<<

CD4+ and CD8+ T Cell Survival Is Regulated Differentially by Protein C θ, c-Rel, and

This information is current as Samuel D. Saibil, Russell G. Jones, Elissa K. Deenick, of September 27, 2021. Nicole Liadis, Alisha R. Elford, Mitchell G. Vainberg, Heather Baerg, James R. Woodgett, Steve Gerondakis and Pamela S. Ohashi J Immunol 2007; 178:2932-2939; ;

doi: 10.4049/jimmunol.178.5.2932 Downloaded from http://www.jimmunol.org/content/178/5/2932

References This article cites 69 articles, 36 of which you can access for free at: http://www.jimmunol.org/content/178/5/2932.full#ref-list-1 http://www.jimmunol.org/

Why The JI? Submit online.

• Rapid Reviews! 30 days* from submission to initial decision

• No Triage! Every submission reviewed by practicing scientists by guest on September 27, 2021 • Fast Publication! 4 weeks from acceptance to publication

*average

Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2007 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology

CD4؉ and CD8؉ T Cell Survival Is Regulated Differentially by ␪, c-Rel, and Protein Kinase B1

Samuel D. Saibil,†‡ Russell G. Jones,§ Elissa K. Deenick,*†‡ Nicole Liadis,*‡ Alisha R. Elford,‡ Mitchell G. Vainberg,*‡ Heather Baerg,‡ James R. Woodgett,* Steve Gerondakis,¶ and Pamela S. Ohashi2*†‡

An effective immune response requires the expansion and survival of a large number of activated T cells. This study compared the role of protein kinase C (PKC)␪ and associated signaling molecules in the survival of activated primary CD4؉ vs CD8؉ murine T cells. We demonstrate that the absence of PKC␪ resulted in a moderate survival defect in CD4؉ T cells and a striking survival defect of CD8؉ T lymphocytes. CD8؉ T cells lacking the c-Rel, but not the NF-␬B1/p50, member of the NF-␬B family of factors displayed a similar impairment in cell survival as PKC␪؊/؊ CD8؉ T lymphocytes. This implicates c-Rel as a key target of PKC␪-mediated survival signals in CD8؉ T cells. In addition, both c-Rel؊/؊ and PKC␪؊/؊ T cells also displayed Downloaded from

impaired expression of the antiapoptotic Bcl-xL protein upon activation. Changes in Bcl-xL expression, however, did not correlate with the survival of CD4؉ or CD8؉ lymphocytes. The addition of protein kinase B-mediated survival signals could restore partially CD4؉ T cell viability, but did not dramatically influence CD8؉ survival. Active protein kinase B was also unable to restore ,proliferative responses in CD8؉ PKC␪؊/؊ T cells. The survival of CD4؉ and CD8؉ T cells deficient in either PKC␪ or c-Rel however, was promoted by the addition of IL-2. Collectively, these data demonstrate that CD4؉ and CD8؉ T cell survival signals http://www.jimmunol.org/ are differentially programmed. The Journal of Immunology, 2007, 178: 2932–2939.

oth immature and mature T cells are destined to die upon the generation of lipid second messengers at the plasma mem- Ag-specific TCR engagement. In thymocytes, TCR-spe- brane. The conversion of -3,4-bisphosphate B cific engagement with high-affinity ligands leads to neg- into phosphatidylinositol-3,4,5-trisphosphate by PI3K prompts the ative selection or clonal deletion (reviewed in Ref. 1). Similarly, in recruitment of PKB to phosphatidylinositol-3,4,5-trisphosphate via the absence of costimulatory and proinflammatory signals, mature its pleckstrin homology domain. Once recruited to the plasma T cells also undergo clonal deletion upon engagement with high- membrane, PKB is subsequently phosphorylated and fully acti-

affinity ligands (2). Therefore, one fundamental issue in T cell vated (9). Active PKB has been demonstrated to promote survival by guest on September 27, 2021 immunity is to understand the nature of survival signals and the due to its ability to phosphorylate, and thereby inhibit, the pro- integration of these signals during T cell activation. Although a apoptotic BH3-only protein BAD (10–12). As well, the forkhead number of molecules, such as Bcl-xL and FLIP, have been shown transcription factors are also direct substrates of PKB (13–15); to play a role in T cell survival (3–7), the signaling pathways that of the forkhead transcription factors FOXO by mediate T cell survival remain unclear. PKB prevents the transcription of the proapoptotic BH3-only pro- 3 The serine/threonine kinase protein kinase B (PKB)␣ (Akt1) tein Bim (16), as well as the transcription of Fas (13). Many has been shown to promote survival in multiple cell lineages, in- other molecules such as GSK-3, Cot, and p27 have been shown to cluding T cells (PKB reviewed in Ref. 8). PKB is activated after be downstream targets of PKB␣ (17–20). In addition to its direct substrates, PKB also promotes cell survival through the activation ␬ *Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Can- of the NF- B family of transcription factors (21–24). ada; †Department of Immunology, University of Toronto, Toronto, Ontario, Canada; In mammals, the NF-␬B transcription factors are composed of ‡Institute for Research, Ontario Cancer Institute, University of Toronto, § homo- or heterodimers of five conserved subunits: p65/RelA, c- Toronto, Ontario, Canada; Abramson Family Cancer Research Institute, Philadel- ␬ ␬ ␬ phia, PA 19104; and ¶Walter and Eliza Hall Institute of Medical Research, Parkville, Rel, RelB, NF- B1/p50, and NF- B2/p52 (NF- B reviewed in Australia Ref. 25). These dimeric transcription factors are essential for T cell Received for publication February 8, 2006. Accepted for publication December activation and regulate genes associated with proliferation, cyto- 21, 2006. kine production, as well as protection against . In T cells, The costs of publication of this article were defrayed in part by the payment of page NF-␬B signaling has been linked not only to the induction of an- charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. tiapoptotic members of the Bcl-2 family of proteins, such as 1 This work was supported by the Canadian Institutes for Health Research and Na- Bcl-xL (26–28), but also to protection from Fas-mediated apopto- tional Cancer Institute of Canada, with funds from the Canadian Cancer Society. sis (29–31). Initiation of NF-␬B-mediated transcription requires S.D.S. is supported by a Canadian Institutes for Health Research MD/PhD award. the activation of the I␬B kinase (IKK) complex. This complex P.S.O. holds a Canada Research Chair. consists of two kinase subunits, IKK␣ and IKK␤, as well as a 2 Address correspondence and reprint requests to Dr. Pamela S. Ohashi, The Campbell ␥ ␬ Institute for Breast Cancer Research, University of Toronto, 610 University Avenue, regulatory subunit IKK /NF- B essential modulator. Activation Room 706, Toronto, Ontario, Canada M5G 2C1. E-mail address: pohashi@uhnres. of the in this complex results in the phosphorylation and utoronto.ca subsequent proteosomal degradation of the I␬B protein. The I␬B 3 Abbreviations used in this paper: PKB, protein kinase B; 7AAD, 7-aminoactino- protein functions by sequestering NF-␬B transcription factors in mycin D; IKK, I␬B kinase; MFI, mean fluorescence intensity; PKC, protein kinase C. the cytosol. Degradation of I␬B allows the nuclear translocation of Copyright © 2007 by The American Association of Immunologists, Inc. 0022-1767/07/$2.00 the NF-␬B and gene transcription.

