Activation-induced cell death (AICD) as well as programmed cell death (PCD) serve to control the expansion of activated T cells to limit untoward side effects of continued effector responses by T cells and to maintain homeostasis. AICD of T cells in tumor immunotherapy can be counterproductive particularly if the activated T cells undergo apoptotic death after the very first secondary encounter of the specific epitope. We examined the extent to which tumor epitope-specific CTLs that are activated and expanded in an in vitro-matured dendritic cell-based primary stimulation protocol undergo AICD following their first secondary encounter of the cognate epitope. Using the MART-127–35 epitope as a prototype vaccine epitope, we also examined whether these CTLs could be rescued from AICD. Our results demonstrate that a substantial fraction of MART-127–35 epitope-specific primary CTLs undergo AICD upon the very first secondary encounter of the cognate epitope. The AICD in these CTLs is neither caspase dependent nor is it triggered by the extrinsic death signaling pathways (Fas, TNFR, etc.). These CTLs, interestingly, could be rescued from AICD by the JNK inhibitor, SP600125. We also found that SP600125 interferes with their IFN-γ response but does not block their cytolytic function. The rescued CTLs, however, regain their capacity to synthesize IFN-γ if continued in culture without the inhibitor. These observations have implications in tumor immunotherapy and in further studies for regulation of AICD in CTLs.

Programmed cell death (PCD)3 and activation-induced cell death (AICD) in T cells are important physiologic processes that prevent untoward side effects of a continued and uncontrolled T cell-mediated effector response as well as maintain homeostasis (1, 2, 3). Both processes involve apoptotic deletion of T cells. PCD entails the deletion of the expanded T cell population usually during the contraction phase of the response. AICD, in contrast, involves the apoptotic deletion of a significant fraction of the activated population after an effector response. In AICD, the effector function and the death are, paradoxically, triggered by TCR-driven signaling (i.e., activation induced). In any event, both processes (i.e., PCD and AICD) are designed to serve useful purposes by limiting the expansion of activated T cells. AICD in activated T cells in tumor immunotherapy, however, can be counterproductive particularly if the activated T cells undergo apoptotic death after the very first encounter of the specific epitope.

Lately, much interest has been generated in cancer vaccine therapy with specific peptides, protein Ags, DNA, etc. Most of these immunogens are “self” Ag (4), yet many cancer patients as well as normal healthy hosts harbor precursor CTLs for such “self” epitopes. Ex vivo stimulation of T cells as well as in vivo immunization with such self peptides or tumor-associated Ags (TAA) lead to the activation and expansion of the Ag-specific CTLs (5, 6). These TAA-specific T cells are also susceptible to AICD. To our knowledge, neither the extent of AICD in these self-but-TAA reactive primary CTLs nor the feasibility of rescuing them from AICD has been carefully examined. We have recently shown that Melan-A/MART-127–35 epitope-specific CTLs expanded in an in vitro dendritic cell (DC)-based stimulation protocol undergo apoptotic death on repetitive stimulation by immature as well as by fully activated DCs (7). Using the Melan-A/MART-127–35 epitope (8, 9) as a prototype self but melanoma-associated Ag, we studied the extent of AICD in MART-127–35 epitope-specific primary CTLs and examined whether it could be prevented. In this study, we show that a large fraction of MART-127–35 epitope-specific CTLs indeed undergo AICD upon the very first secondary encounter of the cognate epitope. The AICD in these CTLs is neither caspase dependent nor is it triggered by the engagement of extrinsic death receptors. We also show that the JNK inhibitor, SP600125, can rescue a significant fraction of them from death. In the process of rescuing, the JNK inhibitor interferes with their capacity to produce IFN-γ but does not interfere with their cytolytic function. Of further interest, the rescued MART-127–35 epitope-specific CTLs, when continued in culture in IL-15 and without the inhibitor, regain capacity to synthesize IFN-γ. These observations, therefore, have implications in cancer immunotherapeutic strategies and in further studies of AICD in CTLs.

The study population consisted of HLA-A2-positive melanoma patients or healthy donors. The participants were included in this study with informed consent.

