed in wild type mice [29], suggesting that factors other than Bim act to keep memory T cells in check. These may involve other BH3-only proteins since, unlike the situation in Bim−/− mice, endogenous memory T cells accumulate rapidly and spontaneously in Bax/Bak double-deficient animals [30].

Discussion

Bcl-2 family members play a critical role in the regulation of immune system homeostasis. These molecules control the survival of both innate and adaptive immune cells. Interestingly, the effects of Bcl-2 family members and death receptors on innate immune cells may affect the magnitude of T cell responses, through how this happens is not clear. On the other hand, the ability of Bim to limit effector T cell survival substantially limits the numbers of cells available for the memory pool. Thus, novel therapies aimed at targeting the Bcl-2 family which are being developed as cancer therapeutics may also prove useful in manipulating and regulating immune homeostasis.

Acknowledgements

The authors thank Ms. Jill Fritz for help organizing the tables. This work was supported by Public Health Service Grants AI-057753 (D.A.H.), AI-18785, 5U19 AI-046374 and CA-108771 (P.C.M.).

References

  • 1.Murali-Krishna K, Altman JD, Suresh M, Sourdive DJ, Zajac AJ, Miller JD, Slansky J, Ahmed R. Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. Immunity. 1998;8:177–187. doi: 10.1016/s1074-7613(00)80470-7. [DOI] [PubMed] [Google Scholar]
  • 2.Brunner T, Mogil RJ, LaFace D, Yoo NJ, Mahboubi A, Echeverri F, Martin SJ, Force WR, Lynch DH, Ware CF, et al. Cell-autonomous Fas (CD95)/Fas-ligand interaction mediates activation-induced apoptosis in T-cell hybridomas. Nature. 1995;373:441–444. doi: 10.1038/373441a0. [DOI] [PubMed] [Google Scholar]
  • 3.Dhein J, Walczak H, Baumler C, Debatin KM, Krammer PH. Autocrine T-cell suicide mediated by APO-1/(Fas/CD95) Nature. 1995;373:438–441. doi: 10.1038/373438a0. [DOI] [PubMed] [Google Scholar]
  • 4.Ju ST, Panka DJ, Cui H, Ettinger R, el-Khatib M, Sherr DH, Stanger BZ, Marshak-Rothstein A. Fas(CD95)/FasL interactions required for programmed cell death after T-cell activation. Nature. 1995;373:444–448. doi: 10.1038/373444a0. [DOI] [PubMed] [Google Scholar]
  • 5.Hildeman DA, Zhu Y, Mitchell TC, Bouillet P, Strasser A, Kappler J, Marrack P. Activated T cell death in vivo mediated by proapoptotic bcl-2 family member bim. Immunity. 2002;16:759–767. doi: 10.1016/s1074-7613(02)00322-9. [DOI] [PubMed] [Google Scholar]
  • 6.Pellegrini M, Belz G, Bouillet P, Strasser A. 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 U S A. 2003;100:14175–14180. doi: 10.1073/pnas.2336198100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Hao Z, Hampel B, Yagita H, Rajewsky K. T cell-specific ablation of Fas leads to Fas ligand-mediated lymphocyte depletion and inflammatory pulmonary fibrosis. J Exp Med. 2004;199:1355–1365. doi: 10.1084/jem.20032196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Song J, So T, Cheng M, Tang X, Croft M. Sustained survivin expression from OX40 costimulatory signals drives T cell clonal expansion. Immunity. 2005;22:621–631. doi: 10.1016/j.immuni.2005.03.012. [DOI] [PubMed] [Google Scholar]