www.jimmunol.org The Journal of Immunology 2933

Protein kinase C (PKC)␪, a member of the calcium-independent annexin V FITC and 7AAD. Samples were acquired on a FACScan in- subfamily of PKCs (PKC␪ reviewed in Ref. 32), has been dem- strument (BD Biosciences) and were analyzed using CellQuest software ␥ ␬ (BD Biosciences). For intracellular Bcl-xL staining, Fc RIII were first onstrated to be an important mediator of NF- B signaling. T lym- ϫ 6 ␪ blocked with an Ab. A total of 1 10 cells was then fixed and perme- phocytes from mice deficient for PKC demonstrate not only im- abilized using BD Cytofix and BD Cytoperm (BD Biosciences) as per the ␬ paired activation of NF- B, but also reduced activation of the manufacturer’s instructions. Cells were then stained with anti-Bcl-xL FITC AP-1 transcription factor (33, 34). The functional consequence of Ab or an isotype control Ab. Samples were then acquired and analyzed. this impaired signaling is that PKC␪Ϫ/Ϫ T cells displayed impaired Western blot analysis proliferation (35) and reduced production of IL-2 (33) upon acti- vation. Recently, a role for PKC␪ in the promotion of survival of Single-cell suspensions were lysed by incubation on ice in Gentle Soft activated T cells has been described (36). Moreover, it has been Buffer (10 mM NaCl, 20 mM PIPES (pH 7.4), 0.5% Nonidet P-40, 5 mM ␮ ␮ suggested previously that PKB and PKC␪ molecules physically EDTA, 5 g/ml leupeptin, 1 mM benzamidine, aprotinin, and 100 M Na3VO4) for 20 min. The lysates were then cleared by centrifugation, and associate (37, 38). Together, these reports suggest interplay be- the supernatants were normalized for total protein (Bio-Rad). Protein was tween the PKB and PKC␪ signaling pathways in the promotion of resolved by 4–20% SDS-PAGE and then electroblotted onto polyvinyli- survival of activated T cells. Therefore, we investigated the roles dene difluoride membrane (Costar). The membranes were blocked in 5% of PKC␪, PKB, and members of the NF-␬B family in controlling nonfat milk in TBST and probed with primary Abs for 12 h at 4°C. Anti- Bcl-xL was purchased from Cell Signaling Technology, and anti-actin was the survival of activated primary murine T cells. Our results dem- purchased from Sigma-Aldrich. onstrate that CD4ϩ and CD8ϩ T cell survival are programmed differentially. PKC␪ signaling events are important for both CD4ϩ Proliferation assay ϩ Downloaded from and CD8 T cell survival, which can be modulated by PKB or Splenocytes from P14 PKC␪Ϫ/Ϫ, P14 PKC␪Ϫ/Ϫ PKB, and P14 PKC␪ϩ/Ϫ IL-2. PKC␪- and c-Rel-mediated signals, however, appear to be mice (2 ϫ 105 cells/well) were incubated 72 h at 37°C in 96-well flat- more important for CD8ϩ T cell survival. bottom plates with increasing concentrations of gp33 peptide. Proliferation was measured by the incorporation of an overnight pulse of 1 ␮Ci of 3 Materials and Methods [ H]thymidine after 72 h of incubation. Mice Results Ϫ Ϫ Ϫ Ϫ Ϫ Ϫ Ϫ Ϫ http://www.jimmunol.org/ The PKC␪ / (33), nf-␬b1 / (p50 / ) (39), and the c-rel / (40) mouse The PKC␪ signals are critical for T cell survival in vitro lines have all been described previously, as have the human CD2-gag-PKB transgenic (B6/PKB) (24), and the P14 TCR transgenic (41). The PKB/ To examine whether PKC␪ conveys survival signals in activated T PKC␪Ϫ/Ϫ line of mice was generated by breeding B6/PKB mice to PKC- cells, T lymphocytes from PKC␪Ϫ/Ϫ, littermate PKC␪ϩ/Ϫ, and deficient mice. These mice were subsequently crossed to the P14 TCR control C57BL/6 mice were stimulated with plate-bound anti-CD3 transgenic mice. C57BL/6J mice were obtained from The Jackson Labo- ratory. All mice were maintained in a specific pathogen-free environment and anti-CD28 Abs. After stimulation, cells were incubated for at the Ontario Cancer Institute, according to institutional guidelines. 20 h without stimulation, and then viability was assessed by flow cytometry using annexin V and 7AAD. Cell survival was moni- Reagents and Abs tored in both the CD4ϩ and the CD8ϩ T cell subsets and was Murine rIL-2 was purchased from PeproTech. Purified anti-CD3 (2C11) normalized in each culture to the percentage of viable cells after by guest on September 27, 2021 and anti-CD28 (37.51) were purchased from eBioscience. Anti-Bcl-x L removal from the stimulation (viability at t0). As compared with FITC Ab was purchased from Southern Biotechnology Associates. Purified the C57BL/6 and littermate heterozygous control cells, which dis- anti-Fc␥RIII/II (2.4G2) was purchased from BD Pharmingen. The follow- ϩ ϩ played similar viability in both the CD4 and the CD8 T cell ing Abs purchased from BD Pharmingen were used for flow cytometry (PE ϩ conjugated): anti-CD4 and anti-CD8. Th annexin V FITC was also pur- compartments, the CD4 T cells lacking PKC␪ displayed a de- chased from BD Pharmingen, whereas the 7-aminoactinomycin D (7AAD) crease in cell viability after 20 h of culture (Fig. 1A). The average was purchased from Sigma-Aldrich. The mouse IgG FITC isotype control viability of the PKC␪-deficient CD4ϩ T cells was 36.7% as com- Ab was purchased from Jackson ImmunoResearch Laboratories. pared with 52.1% for the C57BL/6 cells and 55.2% for the ϩ Ϫ In vitro cell stimulation PKC␪ / control cells. This difference in viability between the C57BL/6 and knockout CD4ϩ T cells is statically significant ( p Ͻ To assay Bcl-xL expression, splenic T cells were purified using the Pan T Cell Isolation Kit on the AutoMACS (Miltenyi Biotec), as per the manu- 0.0003 by two-tailed Student’s t test). The impaired survival in Ϫ/Ϫ ϩ facturer’s instructions. Purified T cells were plated at 1–2 ϫ 106 cells/ml in PKC␪ T cells, however, was much more drastic in the CD8 24-well flat-bottom plates coated with either purified anti-CD3 or com- compartment, because the PKC␪-deficient cells averaged only bined anti-CD3 and anti-CD28. After the indicated times, cells were har- 12.8% viable cells as compared with 59.2% viability for the vested for analysis. PKC␪ϩ/Ϫ CD8ϩ T cells and 64.8% for the C57BL/6 control cells. Cell viability assay These results are consistent with a recent report that suggests PKC␪ is important for CD8ϩ T cell survival (36). Interestingly, we A total of 107 splenocytes was plated at 2 ϫ 106 cells/ml in six-well ␪Ϫ/Ϫ ϩ ϩ flat-bottom plates coated with 2 ␮g/ml anti-CD3 and 2 ␮g/ml anti-CD28. found no enhanced cell death in PKC CD4 or CD8 T cells Cells were stimulated for 72 h in RPMI 1640 medium supplemented with that were subjected to activation-induced cell death via TCR re- 10% heat-inactivated FCS (Invitrogen Life Technologies), 50 ␮M 2-ME, 2 stimulation (data not shown). Thus, PKC␪ may not be required to mM glutamine, and 0.1% penicillin/streptomycin. After 72 h of stimula- protect against all forms of cell death. Our data, however, dem- tion, cells were washed and cell viability was assessed in the CD4 and CD8 onstrate that PKC␪ is required to maintain the survival of activated compartments from a triplicate of aliquots using flow cytometry (viability ϩ ϫ 5 T lymphocytes, particularly those of the CD8 lineage. at t0; t stands for time). A total of 2 10 cells was then plated in triplicate wells in 96-well flat-bottom plates. After 20 h of incubation, cell viability In the next experiments, we sought to ascertain the molecules was assessed again in the CD4 and CD8 compartments using flow cytom- downstream of PKC␪ that are responsible for promoting cell sur- etry. Cell viability is reported as a percentage of the viability of each ␪ ␬ ϭ vival. PKC is known to activate NF- B (33, 34, 42, 43). We culture at t0, using the formula: percentage viability ((viability after ϫ investigated whether T cells lacking only p50 or c-Rel displayed a 20 h)/(viability at t0)) 100. similar phenotype as T cells lacking PKC␪. When cell viability Flow cytometry was assessed in CD4ϩ T cells lacking either p50 or c-Rel, no For cell viability assays, 2 ϫ 105 cells were first stained with either anti- impairment in survival was noted relative to wild-type controls CD4 or anti-CD8 at 4°C. Cell viability was then assessed by staining with (Fig. 1B). This suggests that there might be redundancy between 2934 DIFFERENTIAL PROGRAMMING OF CD4ϩ AND CD8ϩ T CELL SURVIVAL