The MART-127–35 peptide (AAGIGILTV) and MAGE-3271–279 (FLWGPRALV) were purchased from Multiple Peptide Systems (San Diego, CA) while β2-microglobulin was purchased from Sigma-Aldrich (St. Louis, MO). Culture medium consisted of IMDM (Invitrogen Life Technologies, Grand Island, NY) supplemented with 10% FBS (Gemini Bioproducts, Calabasas, CA), 0.55 mM l-arginine, 0.24 mM l-asparagine (both from Invitrogen Life Technologies), 1.5 mM l-glutamine (Sigma-Aldrich), 50 U/ml penicillin, and 50 μg/ml streptomycin (both from Abbott Laboratories, North Chicago, IL). This will be referred to as complete media (CM). The TAP-deficient line, T2, was a gift of P. Cresswell (Yale University, New Haven, CT). Recombinant human GM-CSF was purchased from Immunex (Seattle, WA). Recombinant human IL rhIL-4, rhIL-2, rhIFN-γ was purchased from R&D Systems (Minneapolis, MN). LPS from Escherichia coli 055:B5 was purchased from Sigma-Aldrich. An Annexin V kit to track the early apoptotic cells for exposure of phosphatidylserine was purchased from BD Pharmingen (San Jose, CA). MART-127–35 (EAGIGILTV) tetramer labeled with PE with and without FITC-labeled anti-CD8 was purchased from Beckman Coulter (Fullerton, CA). Fluorochrome-labeled mAbs to CD25, CD27, CD28, CD95, CD95L, 4-1BB, 4-1BBL, OX-40 were purchased from BD Biosciences (San Jose, CA). Inhibitors for various kinase pathways, as SB203580 for p38 kinase, SP600125 for JNK, PD98059 for ERK were purchased from BIOMOL (Plymouth Meeting, PA). Pan caspase inhibitor, human Fas/Fc chimera, human TNF- RI/Fc chimera, human TRAIL-RI/Fc chimera, human TRAIL-RII/Fc chimera, and human IFN-γ RI/Fc chimeric proteins were purchased from R&D Systems.

The procedure for generating myeloid DCs from peripheral blood monocyte was followed. Briefly, circulating monocyte were isolated by 2 h adherence of Ficoll-Hypaque density gradient-cut PBMC as previously described (7). The adherent cells were cultured in CM with 1000 U/ml GM-CSF and 500 U/ml IL-4 for 3–5 days to obtain a population of immature DCs. Maturation of immature DCs was done by first priming in IFN-γ (1000 U/ml) for 2 h and then priming in the presence of 100 ng/ml LPS.

IFN-γ response assay for the effector cells has been described previously (10). Briefly, the effector cells were cocultured with the peptide-pulsed (1 μg/ml) T2 cells. After 4–16 h, culture supernatants were harvested and IFN-γ was measured by ELISA as per manufacturer’s protocol (R&D Systems).

The procedure for phenotypic analyses and for determining the number of epitope-specific cells with tetramer staining by flow cytometry has been described (7). To determine tetramer and annexin V-positive cells, the effector cells were stained with CD8, MART-127–35/HLA-A2 tetramer, and annexin V. The stained cells were then analyzed for single-positive vs double-positive populations in flow cytometry using a FACSCalibur and CellQuest software (BD Biosciences).

The chromium release microcytotoxicity assay has been previously described (11).

The basic procedure for peptide-loaded DC-based in vitro activation and expansion of epitope-specific CD8+ T cells has been described (7). Briefly, Ficoll-Hypaque gradient separated blood mononuclear cells were purified for CD8+ T cells (routinely exceeding 90%) by a Dynal magnetic bead isolation kit (Dynal, Oslo, Norway) and cocultured with autologous DCs pulsed with relevant peptides (100 μg/ml) and 5 μg/ml β2-microglobulin at a CD8+ T cell to DC ratio of 100. For our current purpose, because IL-2 has been known to facilitate apoptosis in activated T cells (12), the cocultures were conducted in the presence of rhIL-15 (10 ng/ml). Before setting up cocultures, the DCs were irradiated to 3000 rad. The activated CTLs were also maintained in culture in IL-15.

To test whether or not the epitope-specific primary CTLs undergo AICD, the activated CTLs were exposed to peptide (1 μg/ml) loaded T2 cells (E:T = 100) at different time points of the cultures. Thereafter, the evidence of apoptosis was determined by flow cytometry with triple color staining (CD8, MART-127–35/HLA-A2 tetramer, and annexin V) at different time points (4–18 h). Experiments were conducted in triplicate wells and significance was calculated by one-way ANOVA using Sigma Stat statistical software (Chicago, IL).

To evaluate the effect of various agents in modulating AICD, the CTLs were preincubated with various compounds at optimal concentration for 45 min at 37°C and then exposed to T2 cells alone or loaded with peptide. The optimal dose used in the experiments shown in this paper was determined by using these compounds at different concentrations in the preliminary experiments (data not shown).

We selected donors (healthy donors as well as melanoma patients) who harbor MART-127–35 epitope-specific CTLs in relatively high frequencies and whose CTL precursors could be easily activated (7). Because IL-2 has been implicated in apoptosis in activated T cells (12), we looked for an alternative cytokine support in the in vitro CTL generation protocol. We found that rIL-15 supports the activation and expansion of the epitope-specific CTLs in the mature DC-based epitope presentation system. IL-15 not only supports the primary activation process, but also supports survival of the activated T cells in an Ag-independent manner (13). IL-15Rα is not down-regulated after T cell activation (14) and IL-15 has an important role in T cell memory generation, as such, in T cell survival (15). Therefore, we conducted all our experiments in the presence of IL-15. The MART-127–35 epitope-specific CTLs were readily activated and expanded when they were stimulated by peptide-pulsed matured DCs in the presence of IL-15, in vitro. Fig. 1,A shows an example of the expansion of the MART-127–35 epitope-specific CTLs–derived from a normal healthy individual and a melanoma patient. The expanded MART-127–35 epitope-specific cells also exhibited IFN-γ response in an epitope-specific manner (Fig. 1 B).