  • 9.Xing Z, Conway EM, Kang C, Winoto A. Essential role of survivin, an inhibitor of apoptosis protein, in T cell development, maturation, and homeostasis. J Exp Med. 2004;199:69–80. doi: 10.1084/jem.20031588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kamath AT, Pooley J, O'Keeffe MA, Vremec D, Zhan Y, Lew AM, D'Amico A, Wu L, Tough DF, Shortman K. The development, maturation, and turnover rate of mouse spleen dendritic cell populations. J Immunol. 2000;165:6762–6770. doi: 10.4049/jimmunol.165.12.6762. [DOI] [PubMed] [Google Scholar]
  • 11.Nopora A, Brocker T. Bcl-2 controls dendritic cell longevity in vivo. J Immunol. 2002;169:3006–3014. doi: 10.4049/jimmunol.169.6.3006. [DOI] [PubMed] [Google Scholar]
  • 12.Ashany D, Savir A, Bhardwaj N, Elkon KB. Dendritic cells are resistant to apoptosis through the Fas (CD95/APO-1) pathway. J Immunol. 1999;163:5303–5311. [PubMed] [Google Scholar]
  • 13.Willems F, Amraoui Z, Vanderheyde N, Verhasselt V, Aksoy E, Scaffidi C, Peter ME, Krammer PH, Goldman M. Expression of c-FLIP(L) and resistance to CD95− mediated apoptosis of monocyte-derived dendritic cells: inhibition by bisindolylmaleimide. Blood. 2000;95:3478–3482. [PubMed] [Google Scholar]
  • 14.Chen M, Wang YH, Wang Y, Huang L, Sandoval H, Liu YJ, Wang J. Dendritic cell apoptosis in the maintenance of immune tolerance. Science. 2006;311:1160–1164. doi: 10.1126/science.1122545. [DOI] [PubMed] [Google Scholar]
  • 15.Chen M, Huang L, Wang J. Deficiency of Bim in dendritic cells contributes to over-activation of lymphocytes and autoimmunity. Blood. 2007 doi: 10.1182/blood-2006-11-056424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Odermatt B, Eppler M, Leist TP, Hengartner H, Zinkernagel RM. Virus-triggered acquired immunodeficiency by cytotoxic T-cell-dependent destruction of antigen-presenting cells and lymph follicle structure. Proc Natl Acad Sci U S A. 1991;88:8252–8256. doi: 10.1073/pnas.88.18.8252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yang J, Huck SP, McHugh RS, Hermans IF, Ronchese F. Perforin-dependent elimination of dendritic cells regulates the expansion of antigen-specific CD8+ T cells in vivo. Proc Natl Acad Sci U S A. 2006;103:147–152. doi: 10.1073/pnas.0509054103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Matsue H, Edelbaum D, Hartmann AC, Morita A, Bergstresser PR, Yagita H, Okumura K, Takashima A. Dendritic cells undergo rapid apoptosis in vitro during antigen-specific interaction with CD4+ T cells. J Immunol. 1999;162:5287–5298. [PubMed] [Google Scholar]
  • 19.Fugier-Vivier I, Servet-Delprat C, Rivailler P, Rissoan MC, Liu YJ, Rabourdin-Combe C. Measles virus suppresses cell-mediated immunity by interfering with the survival and functions of dendritic and T cells. J Exp Med. 1997;186:813–823. doi: 10.1084/jem.186.6.813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Guo Z, Zhang M, Tang H, Cao X. Fas signal links innate and adaptive immunity by promoting dendritic-cell secretion of CC and CXC chemokines. Blood. 2005;106:2033–2041. doi: 10.1182/blood-2004-12-4831. [DOI] [PubMed] [Google Scholar]
  • 21.Rothstein TL, Zhong X, Schram BR, Negm RS, Donohoe TJ, Cabral DS, Foote LC, Schneider TJ. Receptor-specific regulation of B-cell susceptibility to Fas-mediated apoptosis and a novel Fas apoptosis inhibitory molecule. Immunol Rev. 2000;176:116–133. doi: 10.1034/j.1600-065x.2000.00616.x. [DOI] [PubMed] [Google Scholar]