␪Ϫ/Ϫ ϩ FIGURE 2. Impaired expression of Bcl-xL in PKC CD4 and ϩ 6 FIGURE 1. PKC␪ and c-Rel are required for the survival of activated T CD8 T lymphocytes. Purified splenic T cells (1 ϫ 10 cells) from Ϫ Ϫ ϩ Ϫ ϫ 7 ␪Ϫ/Ϫ ␪ϩ/Ϫ PKC␪ / , PKC␪ / , and C57BL/6 mice were stimulated with 2 ␮g/ml

cells. Splenocytes (1 10 cells) from A, PKC , littermate PKC , Downloaded from Ϫ/Ϫ Ϫ/Ϫ ␮ and C57BL/6 mice, or B, c-Rel , p50 , and C57BL/6 mice were stim- anti-CD3 combined with 2 g/ml anti-CD28 Ab for 24 h. Bcl-xL expres- ϩ ϩ ulated with anti-CD3 and anti-CD28 Abs (2 ␮g/ml of each) for 72 h. Cell sion in the CD4 and CD8 T cells was then determined by flow cytom- viability was then assessed by flow cytometry by staining with annexin V etry. Numbers in the histograms indicate the MFIs of staining for Bcl-xL. Specific staining for Bcl-x was confirmed using an isotype control Ab. and 7AAD to determine background death (viability at t0). The average L These data are representative of three independent experiments. viability at t0 ranged from 68 to 85% for the cells in A and 60 to 89% for the cells in B. Cells were then washed and recultured (2 ϫ 105 cells) in

triplicate without stimulation. After 20 h of reculture, cell viability was http://www.jimmunol.org/

assessed by flow cytometry by staining with annexin V and 7AAD. The as significantly (4, 47). All of these findings implicate Bcl-xL as an data are expressed as the percentage of viable cells after 20 h of reculture attractive candidate for a prosurvival molecule regulated by PKC␪ divided by the percentage of viable cells at t (after stimulation). The data 0 and c-Rel. To investigate this possibility, the expression of Bcl-xL are shown as an average value of triplicate wells with error bars showing was assessed in T cells lacking PKC␪. SD. These data are representative of three independent experiments. Purified splenic T cells were stimulated by plate-bound Abs to CD3 and CD28 for 24 h and then stained with either anti-CD4 or

anti-CD8 as well as for intracellular Bcl-xL. As illustrated in Fig. the various subunits of NF-␬B that the absence of one is insuffi- 2, CD4ϩ T cells lacking PKC␪ displayed reduced expression of ϩ ϩ Ϫ ␪ / by guest on September 27, 2021 cient to impair the survival of CD4 T cells. This does not, how- Bcl-xL relative to both the C57BL/6 and the littermate PKC ever, appear to be the case for CD8ϩ T cells. The CD8ϩ T cells controls. PKC␪Ϫ/Ϫ CD8ϩ lymphocytes showed a minor reduction ϩ ϩ ϩ lacking c-Rel displayed a similar phenotype to CD8 T lympho- in Bcl-xL levels. These results demonstrate that CD4 and CD8 ␪ ϩ Ϫ/Ϫ ␪ cytes lacking PKC . The CD8 c-Rel T cells averaged only T cells lacking PKC display impaired expression of Bcl-xL during 24.8% viability as compared with 71.7% viability for the C57BL/6 the process of T cell activation. ␪ cells. These findings implicate the PKC to c-Rel signaling axis as The kinetics of Bcl-xL expression during T cell activation was a major survival signaling pathway in CTL. also assessed by flow cytometry in both CD4ϩ and CD8ϩ T cells. The specific requirement for the c-Rel subunit of NF-␬B down- As can be seen in Fig. 3, stimulation of T lymphocytes from stream of PKC␪ for CD8ϩ T cell survival was demonstrated by the C57BL/6 mice with anti-CD3 Ab alone resulted in ϳ2-fold in- Ϫ/Ϫ lack of impaired survival in the T cells from the p50 mice. The crease in expression of Bcl-xL after 16 h of stimulation. The level ϩ CD8 T cells from these mice displayed a similar profile as the T of Bcl-xL expression increased further after 24 h of stimulation lymphocytes from the C57BL/6 mice (Fig. 1B). The CD8ϩ p50Ϫ/Ϫ with anti-CD3 in both the CD4ϩ and CD8ϩ T cells. Combined T cells had a marginally impaired average survival relative to the anti-CD3 and anti-CD28 Ab treatment of the wild-type T cells

control cells (59.4 vs 71.7%). This difference, however, was not enhanced the expression of Bcl-xL at all time points assayed in statistically significant ( p Ͼ 0.1 by two-tailed Student’s t test). both the CD4ϩ and CD8ϩ T lymphocytes. These data are consis- These results demonstrate that, unlike c-Rel, the p50 subunit of tent with previous studies that demonstrated that signals through ␬ ␪ NF- B is dispensable for the PKC -mediated survival signaling CD28 augment the expression of Bcl-xL (4, 48, 49). In contrast to pathway in activated CD8ϩ T lymphocytes. the control cells, both CD4ϩ and CD8ϩ T cells from the PKC␪Ϫ/Ϫ mice showed a reduced proportion of cells expressing Bcl-x at ␪ L PKC -mediated signals enhance the expression of Bcl-xL upon T ϩ ϩ high levels when stimulated through the Ag alone or with cell activation in both CD4 and CD8 T lymphocytes anti-CD3 and anti-CD28 Abs. This difference was most prominent There exists strong evidence connecting NF-␬B with the expres- after 24 h. These findings suggest that PKC␪ is required down-

sion of Bcl-xL. Mapping of the bcl-x promoter in mouse revealed stream of the Ag receptor for optimal expression of Bcl-xL upon T multiple ␬B consensus binding sites (44, 45). As well, studies in T cell activation. cell lines (26) and primary human T cells (27) demonstrated a requirement for NF-␬B in the expression of Bcl-x . Furthermore, c-Rel deficiency results in diminished expression of Bcl-xL upon L T cell activation the expression of pattern of Bcl-xL supports a role in the survival of activated T cells. Naive T cells express relatively high levels of Our results from Fig. 1 suggest that signals downstream of PKC␪

Bcl-2 (46) and almost undetectable levels of Bcl-xL (47). Upon and c-Rel, but not p50, are important for the survival of activated ϩ activation, however, the expression of Bcl-xL is induced rapidly in CD8 T cells. If Bcl-xL is indeed a key survival effector down- T lymphocytes, whereas the expression of Bcl-2 does not change stream of c-Rel, one prediction would be that the expression of The Journal of Immunology 2935

FIGURE 3. Kinetics of induction ␪Ϫ/Ϫ of Bcl-xL in PKC T cells. Puri- fied splenic T cells (1 ϫ 106 cells) from PKC␪Ϫ/Ϫ (black histogram) and C57BL/6 (gray histogram) mice were stimulated with plate-bound Ab. Cells were stimulated with either 2 ␮g/ml anti-CD3 Ab or 2 ␮g/ml anti-CD3 combined with 2 ␮g/ml anti-CD28

Ab for the indicated times. Bcl-xL ex- pression in the CD4ϩ and CD8ϩ T cells was then determined by flow cy- tometry. Numbers in the histograms indicate the MFIs of staining for ␪Ϫ/Ϫ Bcl-xL for PKC (black) and C57BL/6 (gray) T cells. Specific

staining for Bcl-xL was confirmed us- ing an isotype control Ab. These data are representative of three indepen- dent experiments. Downloaded from