FIGURE 1.

An example of activation and expansion of MART-127–35 epitope-specific CTLs in the in vitro CTL generation culture. A, Number of HLA-A2/MART127–35 tetramer-positive cells from a normal donor (top), and from a melanoma patient (bottom). B, IFN-γ response by the corresponding population (M3 = MAGE-3271–279, M1 = MART-127–35).

FIGURE 1.

An example of activation and expansion of MART-127–35 epitope-specific CTLs in the in vitro CTL generation culture. A, Number of HLA-A2/MART127–35 tetramer-positive cells from a normal donor (top), and from a melanoma patient (bottom). B, IFN-γ response by the corresponding population (M3 = MAGE-3271–279, M1 = MART-127–35).

Close modal

The fate of the MART-127–35 epitope-specific CTLs upon secondary encounter of the cognate epitope (i.e., whether following effector function, they survive or die?) was then examined. The CTLs and the peptide-pulsed T2 cells were cocultured in CM without any cytokine and 4 h later the evidence of early death was examined by annexin V staining. As shown in Fig. 2, although the CTLs were fully functional in IFN-γ assay (Fig. 2,A), a large fraction of these activated CTLs became annexin V positive at 4 h (Fig. 2,B). When the cocultures were continued in IL-15 for 5 days and the number of epitope-specific CTLs was counted, only 20–30% of the starting population could be recovered from the coculture of the CTLs with the cognate target. An example for loss of the epitope-specific CTLs following effector function is shown in Fig. 2,C. Thus, ∼50% of the Ag-specific population showed early evidence of death at 4 h (Fig. 2 B) and a much larger fraction eventually died following secondary encounter of the cognate epitope for the very first time.

FIGURE 2.

Effector function and the fate of MART-127–35 epitope-specific CTLs upon secondary encounter of the cognate epitope. A, IFN-γ response by the effector cells. (M3 = MAGE-3271–279, M1 = MART-127–35). B, Evidence of early death in the Ag-specific effector cells after secondary encounter of the epitope. Top panels, Number of MART-127–35 tetramer+ CD8 T cells; bottom panels, MART-127–35 tetramer+/annexin V+ populations. The numbers at the top of the bottom panels represent the mean ± SE of three replicate samples while superscript letters a, b, c, and d indicate that an increase in the MART-127–35 tetramer+/annexin V+ cells in a, when compared with that in b, c, and d, was significant with p < 0. 001 (one-way ANOVA). C, Survival of the MART-127–35-specific population after secondary encounter of the epitope. The effector cells were coincubated with peptide (1 μg/ml) pulsed irradiated T cells (CTL:T2 = 100). The cultures were washed 18 h later and were then maintained in IL-15-containing medium for 5 days. The recovery of the Ag-specific CTLs was determined by counting the number of viable cells (trypan blue-negative population) and by factoring in the number of the Ag-specific population (percent tetramer+ population). The difference in the percent tetramer+ CTLs rescued after 5 days postsecondary stimulation with MART-1 peptide-loaded T2 cells, when compared with other groups, was significant at ∗, p ≤ 0.001 (one-way ANOVA). A and B represent one of five separate experiments. C represents one of three separate experiments.

FIGURE 2.

Effector function and the fate of MART-127–35 epitope-specific CTLs upon secondary encounter of the cognate epitope. A, IFN-γ response by the effector cells. (M3 = MAGE-3271–279, M1 = MART-127–35). B, Evidence of early death in the Ag-specific effector cells after secondary encounter of the epitope. Top panels, Number of MART-127–35 tetramer+ CD8 T cells; bottom panels, MART-127–35 tetramer+/annexin V+ populations. The numbers at the top of the bottom panels represent the mean ± SE of three replicate samples while superscript letters a, b, c, and d indicate that an increase in the MART-127–35 tetramer+/annexin V+ cells in a, when compared with that in b, c, and d, was significant with p < 0. 001 (one-way ANOVA). C, Survival of the MART-127–35-specific population after secondary encounter of the epitope. The effector cells were coincubated with peptide (1 μg/ml) pulsed irradiated T cells (CTL:T2 = 100). The cultures were washed 18 h later and were then maintained in IL-15-containing medium for 5 days. The recovery of the Ag-specific CTLs was determined by counting the number of viable cells (trypan blue-negative population) and by factoring in the number of the Ag-specific population (percent tetramer+ population). The difference in the percent tetramer+ CTLs rescued after 5 days postsecondary stimulation with MART-1 peptide-loaded T2 cells, when compared with other groups, was significant at ∗, p ≤ 0.001 (one-way ANOVA). A and B represent one of five separate experiments. C represents one of three separate experiments.