  • 22.Craxton A, Draves KE, Gruppi A, Clark EA. BAFF regulates B cell survival by downregulating the BH3-only family member Bim via the ERK pathway. J Exp Med. 2005;202:1363–1374. doi: 10.1084/jem.20051283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Goodyear CS, Corr M, Sugiyama F, Boyle DL, Silverman GJ. Cutting Edge: Bim is required for superantigen-mediated B cell death. J Immunol. 2007;178:2636–2640. doi: 10.4049/jimmunol.178.5.2636. [DOI] [PubMed] [Google Scholar]
  • 24.Jin H, Carrio R, Yu A, Malek TR. Distinct activation signals determine whether IL-21 induces B cell costimulation, growth arrest, or Bim-dependent apoptosis. J Immunol. 2004;173:657–665. doi: 10.4049/jimmunol.173.1.657. [DOI] [PubMed] [Google Scholar]
  • 25.Lesley R, Xu Y, Kalled SL, Hess DM, Schwab SR, Shu HB, Cyster JG. Reduced competitiveness of autoantigen-engaged B cells due to increased dependence on BAFF. Immunity. 2004;20:441–453. doi: 10.1016/s1074-7613(04)00079-2. [DOI] [PubMed] [Google Scholar]
  • 26.Oliver PM, Vass T, Kappler J, Marrack P. Loss of the proapoptotic protein, Bim, breaks B cell anergy. J Exp Med. 2006;203:731–741. doi: 10.1084/jem.20051407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Takeuchi O, Fisher J, Suh H, Harada H, Malynn BA, Korsmeyer SJ. Essential role of BAX,BAK in B cell homeostasis and prevention of autoimmune disease. Proc Natl Acad Sci U S A. 2005;102:11272–11277. doi: 10.1073/pnas.0504783102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Jego G, Pascual V, Palucka AK, Banchereau J. Dendritic cells control B cell growth and differentiation. Curr Dir Autoimmun. 2005;8:124–139. doi: 10.1159/000082101. [DOI] [PubMed] [Google Scholar]
  • 29. Wojciechowski S, Jordan MB, Zhu Y, White J, Zajac AJ, Hildeman DA. Bim mediates apoptosis of CD127(lo) effector T cells and limits T cell memory. Eur J Immunol. 2006;36:1694–1706. doi: 10.1002/eji.200635897.. This paper shows that CD127lo T cells die via Bim, increase expression of CD127 and enter the memory compartment. These data suggest that Bim is a major limiting factor for entrance of effector T cells into the memory pool and argue against terminal differentiation models of memory development.
  • 30.Rathmell JC, Lindsten T, Zong WX, Cinalli RM, Thompson CB. Deficiency in Bak and Bax perturbs thymic selection and lymphoid homeostasis. Nat Immunol. 2002;3:932–939. doi: 10.1038/ni834. [DOI] [PubMed] [Google Scholar]
  • 31.Zhu Y, Liu X, Hildeman D, Peyerl FW, White J, Kushnir E, Kappler J, Marrack P. Bax does not have to adopt its final form to drive T cell death. J Exp Med. 2006;203:1147–1152. doi: 10.1084/jem.20051736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Grayson JM, Zajac AJ, Altman JD, Ahmed R. Cutting edge: increased expression of Bcl-2 in antigen-specific memory CD8+ T cells. J Immunol. 2000;164:3950–3954. doi: 10.4049/jimmunol.164.8.3950. [DOI] [PubMed] [Google Scholar]
  • 33.Mitchell T, Kappler J, Marrack P. Bystander virus infection prolongs activated T cell survival. J Immunol. 1999;162:4527–4535. [PubMed] [Google Scholar]
  • 34. Zhu Y, Swanson BJ, Wang M, Hildeman DA, Schaefer BC, Liu X, Suzuki H, Mihara K, Kappler J, Marrack P. Constitutive association of the proapoptotic protein Bim with Bcl-2-related proteins on mitochondria in T cells. Proc Natl Acad Sci U S A. 2004;101:7681–7686. doi: 10.1073/pnas.0402293101.. Biochemical demonstration that Bim is complexed with Bcl-2 on mitochondria in normal resting T cells. Following T cell activation, Bcl-2 is downregulated and Bim changes its binding partners.