Ϫ/Ϫ Ϫ/Ϫ ␪ Bcl-xL should also be impaired in the c-Rel , but not the p50 PKC to affect Bcl-xL expression. Our laboratory has previously T cells. Accordingly, the expression of Bcl-xL in T cells from described a transgenic model that expresses active PKB in T cells c-RelϪ/Ϫ and p50Ϫ/Ϫ mice was assessed by flow cytometry. In- using the CD2 promoter. We have characterized T cells in these

tracellular Bcl-xL staining confirmed the specific role of c-Rel in mice and have shown that they have increased survival to multiple ␬ http://www.jimmunol.org/ the expression of Bcl-xL (Fig. 4). The mean fluorescence intensity stimuli, enhanced NF- B activation, and increased levels of Bcl-xL of Bcl-xL staining after 24 h of stimulation with anti-CD3 Ab was (24). To examine whether PKB influences Bcl-xL expression in the 16.4 for the C57BL/6 T cells, 14.86 for the p50Ϫ/Ϫ T cells, and absence of PKC␪, the human CD2-gag-PKB␣ transgenic mice Ϫ/Ϫ ␪ only 8.9 for the c-Rel T lymphocytes. The MFI of Bcl-xL stain- were bred onto the PKC -null background. The expression of ing after stimulation through CD3 and CD28 further confirmed Bcl-xL in activated T cells was then assessed by Western blot these findings, because the MFI of Bcl-xL staining was 26.72 for analysis. Ϫ/Ϫ the wild-type T cells, 27.31 for the p50 T cells, and 10.86 for Active PKB rescued the expression of Bcl-xL in the absence of Ϫ/Ϫ ␪ the c-Rel T cells. Thus, our data obtained by intracellular stain- PKC . As can be seen in Fig. 5A, the expression of Bcl-xL in ␬ ␪Ϫ/Ϫ

ing indicate that the c-Rel, but not the p50 subunit of NF- B plays activated PKC PKB T cells was higher than that of the by guest on September 27, 2021 ␪Ϫ/Ϫ a nondispensable role in the up-regulation of Bcl-xL in activated T PKC T cells after both 16 and 24 h of stimulation. The level lymphocytes.

Active PKB can rescue Bcl-xL expression in the absence of PKC␪ in CD8ϩ T cells Multiple studies (21–24, 50) have demonstrated that PKB can en- hance the activation of NF-␬B. Studies by Kane et al. (23, 50) have suggested that PKB exerts a greater affect upon the transcription of NF-␬B-dependent genes, such as IL-2, when it is activated in con- junction with other signaling pathways, particularly PKC␪. Ac- cordingly, we wanted to establish whether transgenic expression of

active PKB could rescue the expression of Bcl-xL in T cells lack- ing PKC␪, or whether alternatively, PKB requires the presence of

FIGURE 5. Transgenic expression of PKB can rescue the expression of ϩ ␪Ϫ/Ϫ ϫ 6 Bcl-xL in CD8 PKC T cells. A, Purified splenic T cells (2 10 cells) from PKC␪Ϫ/Ϫ (␪), PKC␪Ϫ/Ϫ PKB (␪P), and C57BL/6 (B) mice were stimulated with plate-bound Ab (1 ␮g/ml anti-CD3 combined with 1 ␮ g/ml anti-CD28 Ab) for the indicated times. Bcl-xL expression was de- termined by SDS-PAGE and Western blot analysis. B, Purified splenic T Ϫ/Ϫ ϫ 6 ␪Ϫ/Ϫ ␪Ϫ/Ϫ FIGURE 4. Impaired induction of Bcl-xL in c-Rel T cells. Purified cells (1 10 cells) from PKC , littermate PKC PKB, and splenic T cells (1 ϫ 106 cells) from c-RelϪ/Ϫ (red histogram), p50Ϫ/Ϫ (blue C57BL/6 mice were stimulated with plate-bound Ab. Cells were stimulated histogram), and C57BL/6 (black histogram) mice were stimulated with with 2 ␮g/ml anti-CD3 Ab combined with 2 ␮g/ml anti-CD28 Ab for 24 h. ␮ ϩ ϩ plate-bound Ab. Cells were stimulated with 2 g/ml anti-CD3 Ab or 2 Bcl-xL expression in CD4 and CD8 T cells was then determined by flow ␮ ␮ Ϯ g/ml anti-CD3 combined with 2 g/ml anti-CD28 Ab for 24 h. Bcl-xL cytometry. The average MFI of Bcl-xL staining of triplicate samples SDs expression was then determined by flow cytometry. Specific staining was are plotted. Specific staining for Bcl-xL was confirmed using an isotype confirmed using an isotype control Ab (dotted histogram). These data are control Ab (data not shown). These data are representative of three inde- representative of three independent experiments. pendent experiments. 2936 DIFFERENTIAL PROGRAMMING OF CD4ϩ AND CD8ϩ T CELL SURVIVAL

␪Ϫ/Ϫ of Bcl-xL expression in the PKC PKB T cells, however, was

slightly reduced as compared with the amount of Bcl-xL expressed by the C57BL/6 control cells. Thus, although transgenic expres-

sion of PKB could rescue the expression of Bcl-xL in cells lacking PKC␪, this rescue did not appear to be complete.

Given that we had found a difference in the expression of Bcl-xL in CD4ϩ and CD8ϩ cells (Fig. 2), we next examined whether PKB ϩ ϩ had a differential ability to restore Bcl-xL in CD4 and CD8 T cells that lacked PKC␪. Interestingly, as can be seen in Fig. 5B, ϩ active PKB was unable to rescue the expression of Bcl-xL in CD4

T cells, because the average MFI of Bcl-xL staining in the PKC␪Ϫ/Ϫ PKB CD4ϩ T lymphocytes was comparable to that of ␪Ϫ/Ϫ the PKC T cells. Conversely, the average MFI of Bcl-xL staining in the PKC␪Ϫ/Ϫ PKB CD8ϩ T cells was 15.6 as compared with 10.0 for the PKC␪Ϫ/Ϫ CD8ϩ T lymphocytes. This difference is statistically significant ( p Ͻ 0.0006 by two-tailed Student’s t

test) and demonstrates that active PKB can rescue Bcl-xL expres- ϩ ␪ sion in CD8 T cells lacking PKC . This rescue of Bcl-xL ex- Downloaded from pression, however, is not total, because the MFI of Bcl-xL staining in the control C57BL/6 CD8ϩ T was 22.4. These data demonstrate that in CD8ϩ T cells, PKB can still promote the expression of ␪ Bcl-xL in the absence of PKC . This suggests that there exists a

signaling pathway that links PKB to Bcl-xL expression that does not require PKC␪ in CTL. http://www.jimmunol.org/

PKB partially restores cell viability in CD4ϩ PKC␪Ϫ/Ϫ T cells Next, we examined whether PKB expression was sufficient to res- cue cell survival. Like in the previous survival experiments, the viability of in vitro-activated T cells from C57BL/6, PKC␪Ϫ/Ϫ, and PKC␪Ϫ/Ϫ PKB mice was assessed by flow cytometry using annexin V and 7AAD staining 20 h after their removal from plate-

bound Ab stimulation. Fig. 6A shows the percentage of viable by guest on September 27, 2021 CD4ϩ and CD8ϩ cells in each culture after their removal from

stimulation (viability at t0) as well as after 20 h of reculture. These data show that PKB was able to rescue partially the survival of PKC␪Ϫ/Ϫ CD4ϩ T cells. When the survival in each culture was FIGURE 6. Expression of active PKB partially restores CD4ϩ T cell survival. Splenocytes (1 ϫ 107 cells) from PKC␪Ϫ/Ϫ, PKC␪Ϫ/Ϫ PKB, and normalized to the viability at t0 (data not shown), the average viability of the PKC␪Ϫ/Ϫ CD4ϩ T cells was 30.0 vs 49.1% for the C57BL/6 mice were stimulated with anti-CD3 and anti-CD28 Abs (2 ␮ PKC␪Ϫ/Ϫ PKB CD4ϩ T cells. This difference is statistically sig- g/ml of each) for 72 h. Cell viability was assessed by flow cytometry by Ͻ staining with annexin V and 7AAD to determine background death (via- nificant ( p 0.003 by two-tailed Student’s t test) and demon- ϫ 5 bility at t0). Cells were then washed and recultured (2 10 cells) in strates that the PKB transgene can affect cell viability. The effect triplicate without stimulation. After 20 h of reculture, cell viability was is through a pathway that does not alter Bcl-xL expression, because assessed by flow cytometry by staining with annexin V and 7AAD. FACS we demonstrated active PKB does not restore Bcl-xL expression in plots at each time point with the nonadjusted percentage of viable cells CD4ϩ T cells (Fig. 5). This rescue of cell survival, however, is not (percentage of annexin VϪ7AADϪ cells) are shown in A. Splenocytes from Ϫ Ϫ Ϫ Ϫ ϩ Ϫ complete. The PKC␪Ϫ/Ϫ PKB CD4ϩ T cells were not as viable P14 PKC␪ / , P14 PKC␪ / PKB, and P14 PKC␪ / mice (2 ϫ 105 cells/ after 20 h as the C57BL/6 CD4ϩ T cells, which had an average of well) were incubated with increasing concentrations of p33 peptide. Pro- 3 69.5% live cells in culture relative to their viability at t . Thus, liferation measured by the incorporation of [ H]thymidine after 72 h is 0 shown in B. These data are representative of four independent experiments. active PKB expression only partially restored T cell survival in ␪Ϫ/Ϫ ϩ PKC CD4 T cells through a Bcl-xL-independent pathway. In CD8ϩ cells, the PKB transgene had a reduced effect on cell Ϫ/Ϫ viability. The PKC␪Ϫ/Ϫ CD8ϩ T cells averaged 4.2% living cells PKB-mediated signals do not rescue proliferation in PKC␪ in culture vs 10.6% for the PKC␪Ϫ/Ϫ PKB CD8ϩ T cells. Al- T cells though this difference is statistically significant ( p Ͻ 0.02 by two- T cells lacking PKC␪ display poor proliferative responses in vitro Ϫ Ϫ tailed Student’s t test), the average viability of the PKC␪ / PKB when stimulated with Abs (33, 34) or cognate peptide Ag (35). We ϩ CD8 T cells was still reduced greatly as compared with the also wanted to determine whether active PKB could provide ap- ϩ C57BL/6 CD8 T cell cultures. The control cell cultures had an propriate signals to restore the proliferative defect in vitro. To do average of 61.9% viable cells after 20 h relative to the viability at these experiments, P14 transgenic mice expressing a TCR specific