Close modal

We then examined the mechanism of AICD on these CTLs and also examined whether these CTLs could be rescued from AICD by interfering with the death signaling pathway(s). We examined the effect of interfering with the external death signal receptors (such as Fas, TNFR, TRAIL, etc.) on AICD. We also examined whether any of the MAPK inhibitors could block the AICD in these CTLs because MAPKs have been implicated in the negative selection of developing thymocytes (16, 17). As shown in Fig. 3,A, the death of these CTLs was not caspase dependent, as the pan caspase inhibitor, z-VAD-fmk did not prevent apoptosis. The apoptosis was not also affected by the blockade of the common extrinsic death signal receptors such as Fas, TNFR, or TRAIL. However, we found that while the p38 and ERK inhibitors had no effect, the JNK inhibitor, SP600125, rescued a significant fraction of the CTLs from AICD. Interestingly, SP600125 also inhibited the IFN-γ response by these CTLs (Fig. 3,B) but did not affect their cytotoxic function (Fig. 3 C) suggesting that the cytolytic machinery and the IFN-γ response pathways are differently regulated in these CTLs. Induction of AICD upon secondary exposure to the cognate epitope in the primary CTLs and the protective effect of SP600125 from AICD were observed in CTLs generated from two normal donors and two melanoma patients (collective data not shown).

FIGURE 3.

Effect of external death receptor blockade and MAPK inhibitors on AICD in the epitope-specific CTLs upon secondary encounter of the cognate epitope. The CTLs were preincubated at optimal concentration for 45 min at 37°C with pan caspase inhibitor -z-VAD-fmk (100 μM); human Fas/Fc chimera, human TNF- RI/Fc chimera, human TRAIL-RI/Fc chimera, human TRAIL-RII/Fc chimera, and human IFN-γ RI/Fc chimeric proteins (10 μg/ml); p38 kinase inhibitor SB203580, JNK inhibitor SP600125 JNK, ERK inhibitor PD98059 (25 μM). Then the pretreated as well as untreated CTLs were incubated with T2 cells either alone or loaded with peptide. Four hours after secondary exposure, cells were stained for determining A, the number of tetramer+ cells of the CD8 T cells (top panels) and the number of tetramer+/annexin V+ and tetramer+/annexin populations (bottom panels). The numbers on the top margin of the bottom panels represent tetramer+/annexin from the mean ± SE of three replicate samples while superscript letters a, b, c, and d indicate that the increase in the number of tetramer+/annexin V population in a, when compared with that in b, c, and d, was significant with p < 0.001 (one-way ANOVA). B, Effect of the MAPK inhibitors on IFN-γ response by the effector cells. (M3 = MAGE-3271–279, M1 = MART-127–35). The reduction in IFN-γ synthesis in a, when compared with b, c, and d, was significant at p ≤ 0.001 by one-way ANOVA. M3 = MAGE-3271–279, M1 = MART-127–35. C, Effect of the MAPK inhibitor on cytotoxic response by the effector cells. (M3 = MAGE-3271–279, M1 = MART-127–35). The difference in the percent-specific lysis in the MART-1 peptide-loaded T2 cells was significant (∗, p ≤ 0.001 by Student’s t test) only when compared with that of MAGE-3271–279 peptide-loaded T2 cells at all E:T ratios (E:T). A represents one of four separate experiments while B and C represent one of two separate experiments.

FIGURE 3.

Effect of external death receptor blockade and MAPK inhibitors on AICD in the epitope-specific CTLs upon secondary encounter of the cognate epitope. The CTLs were preincubated at optimal concentration for 45 min at 37°C with pan caspase inhibitor -z-VAD-fmk (100 μM); human Fas/Fc chimera, human TNF- RI/Fc chimera, human TRAIL-RI/Fc chimera, human TRAIL-RII/Fc chimera, and human IFN-γ RI/Fc chimeric proteins (10 μg/ml); p38 kinase inhibitor SB203580, JNK inhibitor SP600125 JNK, ERK inhibitor PD98059 (25 μM). Then the pretreated as well as untreated CTLs were incubated with T2 cells either alone or loaded with peptide. Four hours after secondary exposure, cells were stained for determining A, the number of tetramer+ cells of the CD8 T cells (top panels) and the number of tetramer+/annexin V+ and tetramer+/annexin populations (bottom panels). The numbers on the top margin of the bottom panels represent tetramer+/annexin from the mean ± SE of three replicate samples while superscript letters a, b, c, and d indicate that the increase in the number of tetramer+/annexin V population in a, when compared with that in b, c, and d, was significant with p < 0.001 (one-way ANOVA). B, Effect of the MAPK inhibitors on IFN-γ response by the effector cells. (M3 = MAGE-3271–279, M1 = MART-127–35). The reduction in IFN-γ synthesis in a, when compared with b, c, and d, was significant at p ≤ 0.001 by one-way ANOVA. M3 = MAGE-3271–279, M1 = MART-127–35. C, Effect of the MAPK inhibitor on cytotoxic response by the effector cells. (M3 = MAGE-3271–279, M1 = MART-127–35). The difference in the percent-specific lysis in the MART-1 peptide-loaded T2 cells was significant (∗, p ≤ 0.001 by Student’s t test) only when compared with that of MAGE-3271–279 peptide-loaded T2 cells at all E:T ratios (E:T). A represents one of four separate experiments while B and C represent one of two separate experiments.