  • 35.Mitchell TC, Hildeman D, Kedl RM, Teague TK, Schaefer BC, White J, Zhu Y, Kappler J, Marrack P. Immunological adjuvants promote activated T cell survival via induction of Bcl-3. Nat Immunol. 2001;2:397–402. doi: 10.1038/87692. [DOI] [PubMed] [Google Scholar]
  • 36.Bauer A, Villunger A, Labi V, Fischer SF, Strasser A, Wagner H, Schmid RM, Hacker G. The NF-kappaB regulator Bcl-3 and the BH3-only proteins Bim and Puma control the death of activated T cells. Proc Natl Acad Sci U S A. 2006;103:10979–10984. doi: 10.1073/pnas.0603625103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sandalova E, Wei CH, Masucci MG, Levitsky V. Regulation of expression of Bcl-2 protein family member Bim by T cell receptor triggering. Proc Natl Acad Sci U S A. 2004;101:3011–3016. doi: 10.1073/pnas.0400005101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Seward RJ, von Haller PD, Aebersold R, Huber BT. Phosphorylation of the pro-apoptotic protein Bim in lymphocytes is associated with protection from apoptosis. Mol Immunol. 2003;39:983–993. doi: 10.1016/s0161-5890(03)00047-6. [DOI] [PubMed] [Google Scholar]
  • 39.Strasser A. The role of BH3-only proteins in the immune system. Nat Rev Immunol. 2005;5:189–200. doi: 10.1038/nri1568. [DOI] [PubMed] [Google Scholar]
  • 40.Bouillet P, Metcalf D, Huang DC, Tarlinton DM, Kay TW, Kontgen F, Adams JM, Strasser A. Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity. Science. 1999;286:1735–1738. doi: 10.1126/science.286.5445.1735. [DOI] [PubMed] [Google Scholar]
  • 41.Villunger A, Michalak EM, Coultas L, Mullauer F, Bock G, Ausserlechner MJ, Adams JM, Strasser A. p53- and drug-induced apoptotic responses mediated by BH3-only proteins puma and noxa. Science. 2003;302:1036–1038. doi: 10.1126/science.1090072. [DOI] [PubMed] [Google Scholar]
  • 42.Erlacher M, Labi V, Manzl C, Bock G, Tzankov A, Hacker G, Michalak E, Strasser A, Villunger A. Puma cooperates with Bim, the rate-limiting BH3-only protein in cell death during lymphocyte development, in apoptosis induction. J Exp Med. 2006;203:2939–2951. doi: 10.1084/jem.20061552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chang JT, Palanivel VR, Kinjyo I, Schambach F, Intlekofer AM, Banerjee A, Longworth SA, Vinup KE, Mrass P, Oliaro J, et al. Asymmetric T lymphocyte division in the initiation of adaptive immune responses. Science. 2007;315:1687–1691. doi: 10.1126/science.1139393. [DOI] [PubMed] [Google Scholar]
  • 44.Kaech SM, Tan JT, Wherry EJ, Konieczny BT, Surh CD, Ahmed R. Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells. Nat Immunol. 2003;4:1191–1198. doi: 10.1038/ni1009. [DOI] [PubMed] [Google Scholar]
  • 45.Huster KM, Busch V, Schiemann M, Linkemann K, Kerksiek KM, Wagner H, Busch DH. 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 U S A. 2004;101:5610–5615. doi: 10.1073/pnas.0308054101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lacombe MH, Hardy MP, Rooney J, Labrecque N. IL-7 receptor expression levels do not identify CD8+ memory T lymphocyte precursors following peptide immunization. J Immunol. 2005;175:4400–4407. doi: 10.4049/jimmunol.175.7.4400. [DOI] [PubMed] [Google Scholar]
  • 47.Fuller MJ, Hildeman DA, Sabbaj S, Gaddis DE, Tebo AE, Shang L, Goepfert PA, Zajac AJ. Cutting edge: emergence of CD127high functionally competent memory T cells is compromised by high viral loads and inadequate T cell help. J Immunol. 2005;174:5926–5930. doi: 10.4049/jimmunol.174.10.5926. [DOI] [PubMed] [Google Scholar]
  • 48. Tripathi P, Mitchell TC, Finkelman F, Hildeman DA. Cutting Edge: Limiting Amounts of IL-7 Do Not Control Contraction of CD4+ T Cell Responses. J Immunol. 2007;178:4027–4031. doi: 10.4049/jimmunol.178.7.4027.. This study shows that exogenous IL-7 can prevent contraction of T cell responses through induction of Bcl-2. Interestingly, neutralization of either IL-7 or Bcl-2 does not exacerbate contraction, suggesting that effector T cells do not require IL-7 or Bcl-2 to survive contraction or the response.