t0. Thus, although PKB could rescue Bcl-xL expression in for the lymphocytic choriomeningitis virus glycoprotein peptide, PKC␪Ϫ/Ϫ CD8ϩ T cells (Fig. 5), it was unable to markedly im- gp33, presented in the context of H-2Db were bred with PKC␪Ϫ/Ϫ prove the cell survival of activated PKC␪Ϫ/Ϫ CD8ϩ T cells in and PKC␪Ϫ/Ϫ PKB mice. Spleen cells from P14 PKC␪ϩ/Ϫ, P14 vitro. These findings further emphasize the different mechanisms PKC␪Ϫ/Ϫ, and P14 PKC␪Ϫ/Ϫ PKB mice were then cultured with that modulate CD4ϩ and CD8ϩ T cell survival. varying concentrations of the gp33 peptide and proliferation was The Journal of Immunology 2937

T cells as compared with activation with Abs alone (data not shown). These data suggest that the survival-promoting effects of exogenous IL-2 we observed in the c-RelϪ/Ϫ and PKC␪Ϫ/Ϫ T

lymphocytes were independent of Bcl-xL expression.

Discussion Signaling molecules promoting T cell survival The signaling pathways that govern activated T cell survival have FIGURE 7. Exogenous IL-2 rescues the survival of activated PKC␪Ϫ/Ϫ yet to be elucidated fully. This is the first study that directly com- ϩ and c-RelϪ/Ϫ T cells. Splenocytes (1 ϫ 107 cells) from PKC␪Ϫ/Ϫ, pares the role of PKC␪, c-Rel, and PKB in the survival of CD4 ϩ ϩ c-RelϪ/Ϫ, and C57BL/6 mice were stimulated with anti-CD3 and anti- and CD8 T cells. Our study has shown that both CD4 and CD28 Abs (2 ␮g/ml of each) for 72 h. Cell viability was assessed by flow CD8ϩ T lymphocytes require PKC␪-derived signals to survive cytometry by staining with annexin V and 7AAD to determine background (Fig. 1A). In the absence of PKC␪, however, CD4ϩ T cell survival death (viability at t0). The average viability at t0 of the cells ranged from can be restored partially by PKB (Fig. 6), whereas PKB does not ϫ 5 65 to 84%. Cells were then washed and recultured (2 10 cells) in have a major impact upon the survival of CD8ϩ T cells. These data triplicate without stimulation in the presence or absence of 10 U/ml IL-2. demonstrate a different requirement for PKB-mediated survival After 20 h of reculture, cell viability was assessed by flow cytometry by ϩ ϩ staining with annexin V and 7AAD. The data are expressed as the per- signals in CD4 and CD8 T cells. These findings also indicate that PKC␪ and PKB signal independently of each other to promote Downloaded from centage of viable cells after 20 h of reculture divided by the percentage of ϩ viable cells at t (after stimulation). The data are shown as an average value CD4 T cell survival. The importance of active PKB for the sur- 0 ϩ of triplicate wells with error bars showing SD. These data are representa- vival of CD4 T lymphocytes is supported by studies by Song tive of five independent experiments. et al. (54). These authors demonstrated that signals activating PKB are important in maintaining the long-term survival of CD4ϩ T cells in vivo (55). 3

measured by [ H]thymidine incorporation at different time points. Although our results indicate the PKC␪ is required for the sur- http://www.jimmunol.org/ Fig. 6B shows that the PKB transgene could not rescue the pro- vival of both CD4ϩ and CD8ϩ T lymphocytes, our data also in- Ϫ Ϫ liferation of the PKC␪ / T cells to a level comparable to the dicate that PKC␪ delivers a more vital survival signal in activated ϩ Ϫ PKC␪ / after 72 h of stimulation. These findings demonstrate CD8ϩ T lymphocytes than in CD4ϩ T cells. CD8ϩ cells lacking

that neither the restoration of Bcl-xL expression nor the potential PKC␪ display a much more striking impairment in survival than do alternative signals provided by PKB is sufficient to restore the CD4ϩ PKC␪Ϫ/Ϫ T cells (Fig. 1A). The importance of PKC␪ in Ϫ Ϫ proliferative response to Ag in PKC␪ / T cells in vitro. CD8ϩ T cell survival in vivo has been illustrated previously by work by Barouch-Bentov et al. (36). Our data confirm the findings ␪Ϫ/Ϫ Ϫ/Ϫ IL-2 rescues the survival of PKC and c-Rel T cells of Barouch-Bentov et al. (36) and further suggest that c-Rel is Because the expression of IL-2 has been linked to PKC␪ signals required to relay PKC␪-mediated survival signals in activated by guest on September 27, 2021 (33) and to the c-Rel transcription factor (40, 51), we tested CD8ϩ T cells, but not in CD4ϩ T cells (Fig. 1B). This finding is whether the addition of exogenous IL-2 was able to promote the consistent with a previous report that has linked PKC␪ to the mo- survival of PKC␪Ϫ/Ϫ and c-RelϪ/Ϫ T cells. To do this, in vitro- bilization of c-Rel (56). It is also consistent with studies that have activated splenic T cells from PKC␪Ϫ/Ϫ, c-RelϪ/Ϫ, and C57BL/6 demonstrated a more profound impact upon activated CD8ϩ T cell mice were cultured for 20 h in the presence or absence of 10 U/ml survival than activated CD4ϩ T cell survival in mice with impaired IL-2. Cell viability was then assessed by flow cytometry using ability to activate NF-␬B (30, 57). The target genes of c-Rel that annexin V and 7AAD staining. As can be seen in Fig. 7, the ad- are important for the survival of CD8ϩ T lymphocytes remain to dition of exogenous IL-2 was able to restore the survival of PKC␪- be elucidated fully. ϩ ␪Ϫ/Ϫ deficient CD4 T cells. The average survival of the PKC A multitude of studies have suggested that Bcl-xL is the key CD4ϩ T cells in the presence of IL-2 was 72.0%. Although this prosurvival molecule of activated T cells (4, 47, 58). Given that ϩ was slightly lower than the average survival of the control CD4 our results indicate that the expression of Bcl-xL is affected by both T cells, it represents a much more substantial rescue of cell via- PKC␪ (Figs. 2 and 3) and c-Rel (Fig. 4), we suspected that the bility than was provided by the PKB transgene (Fig. 6A). Simi- diminished survival of CD8ϩ T cells we observed (Fig. 1) was due ϩ larly, in the case of the CD8 lymphocytes, IL-2 had a profound to an impaired ability to express Bcl-xL. Thus, we were surprised