Close modal

The caspase-independent death in these primary CTLs upon TCR engagement was confirmed as poly(ADP-ribose) polymerase (PARP) was found to be uncleaved (Fig. 4,A). Of note, PARP was cleaved in staurosporine-treated Jurkat cells and z-VAD-fmk blocked the PARP cleavage. We also confirmed that SP600125 abrogates c-jun activation (Fig. 4 B).

FIGURE 4.

Evidence for a caspase-independent pathway for apoptosis in MART-1-specific primary CTLs. A, The CTLs were preincubated at optimal concentration for 45 min at 37°C with p38 kinase inhibitor SB203580 and JNK inhibitor SP600125 (25 μM). The pretreated as well as untreated CTLs were then incubated with T2 cells either alone or loaded with peptide. Six hours after secondary exposure cells were used for Western blot analysis for PARP cleavage (top panel). Jurkat T cells either preincubated with pan caspase inhibitor -z-VAD-fmk (100 μM) or untreated were exposed with staurosporine (1 μM) to establish the PARP cleavage alongside. B, Jurkat T cells were stimulated with PMA (10 ng/ml)/ionomycin (0.5 μM) for the indicated time both in the presence and absence of JNK inhibitor SP600125 JNK (25 μM). Immunoprecipitation was done to ascertain the blockade of c-jun phosphorylation/activation in the presence of SP600125, as seen in the blot.

FIGURE 4.

Evidence for a caspase-independent pathway for apoptosis in MART-1-specific primary CTLs. A, The CTLs were preincubated at optimal concentration for 45 min at 37°C with p38 kinase inhibitor SB203580 and JNK inhibitor SP600125 (25 μM). The pretreated as well as untreated CTLs were then incubated with T2 cells either alone or loaded with peptide. Six hours after secondary exposure cells were used for Western blot analysis for PARP cleavage (top panel). Jurkat T cells either preincubated with pan caspase inhibitor -z-VAD-fmk (100 μM) or untreated were exposed with staurosporine (1 μM) to establish the PARP cleavage alongside. B, Jurkat T cells were stimulated with PMA (10 ng/ml)/ionomycin (0.5 μM) for the indicated time both in the presence and absence of JNK inhibitor SP600125 JNK (25 μM). Immunoprecipitation was done to ascertain the blockade of c-jun phosphorylation/activation in the presence of SP600125, as seen in the blot.

Close modal

We examined the status of the mitochondrial membrane potential in the CTLs during AICD. As shown in Fig. 5 A, the CTLs surprisingly exhibited hyperpolarization of the mitochondrial transmembrane potential (Δψm) (18, 19) while the apoptotic Jurkat cells exhibited hypopolarization of the mitochondrial membrane. Hyperpolarization has been associated with the apoptotic process in peripheral blood lymphocytes and has also been shown to occur independently from activation of caspases (20, 21). Further, hyperpolarization of mitochondrial membrane potential has been shown to be a reversible stage in the mitochondrial death decision process while hypopolarization usually represents a point of irreversible commitment to death (18, 19). Interestingly, SP600125 (JNK inhibitor) as well as SB203580 (p38 inhibitor) decreased the level of hyperpolarization induced by the restimulation with the cognate peptide (data not shown) suggesting that SP600125-mediated rescue is not specifically mediated through the modulation of mitochondrial membrane potential.

FIGURE 5.

Effect of Ag-specific secondary stimulation on mitochondrial transmembrane potential (Δψm), pro- and antiapoptotic protein, and expression of cell surface molecules. A, Differential effect on membrane polarization in MART-1 primary CTLs (top panels) vs Jurkat T cells (bottom panels). CTLs were incubated with T2 cells either alone or loaded with peptide. After overnight secondary exposure, cells were stained with MART-1 tetramer/CD8 and DiOC6. The histogram represents the fluorescence of DiOC6 in the gated Ag-specific tetramer+ population (top panels). Jurkat T cells were also analyzed for modulation of membrane potential after TCR-driven stimulation (bottom panels). Values represent the mean ± SD of fluorescence intensity triplicates from one of three separate experiments. ∗, p < 0.001 (one-way ANOVA). B, Left panel, Western blots for anti- and proapoptotic proteins using the whole CTL population following secondary stimulation in the presence or absence of the MAPK inhibitors. Right panel, The densitometric analysis of the relevant Bcl family proteins normalized to β-actin controls using OPTIMAS version 5.2 software (Bioscan, Bothell, WA). C, Expression of different cell surface receptors for survival or death signal in the Ag-specific population upon secondary stimulation. Values in the upper left corner represent the geometric mean of the fluorescence intensity for that particular marker when stimulated with T2/MAGE-3271–279 while those on the upper right corner represent the geometric mean of the fluorescence intensity when stimulated with T2/MART-127–35.