  • 49. Yajima T, Yoshihara K, Nakazato K, Kumabe S, Koyasu S, Sad S, Shen H, Kuwano H, Yoshikai Y. IL-15 regulates CD8+ T cell contraction during primary infection. J Immunol. 2006;176:507–515. doi: 10.4049/jimmunol.176.1.507.. An intriguing study suggesting that IL-15 may contribute to effector T cell survival during contraction of the response.
  • 50.Becker TC, Wherry EJ, Boone D, Murali-Krishna K, Antia R, Ma A, Ahmed R. Interleukin 15 is required for proliferative renewal of virus-specific memory CD8 T cells. J Exp Med. 2002;195:1541–1548. doi: 10.1084/jem.20020369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Schluns KS, Kieper WC, Jameson SC, Lefrancois L. Interleukin-7 mediates the homeostasis of naive and memory CD8 T cells in vivo. Nat Immunol. 2000;1:426–432. doi: 10.1038/80868. [DOI] [PubMed] [Google Scholar]
  • 52.Murali-Krishna K, Lau LL, Sambhara S, Lemonnier F, Altman J, Ahmed R. Persistence of memory CD8 T cells in MHC class I-deficient mice. Science. 1999;286:1377–1381. doi: 10.1126/science.286.5443.1377. [DOI] [PubMed] [Google Scholar]
  • 53.Lenz DC, Kurz SK, Lemmens E, Schoenberger SP, Sprent J, Oldstone MB, Homann D. IL-7 regulates basal homeostatic proliferation of antiviral CD4+T cell memory. Proc Natl Acad Sci U S A. 2004;101:9357–9362. doi: 10.1073/pnas.0400640101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Seddon B, Tomlinson P, Zamoyska R. Interleukin 7 and T cell receptor signals regulate homeostasis of CD4 memory cells. Nat Immunol. 2003;4:680–686. doi: 10.1038/ni946. [DOI] [PubMed] [Google Scholar]
  • 55.Swain SL, Hu H, Huston G. Class II-independent generation of CD4 memory T cells from effectors. Science. 1999;286:1381–1383. doi: 10.1126/science.286.5443.1381. [DOI] [PubMed] [Google Scholar]
  • 56.Sabbagh L, Srokowski CC, Pulle G, Snell LM, Sedgmen BJ, Liu Y, Tsitsikov EN, Watts TH. A critical role for TNF receptor-associated factor 1 and Bim down-regulation in CD8 memory T cell survival. Proc Natl Acad Sci U S A. 2006;103:18703–18708. doi: 10.1073/pnas.0602919103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Riou C, Yassine-Diab B, Van grevenynghe J, Somogyi R, Greller LD, Gagnon D, Gimmig S, Wilkinson P, Shi Y, Cameron MJ, et al. Convergence of TCR and cytokine signaling leads to FOXO3a phosphorylation and drives the survival of CD4+ central memory T cells. J Exp Med. 2007;204:79–91. doi: 10.1084/jem.20061681.. A study in human CD4 memory T cells shows differential Bim expression within effector versus central memory T cells is likely due to differential Foxo3a phosphorylation. These data may explain the gradual accumulation of central memory T cells.

RESOURCES