effect upon survival. The addition of IL-2 was able to rescue fully to find that rescuing Bcl-xL expression with the PKB transgene in the survival defect in both the c-RelϪ/Ϫ and PKC␪Ϫ/Ϫ CD8ϩ T cytotoxic lymphocytes did not significantly alter their survival Ϫ/Ϫ cells. The average survival of the c-Rel T lymphocytes was (Fig. 6). These data question the importance of Bcl-xL expression actually slightly enhanced compared with the average survival of downstream of PKC␪ and c-Rel for the survival of activated CD8ϩ the wild-type CD8ϩ T cells cultured with IL-2. Collectively, these T lymphocytes. Consistent with these findings, Zhang and He (59)

data demonstrate that unlike PKB, IL-2 can restore fully the sur- recently reported that Bcl-xL is dispensable for the survival of vival of T cells lacking PKC␪ or c-Rel. CD8ϩ T cells. Our results would agree with these data and suggest Because signaling through the IL-2R has been linked to the that perhaps IL-2 is the important c-Rel target gene downstream of ␪ ϩ expression of Bcl-xL (52, 53), we wanted to ensure that the addi- PKC for the survival of CD8 T lymphocytes (Fig. 7). The im- ␪ tion of exogenous IL-2 was not altering the expression of Bcl-xL portance of IL-2 gene expression downstream of PKC in sup- in activated c-RelϪ/Ϫ and PKC␪Ϫ/Ϫ T cells. To do this, purified porting the survival of CD8ϩ T cells, however, may be dependent splenic T cells from c-RelϪ/Ϫ and PKC␪Ϫ/Ϫ mice were stimulated upon the context of T cell activation. Work in our own laboratory for 24 h with plate-bound Ab in the presence or absence of IL-2. (35) and that of others (60) has demonstrated that the absence of ␪ ϩ After 24 h, the expression of Bcl-xL was assessed using flow cy- the entire PKC signaling pathway in CD8 T cells can be over- tometry. The addition of either 10 or 50 U/ml IL-2 did not alter the come via the provision of strong pathogen-derived signals in vivo. Ϫ/Ϫ ␪Ϫ/Ϫ ␪ expression of Bcl-xL in either the activated c-Rel or PKC PKC -mediated survival signals, however, may be more important 2938 DIFFERENTIAL PROGRAMMING OF CD4ϩ AND CD8ϩ T CELL SURVIVAL when pathogen-mediated signals are absent, such as in the context Acknowledgments of autoimmunity or during an antitumor response. We thank Rosa Pileggi and Sandra McGugan for administrative help. Bcl-x expression is not sufficient to restore proliferation L Disclosures ϩ Previous work by Zheng et al. (61) demonstrated that CD4 T The authors have no financial conflict of interest. cells lacking both p50 and c-Rel did not proliferate when stimu- lated. These cells failed to induce the expression of Bcl-xL as well References as demonstrating impaired c- expression upon stimulation. 1. Werlen, G., B. Hausmann, D. Naeher, and E. Palmer. 2003. Signaling life and Thus, these authors concluded that the reasons for failed prolifer- death in the thymus: timing is everything. Science 299: 1859–1863. 2. Steinman, R. M., D. Hawiger, and M. C. Nussenzweig. 2003. Tolerogenic den- ation were 2-fold. Not only were the cells unable to induce the dritic cells. Annu. Rev. Immunol. 21: 685–711. genes required for survival, but they also did not initiate the ap- 3. Marrack, P., and J. Kappler. 2004. Control of T cell viability. Annu. Rev. Immu- propriate genetic program required to enter . Our results nol. 22: 765–787. 4. Boise, L. H., A. J. Minn, P. J. Noel, C. H. June, M. A. Accavitti, T. Lindsten, and support the conclusion that restoring Bcl-xL expression alone is C. B. Thompson. 1995. CD28 costimulation can promote T cell survival by insufficient to rescue proliferation, because returning the expres- enhancing the expression of Bcl-xL. Immunity 3: 87–98. ␪Ϫ/Ϫ ϩ 5. Irmler, M., M. Thome, M. Hahne, P. Schneider, K. Hofmann, V. Steiner, J.-L. sion of Bcl-xL in PKC CD8 T cells was unable to enhance Bodmer, M. Schroter, K. Burns, C. Mattmann, et al. 1997. Inhibition of death proliferation (Fig. 6B). Similar conclusions have been reached in receptor signals by cellular FLIP. Nature 388: 190–195. other systems. In B lymphocytes lacking c-Rel, expression of 6. Chau, H., V. Wong, N. J. Chen, H. L. Huang, W. J. Lin, C. Mirtsos, A. R. Elford, M. Bonnard, A. Wakeham, T. You, et al. 2005. Cellular FLICE-inhibitory protein Bcl-xL was not sufficient to restore proliferation. Expression of is required for T cell survival and cycling. J. Exp. Med. 202: 405–413. Downloaded from cylin E in conjunction with Bcl-xL was required to restore prolif- 7. Zhang, N., and Y. W. He. 2005. An essential role for c-FLIP in the efficient eration (62). As well, in T cells, retroviral expression of Bcl-x in development of mature T lymphocytes. J. Exp. Med. 202: 395–404. L 8. Woodgett, J. R. 2005. Recent advances in the protein kinase B signaling pathway. T cells lacking CD28 did not rescue proliferation (63). Collec- Curr. Opin. Cell Biol. 17: 150–157. 9. Scheid, M. P., P. A. Marignani, and J. R. Woodgett. 2002. Multiple phospho- tively, these findings indicate that expression of Bcl-xL alone is inositide 3-kinase-dependent steps in activation of protein kinase B. Mol. Cell. insufficient to promote proliferation. Moreover, because we used Biol. 22: 6247–6260. the active PKB transgene to increase Bcl-xL expression, our results 10. Datta, S. R., H. Dudek, X. Tao, S. Masters, H. Fu, Y. Gotoh, and M. E. Greenberg. 1997. Akt phosphorylation of BAD couples survival signals to also suggest that in the absence of PKC␪, signals downstream of http://www.jimmunol.org/ the cell-intrinsic death machinery. Cell 91: 231–241. PKB are unable to affect proliferation. This result is interesting 11. Del Peso, L., M. Gonzalez-Garcia, C. Page, R. Herrera, and G. Nunez. 1997. because PKB has many downstream effector molecules and has Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt. been demonstrated previously to promote proliferation, in part via Science 278: 687–689. 12. Blume-Jensen, P., R. Janknecht, and T. Hunter. 1998. The receptor promotes the phosphorylation of the cell cycle inhibitor p27 (19, 20). cell survival via activation of PI 3-kinase and subsequent Akt-mediated phos- phorylation of Bad on Ser136. Curr. Biol. 8: 779–782. Unique functions of NF-␬B family members 13. Brunet, A., A. Bonni, M. J. Zigmond, M. Z. Lin, P. Juo, L. S. Hu, M. J. Anderson, K. C. Arden, J. Blenis, and M. E. Greenberg. 1999. Akt promotes cell survival by In this study, we have demonstrated that c-Rel is able to provide an phosphorylating and inhibiting a Forkhead transcription factor. Cell 96: 857–868. important survival signal in CD8ϩ T cells. Recent work in our 14. Rena, G., S. Guo, S. C. Cichy, T. G. Unterman, and P. Cohen. 1999. Phosphor-