FIGURE 5.

Effect of Ag-specific secondary stimulation on mitochondrial transmembrane potential (Δψm), pro- and antiapoptotic protein, and expression of cell surface molecules. A, Differential effect on membrane polarization in MART-1 primary CTLs (top panels) vs Jurkat T cells (bottom panels). CTLs were incubated with T2 cells either alone or loaded with peptide. After overnight secondary exposure, cells were stained with MART-1 tetramer/CD8 and DiOC6. The histogram represents the fluorescence of DiOC6 in the gated Ag-specific tetramer+ population (top panels). Jurkat T cells were also analyzed for modulation of membrane potential after TCR-driven stimulation (bottom panels). Values represent the mean ± SD of fluorescence intensity triplicates from one of three separate experiments. ∗, p < 0.001 (one-way ANOVA). B, Left panel, Western blots for anti- and proapoptotic proteins using the whole CTL population following secondary stimulation in the presence or absence of the MAPK inhibitors. Right panel, The densitometric analysis of the relevant Bcl family proteins normalized to β-actin controls using OPTIMAS version 5.2 software (Bioscan, Bothell, WA). C, Expression of different cell surface receptors for survival or death signal in the Ag-specific population upon secondary stimulation. Values in the upper left corner represent the geometric mean of the fluorescence intensity for that particular marker when stimulated with T2/MAGE-3271–279 while those on the upper right corner represent the geometric mean of the fluorescence intensity when stimulated with T2/MART-127–35.

Close modal

The status of Bcl family pro- and antiapoptotic proteins and costimulation through certain TCRs (CD28, 4-1BB, OX-40, etc.) influence the survival of activated T cells (22). As such, we examined the expression levels of the anti- and proapoptotic Bcl family proteins and these receptors in the CTLs in a condition that induces AICD. As shown in Fig. 5,B, while Bcl-2 and Bcl-xL were nonselectively up-regulated by SB203580 and SP600125, only phosphorylated Bcl-2 and Mcl-1 were selectively up-regulated by the JNK inhibitor SP600125. Bim–a pro-apoptotic protein–was markedly up-regulated (or released from sequestered sites) by the CTLs undergoing AICD. Of interest, the CTLs up-regulated 4-1BB (10- to 20-fold increase of the geometric mean intensity of fluorescence) and CD25 (3- to 6-fold increase of the geometric mean intensity of fluorescence) upon secondary encounter of the Ag (Fig. 5 C).

Finally, we examined the functional status of SP600125-rescued CTLs upon continuous culture. After overnight exposure of the CTLs to the cognate epitope in the presence or absence of SP600125, the CTLs were washed and then maintained in continuous culture in IL-15 containing CM without the inhibitor. Five days after, the number of viable MART-127–35 tetramer-positive cells were determined and their function was tested in the IFN-γ response assay. Fig. 6,A shows that only a small fraction of the starting population could be recovered from the cognate epitope-exposed culture in the absence of the JNK inhibitor. A larger fraction of the starting epitope-specific population was recovered from the coculture that was started in the presence of the JNK inhibitor, SP600125. Remarkably, the rescued CTLs regained their capacity to synthesize IFN-γ (Fig. 6 B).

FIGURE 6.

Survival of the Ag-specific CTLs following secondary encounter of the epitope in the presence of SP600125 and recovery of function in the SP600125-rescued CTLs. Effector cells were cocultured with peptide-pulsed T2 cells in the presence or absence of the kinase inhibitors (25 μM). The cultures were washed 18 h later and then recultured in IL-15-containing medium. On day 5, the numbers of the viable Ag-specific CTLs were determined. A, Top panels, The forward and side scattering of the total population on day 5 and the number of viable cells that could be recovered and gated for analysis. The number (11%) of MART-127–35 tetramer+/CD8+ population exposed to MART-127–35 peptide-pulsed T2 cells (T2/M1) is a reflection of loss of epitope-specific CTLs from AICD. Bottom panels, The percentage of MART-127–35 tetramer+/CD8+ cells recovered 5 days after coculture of the CTLs with peptide-pulsed T2 cells in the presence or absence of SP600125. B, IFN-γ response by the recovered populations (M3 = MAGE-3271–279, M1 = MART-127–35).

FIGURE 6.

Survival of the Ag-specific CTLs following secondary encounter of the epitope in the presence of SP600125 and recovery of function in the SP600125-rescued CTLs. Effector cells were cocultured with peptide-pulsed T2 cells in the presence or absence of the kinase inhibitors (25 μM). The cultures were washed 18 h later and then recultured in IL-15-containing medium. On day 5, the numbers of the viable Ag-specific CTLs were determined. A, Top panels, The forward and side scattering of the total population on day 5 and the number of viable cells that could be recovered and gated for analysis. The number (11%) of MART-127–35 tetramer+/CD8+ population exposed to MART-127–35 peptide-pulsed T2 cells (T2/M1) is a reflection of loss of epitope-specific CTLs from AICD. Bottom panels, The percentage of MART-127–35 tetramer+/CD8+ cells recovered 5 days after coculture of the CTLs with peptide-pulsed T2 cells in the presence or absence of SP600125. B, IFN-γ response by the recovered populations (M3 = MAGE-3271–279, M1 = MART-127–35).

Close modal

Ashwell et al. (1) first noted growth arrest in T cell hybridoma upon TCR-mediated stimulation. Shi et al. (2) thereafter showed that TCR- driven signaling leads to death of the cells and coined the term AICD. In physiology, T cell responses follow three phases: amplification, contraction, and memory generation. During the contraction phase of a primary T cell response, the majority of the Ag-specific T cells undergo apoptotic death (23) as a form of PCD primarily to maintain homeostasis (24). It has been thought for some time that the apoptotic signal is initiated through the Fas and TNF receptors and that the cell death is mediated by the eventual activation of an executioner caspase (25, 26). A universal role of the Fas and TNF family receptor-driven signaling for T cell apoptosis has, however, been controversial (27) and both caspase-dependent and caspase-independent deaths in T cells have been described (28). Further, it is now increasingly apparent that an intrinsic cell death pathway triggers AICD in CTLs without involving the extrinsic death receptors such as Fas and/or TNF family receptors (29, 30). Although further studies will be needed to establish the mechanism underlying apoptosis in the epitope-specific primary CTLs in our system, our data lend support to the latter view.

The results are also noteworthy for a number of other reasons. First, the data show that a large fraction of the epitope-specific primary CTLs generated, in vitro, are indeed susceptible to AICD from their very first re-encounter of the Ag even during the amplification phase (Fig. 2). It should be mentioned that the MART-127–35 epitope-specific populations expanded more than 1000-fold in IL-15 without a second round of stimulation and then senesced around day 50. These CTLs, interestingly, exhibited propensity to undergo AICD as early as on day 9 (collective data not shown). This, therefore, suggests that the apoptotic program in this in vitro CTL activation system is turned on quite early and lends support to the recent observations that the apoptotic program in viral Ag-specific CTLs gets activated substantially before the contraction phase, in vivo (31). Of interest, stimulation of the CTLs with the cognate peptide that induced apoptosis paradoxically up-regulated the expressions of IL-2Rα (CD25) and 4-1BB (CD137) substantially (Fig. 5,C). Although these molecules are associated with T cell growth and survival, 4-1BB has lately been implicated in apoptosis and has also been shown to interfere with IFN-γ response but not with cytotoxicity in CTLs (32, 33). A connection between 4-1BB-mediated signaling and activation of the JNK pathway has also been suggested (34). Second, the MART-127–35 epitope-specific CTLs generated in the in vitro DC-based epitope presentation protocol in the presence of IL-15 and maintained in IL-15 were just as susceptible to AICD as the CTLs that were generated and maintained in IL-2 (collective data not shown). Third, the results clearly reveal that SP600125 is capable of protecting these CTLs from AICD (Fig. 3). Because SP600125 functions as an inhibitor of the JNK, the data suggest a role for JNK in the apoptotic process. Fourth, while SP600125 negatively regulates the IFN-γ responsiveness of the CTLs, it does not affect their cytotoxic function, suggesting that the two functional pathways in CTLs are differently regulated. Finally, and most importantly, the rescued CTLs regained IFN-γ responsiveness in continued culture after the inhibitor being washed off.

The JNK is a member of the MAPK family of signaling proteins. JNK has been associated with a variety of biological processes–most notably in inflammation and transformation (35). All three MAPKs (i.e., p38, JNK, and ERK) have been found to have different roles in T cell biology such as in proliferation, effector function, survival as well as death (16, 17, 36, 37, 38, 39, 40). Different isoforms of JNK have been found to have divergent roles in CD4+ and CD8+ T cells (17). For example, JNK1 is needed for the expression of IL-2Rα (CD25) upon activation and for proliferation. As such, jnk-1-null mice exhibit marked reduction in the expansion of Ag-driven CD8+ T cells (39). JNK2 down-regulates IL-2 production in CD8+ T cells (39, 40). JNK has also been implicated in negative selection of thymocytes (17, 40). Both JNK1 and JNK2 have been found to have stage-dependent roles in T cell development. For example, while JNK1 has been associated with anti-CD3-induced apoptosis of double-positive thymocytes, jnk2-null double-positive thymocytes are resistant to anti-CD3-induced apoptosis although they are sensitive to apoptosis induced by dexamethasone, anti-Fas Ab, or UV radiation (38, 39). Thus, immature thymocytes seem to need one form of JNK or another for undergoing receptor-driven apoptosis but mature T cells need JNK1 for proliferation and IL-2 synthesis, and they do not seem to need JNK2 for activation-induced apoptosis (39, 40). Although these observations suggest a role for JNK in apoptosis in immature thymocytes but not in mature T cells, the results of our study reveal that JNK may be an important player in apoptotic death in CTLs in the periphery. A careful examination of this type of AICD in CTLs with specificity for a “self” vs a “dangerous” Ag in the same donor will be informative.