ylation of the transcription factor forkhead family member FKHR by protein by guest on September 27, 2021 laboratory has established that p50 is required for protection kinase B. J. Biol. Chem. 274: 17179–17183. against Fas-mediated apoptosis, whereas c-Rel is dispensable for 15. Kops, G. J., N. D. de Ruiter, A. M. De Vries-Smits, D. R. Powell, J. L. Bos, and this protection (31); now, we have demonstrated a survival signal- B. M. Burgering. 1999. Direct control of the Forkhead transcription factor AFX by protein kinase B. Nature 398: 630–634. ing pathway engaged during T cell activation in which the con- 16. Dijkers, P. F., R. H. Medema, J.-W. J. Lammers, L. Koenderman, and P. J. Coffer. verse is true: c-Rel is required, whereas p50 is dispensable. This 2000. Expression of the pro-apoptotic Bcl-2 family member Bim is regulated by the forkhead transcription factor FKHR-L1. Curr. Biol. 10: 1201–1204. pattern also holds true for the expression of Bcl-xL. We demon- 17. Scheid, M. P., and J. R. Woodgett. 2001. PKB/AKT: functional insights from strate c-Rel is involved in the expression of Bcl-xL upon the ac- genetic models. Nat. Rev. Mol. Cell Biol. 2: 760–768. tivation of T cells, whereas p50 is not required (Fig. 4). These data 18. Kane, L. P., M. N. Mollenauer, Z. Xu, C. W. Turck, and A. Weiss. 2002. Akt- dependent phosphorylation specifically regulates Cot induction of NF-␬B-depen- indicate that the signaling cascade leading to the expression of dent transcription. Mol. Cell. Biol. 22: 5962–5974. Bcl-xL in T cells is similar to the one described in B cells. 19. Liang, J., J. Zubovitz, T. Petrocelli, R. Kotchetkov, M. K. Connor, K. Han, In B lymphocytes, the PKC␤ isoform is required to activate the J. H. Lee, S. Ciarallo, C. Catzavelos, R. Beniston, et al. 2002. PKB/Akt phos- phorylates p27, impairs nuclear import of p27 and opposes p27-mediated G1 IKK complex and initiate the expression of Bcl-xL (64, 65). As arrest. Nat. Med. 8: 1153–1160. well, c-Rel has also been demonstrated to be essential for the ex- 20. Shin, I., F. M. Yakes, F. Rojo, N. Y. Shin, A. V. Bakin, J. Baselga, and C. L. Arteaga. 2002. PKB/Akt mediates cell-cycle progression by phosphoryla- pression of Bcl-xL in B lymphocytes stimulated through the BCR tion of p27Kip1 at threonine 157 and modulation of its cellular localization. Nat. or CD40 (66, 67). Our data, and those from the B cell studies, Med. 8: 1145–1152. indicate that a signaling paradigm linking PKC isoforms to Bcl-x 21. Ozes, O. N., L. D. Mayo, J. A. Gustin, S. R. Pfeffer, L. M. Pfeffer, and L D. B. Donner. 1999. NF-␬B activation by tumor necrosis factor requires the Akt through the induction of c-Rel may be conserved in cells of lym- serine-threonine kinase. Nature 401: 82–85. phoid, and potentially, all of the hemopoietic lineages. This model 22. Romashkova, J. A., and S. S. Makarov. 1999. NF-␬B is a target of AKT in would fit with the expression of c-Rel, because its expression is anti-apoptotic PDGF signalling. Nature 401: 86–90. 23. Kane, L. P., V. S. Shapiro, D. Stokoe, and A. Weiss. 1999. Induction of NF-␬B confined to the hemopoietic compartment (68, 69). by the Akt/PKB kinase. Curr. Biol. 9: 601–604. 24. Jones, R. G., M. Parsons, M. Bonnard, V. S. F. Chan, W. C. Yeh, J. R. Woodgett, Concluding remarks and P. S. Ohashi. 2000. Protein kinase B (PKB) regulates T lymphocytes sur- ␬ J. Exp. Med. ϩ ϩ vival, Bcl-XL levels, and NF- B activation in vivo. 191: 1721–1733. In summary, this work demonstrates that CD4 and CD8 T cell 25. Li, Q., and I. M. Verma. 2002. NF-␬B regulation in the immune system. Nat. Rev. survival is differentially regulated and is not strictly governed by Immunol. 2: 725–734. 26. Chen, C., L. C. Edelstein, and C. Gelinas. 2000. The Rel/NF-␬B family directly Bcl-x expression. In CD4ϩ T lymphocytes, PKC␪ provides a sub- L activates expression of the apoptosis inhibitor Bcl-xL. Mol. Cell. Biol. 20: set of survival signals that can be augmented by PKB and restored by 2687–2695. ␪ ϩ 27. Khoshnan, A., C. Tindell, I. Laux, D. Bae, C. L. Bennett, and A. E. Nel. 2000. IL-2. In contrast, PKC and c-Rel are vital to CD8 T cell survival, ␬ The NF- B cascade is important in Bcl-xL expression and for the anti-apoptotic whereas PKB plays a limited role in the survival of CTL. Delineation effects of the CD28 receptor in primary human CD4ϩ lymphocytes. J. Immunol. of all of the differences in the CD4ϩ vs CD8ϩ survival programs 165: 1743–1754. 28. Lee, H. W., S. J. Park, B. K. Choi, H. H. Kim, K. O. Nam, and B. S. Kwon. 2002. requires further investigation, but may reveal potential subset-specific 4-1BB promotes the survival of CD8ϩ T lymphocytes by increasing expression therapeutic targets. of Bcl-xL and Bfl-1. J. Immunol. 169: 4882–4888. The Journal of Immunology 2939

29. Dudley, E., F. Hornung, L. Zheng, D. Scherer, D. Ballard, and M. Lenardo. 1999. 50. Kane, L. P., P. G. Andres, K. C. Howland, A. K. Abbas, and A. Weiss. 2001. Akt NF-␬B regulates Fas/APO-1/CD95- and TCR-mediated apoptosis of T lympho- provides the CD28 costimulatory signal for up-regulation of IL-2 and IFN-␥ but cytes. Eur. J. Immunol. 29: 878–886. not TH2 cytokines. Nat. Immunol. 2: 37–44. 30. Mora, A. L., R. A. Corn, A. K. Stanic, S. Goenka, M. Aronica, S. Stanley, 51. Rao, S., S. Gerondakis, D. Woltring, and M. F. Shannon. 2003. c-Rel is required D. W. Ballard, S. Joyce, and M. Boothby. 2003. Antiapoptotic function of NF-␬B for chromatin remodeling across the IL-2 gene promoter. J. Immunol. 170: in T lymphocytes is influenced by their differentiation status: roles of Fas, c-FLIP, 3724–3731. and Bcl-xL. Cell Death Differ. 10: 1032–1044. 52. Lord, J. D., B. C. McIntosh, P. D. Greenberg, and B. H. Nelson. 1998. The IL-2 31. Jones, R. G., S. D. Saibil, J. M. Pun, A. R. Elford, M. Bonnard, M. Pellegrini, receptor promotes proliferation, bcl-2 and bcl-x induction, but not cell viability S. Arya, M. E. Parsons, C. M. Krawczyk, S. Gerondakis, et al. 2005. NF-␬B through the adapter molecule Shc. J. Immunol. 161: 4627–4633. couples protein kinase B/Akt signaling to distinct survival pathways and the 53. Lord, J. D., B. C. McIntosh, P. D. Greenberg, and B. H. Nelson. 2000. The IL-2 regulation of lymphocyte homeostasis in vivo. J. Immunol. 175: 3790–3799. receptor promotes lymphocyte proliferation and induction of the c-myc, bcl-2, ␪ 32. Isakov, N., and A. Altman. 2002. Protein kinase C in T cell activation. Annu. and bcl-x genes through the trans-activation domain of Stat5. J. Immunol. 164: Rev. Immunol. 20: 761–794. 2533–2541. 33. Sun, Z., C. W. Arendt, W. Ellmeier, E. M. Schaeffer, M. J. Sunshine, L. Gandhi, 54. Song, J., S. Salek-Ardakani, P. R. Rogers, M. Cheng, L. van Parijs, and M. Croft. J. Annes, D. Petrzilka, A. Kupfer, P. L. Schwartzberg, and D. R. Littman. 2000. ␪ ␬ 2004. The costimulation-regulated duration of PKB activation controls T cell PKC- is required for TCR-induced NF- B activation in mature but not immature longevity. Nat. Immunol. 5: 150–158. T lymphocytes. Nature 404: 402–407. 55. Rogers, P. R., J. Song, I. Gramaglia, N. Killeen, and M. Croft. 2001. OX40 34. Pfeifhofer, C., K. Kofler, T. Gruber, N. G. Tabrizi, C. Lutz, K. Maly, M. Leitges, ϩ promotes Bcl-x and Bcl-2 expression and is essential for long-term survival of and G. Baier. 2003. Protein kinase C␪ affects Ca2 mobilization and NFAT cell L CD4 T cells. Immunity 15: 445–455. activation in primary mouse T cells. J. Exp. Med. 197: 1525–1535. 35. Berg-Brown, N. N., M. A. Gronski, R. G. Jones, A. R. Elford, 56. Grumont, R., P. Lock, M. Mollinari, F. M. Shannon, A. Moore, and E. K. O. B. Deenick, D. R. Littman, and P. S. Ohashi. 2004. PKC␪ signals S. Gerondakis. 2004. The mitogen-induced increase in T cell size involves PKC ␬ activation versus tolerance in vivo. J. Exp. Med. 199: 743–744. and NFAT activation of Rel/NF- B-dependent c-myc expression. Immunity 21: 36. Barouch-Bentov, R., E. E. Lemmens, J. Hu, E. M. Janssen, N. M. Droin, J. Song, 19–30. S. P. Schoenberger, and A. Altman. 2005. Protein kinase C-␪ is an early survival 57. Boothby, M. R., A. L. Mora, D. C. Scherer, J. A. Brockman, and D. W. Ballard. factor required for differentiation of effector CD8ϩ T cells. J. Immunol. 175: 1997. Perturbation of the T lymphocyte lineage in transgenic mice expressing a Downloaded from ␬ 5126–5134. constitutive repressor of nuclear factor (NF)- B. J. Exp. Med. 185: 1897–1907. 37. Bauer, B., N. Krumbo¨ck, F. Fresser, F. Hochholdinger, M. Spitaler, A. Simm, 58. Song, A., X. Tang, K. M. Harms, and M. Croft. 2005. OX40 and Bcl-xL promote F. U¨ berall, B. Schraven, and G. Baier. 2001. Complex formation and cooperation the persistence of CD8 T cells to recall tumor-associated antigen. J. Immunol. of protein kinase C␪ and Akt1/protein kinase B␣ in the NF-␬B transactivation 175: 3534–3541. cascade in Jurkat T cells. J. Biol. Chem. 276: 31627–31634. 59. Zhang, N., and Y. W. He. 2005. The antiapoptotic protein Bcl-xL is dispensable 38. Bauer, B., M. Jenny, F. Fresser, F. U¨ berall, and G. Baier. 2003. AKT1/PKB␣ is for the development of effector and memory T lymphocytes. J. Immunol. 174: recruited to lipid rafts and activated downstream of PKC isotypes in CD3-induced 6967–6973.

T cell signaling. FEBS Lett. 541: 155–162. 60. Marsland, B. J., C. Nembrini, N. Schmitz, B. Abel, S. Krautwald, http://www.jimmunol.org/ 39. Sha, W. C., H.-C. Liou, E. L. Tuomanen, and D. Baltimore. 1995. Targeted M. F. Bachmann, and M. Kopf. 2005. Innate signals compensate for the absence disruption of the p50 subunit of NF-␬B leads to multifocal defects in immune of PKC-␪ during in vivo CD8ϩ T cell effector and memory responses. Proc. Natl. responses. Cell 80: 321–330. Acad. Sci. USA 102: 14374–14379. 40. Kontgen, F., R. J. Grumont, A. Strasser, D. Metcalf, R. Li, D. Tarlinton, and 61. Zheng, Y., M. Vig, J. Lyons, L. van Parijs, and A. A. Beg. 2003. Combined S. Gerondakis. 1995. Mice lacking the c-rel proto- exhibit defects in deficiency of p50 and cRel in CD4ϩ T cells reveals an essential requirement for lymphocyte proliferation, humoral immunity and interleukin-2 expression. Genes nuclear factor ␬B in regulating mature T cell survival and in vivo function. Dev. 9: 1965–1977. J. Exp. Med. 197: 861–874. 41. Pircher, H., K. Bu¨rki, R. Lang, H. Hengartner, and R. Zinkernagel. 1989. Tol- 62. Cheng, S., C. Y. Hsia, G. Leone, and H. C. Liou. 2003. and Bcl-xL erance induction in double specific T-cell receptor transgenic mice varies with cooperatively induce cell cycle progression in c-RelϪ/Ϫ B cells. Oncogene 22: antigen. Nature 342: 559–561. 8472–8486. 42. Lin, X., A. O’Mahony, Y. Mu, R. Geleziunas, and W. C. Greene. 2000. Protein 63. Dahl, A. M., C. Klein, P. G. Andres, C. A. London, M. P. Lodge, R. C. Mulligan,

␪ ␬ by guest on September 27, 2021 kinase C- participates in NF- B activation induced by CD3-CD28 costimulation and A. K. Abbas. 2000. Expression of Bcl-x restores cell survival, but not ␬ ␤ L through selective activation of I B kinase . Mol. Cell. Biol. 20: 2933–2940. proliferation and effector differentiation, in CD28-deficient T lymphocytes. ␬ 43. Coudronniere, N., M. Villalba, N. Englund, and A. Altman. 2000. NF- B acti- J. Exp. Med. 191: 2031–2037. vation induced by T cell receptor/CD28 costimulation is mediated by protein 64. Saijo, K., I. Mecklenbra¨uker, A. Santana, M. Leitger, C. Schmedt, and kinase C-␪. Proc. Natl. Acad. Sci. USA 97: 3394–3399. A. Tarakhovsky. 2002. Protein kinase C␤ controls nuclear factor ␬B activation in 44. Grillot, D. A., M. Gonzalez-Garcia, D. Ekhterae, L. Duan, N. Inohara, S. Ohta, B cells through selective regulation of the I␬B kinase ␣. J. Exp. Med. 195: M. F. Seldin, and G. Nunez. 1997. Genomic organization, promoter region anal- 1647–1652. ysis, and chromosome localization of the mouse bcl-x gene. J. Immunol. 158: 65. Su, T. T., B. Guo, Y. Kawakami, K. Sommer, K. Chae, L. A. Humphries, 4750–4757. ␤ ␬ 45. Glasgow, J. N., T. Wood, and J. R. Perez-Polo. 2000. Identification and charac- R. M. Kato, S. Kang, L. Patrone, R. Wall, et al. 2002. PKC- controls I B kinase terization of nuclear factor ␬B binding sites in the murine bcl-x promoter. J. Neu- lipid raft recruitment and activation in response to BCR signaling. Nat. Immunol. rochem. 75: 1377–1389. 3: 780–786. ␬ 46. Veis, D. J., C. L. Sentman, E. A. Bach, and S. J. Korsmeyer. 1993. Expression 66. Lee, H. H., H. Dadgostar, Q. Cheng, J. Shu, and G. Cheng. 1999. NF- B-me- of the bcl-2 protein in murine and human thymocytes and in peripheral T lym- diated up-regulation of Bcl-x and Bfl-1/A1 is required for CD40 survival signal- phocytes. J. Immunol. 151: 2546–2554. ing in B lymphocytes. Proc. Natl. Acad. Sci. USA 96: 9136–9141. 47. Broome, H. E., C. M. Dargan, S. Krajewski, and J. C. Reed. 1995. Expression of 67. Owyang, A. M., J. R. Tumang, B. R. Schram, C. Y. Hsia, T. W. Behrens, Bcl-2, Bcl-x, and Bax after T cell activation and IL-2 withdrawal. J. Immunol. T. L. Rothstein, and H. C. Liou. 2001. c-Rel is required for the protection of B 155: 2311–2317. cells from antigen receptor-mediated, but not Fas-mediated, apoptosis. J. Immu- 48. Okkenhaug, K., L. Wu, K. M. Garza, J. La Rose, W. Khoo, B. Odermatt, nol. 167: 4948–4956. T. W. Mak, P. S. Ohashi, and R. Rottapel. 2001. A point mutation in CD28 68. Brownell, E., B. Mathieson, H. A. Young, J. Keller, J. N. Ihle, and N. R. Rice. distinguishes proliferative signals from survival signals. Nat. Immunol. 2: 1987. Detection of c-rel-related transcripts in mouse hematopoietic tissues, frac- 325–332. tionated lymphocyte populations, and cell lines. Mol. Cell. Biol. 7: 1304–1309. 49. Burr, J. S., N. D. L. Savage, G. E. Messah, S. L. Kimzey, A. S. Shaw, R. H. Arch, 69. Grumont, R. J., and S. Gerondakis. 1990. The murine c-rel proto-oncogene en- and J. M. Green. 2001. Distinct motifs within CD28 regulate T codes two mRNAs the expression of which is modulated by lymphoid stimuli. and induction of Bcl-xL. J. Immunol. 166: 5331–5335. Oncog. Res. 5: 245–254.