JNK is activated by dual phosphorylation of the Thr and Tyr residues within the Thr-Pro-Tyr motif and it can be activated by cytokines and environmental stress. Its activation leads to the up-regulation of c-jun and eventual phosphorylation of transcription factors such as AP-1 and other proteins some of which are associated with apoptosis. Given that death in CD8+ T cells is turning out to be mostly driven by the internal pathway of apoptosis and results from mitochondrial dysfunction, the release of reactive oxygen intermediates, and stress (29, 30), and as JNK can play a role in stress-induced activation of the cytochrome c-mediated death pathway (41), a role for JNK in AICD in Ag-specific CD8+ CTLs can be envisioned.

As mentioned earlier, Bcl family pro- and antiapoptotic proteins play an important role in the survival or death of cells (22). Our observation on the modulation of some of these proteins, therefore, is of interest. The JNK inhibitor, SP600125, increased the level of pBcl-2 and Mcl-1. The effect of phosphorylated Bcl-2 on apoptosis of T cells remains an unsettled issue (42, 43). However, Mcl-1 is an important Bcl family member with a positive effect on T cell survival (44). It is therefore tempting to suggest that the JNK inhibitor SP600125 prevents AICD in these CTLs by up-regulating Mcl-1. Admittedly, more work will be needed to establish this issue.

A role for JNK in AICD in CTLs in an Ag-specific manner in our study has been based from the observation of the effect of SP600125 on AICD in these cells (Fig. 3). SP600125 has been extensively used as a JNK inhibitor in numerous studies although it is not absolutely specific for JNK (35). The possibility exists that the rescuing effect of SP600125 could have resulted from its effect on some other molecule(s). More work will be needed to firmly establish a role for JNK in AICD in Ag-specific CTLs and to “connect” JNK with the upstream and downstream events in the apoptotic processes in primary CTLs. Meanwhile, considering that the antitumor effects of the CTLs–induced by active vaccination or adoptively transferred–can be vastly enhanced by preventing their “premature” death and as JNK inhibitors have generated much interest in inflammatory diseases and have also entered into clinical trials (35), our observations have implications in active as well as adoptive immunotherapeutic strategies for cancer and in further studies of the regulation of survival and death in CTLs.

We thankfully acknowledge Drs. Leo Lefrancois for his careful reading of the manuscript, Meenal Mehrotra for help in some of the initial experiments, and S. K. Suneja for help with densitometric analysis.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1

The work was supported by Public Health Service Grants CA 61398, CA 83130, and CA 88059.

3

Abbreviations used in this paper: PCD, programmed cell death; AICD, activation-induced cell death; TAA, tumor-associated Ag; DC, dendritic cell; CM, complete media; PARP, poly(ADP-ribose) polymerase; Δψm, mitochondrial transmembrane potential.

1
Ashwell, J. D., R. E. Cunningham, P. D. Noguchi, D. Hernandez.
1987
. Cell growth cycle block of T cell hybridomas upon activation with antigen.
J. Exp. Med.
165
:
173
.
2
Shi, Y. F., B. Sahai, D. R. Green.
1989
. Cyclosporin A inhibits activation-induced cell death in T-cell hybridomas and thymocytes.
Nature
339
:
625
.
3
Budd, R. C..
2001
. Activation-induced cell death.
Curr. Opin. Immunol.
13
:
356
.
4
Houghton, A. N..
1994
. Cancer antigens: immune recognition of self and altered self.
J. Exp. Med.
180
:
1
.
5
Parmiani, G., C. Castelli, P. Dalerba, R. Mortarini, L. Rivoltini, F. M. Marincola, A. Anichini.
2002
. Cancer immunotherapy with peptide based vaccines: what have we achieved: where are we going?.
J. Natl. Cancer Inst.
94
:
805
.
6
Mukherji, B., N. G. Chakraborty, S. Yamasaki, T. Okino, H. Yamase, J. R. Sporn, S. K. Kurtzman, M. T. Ergin, J. Ozols, J. Meehan.
1995
. Induction of antigen-specific cytolytic T cells in situ in human melanoma by immunization with synthetic peptide-pulsed autologous antigen presenting cells.
Proc. Natl. Acad. Sci. USA
92
:
8078
.
7
Mehrotra, S., R. Stevens, R. Zengou, N. G. Chakraborty, L. H. Butterfield, J. S. Economou, D. I. Dorsky, B. Mukherji.
2003
. Regulation of melanoma epitope-specific cytolytic T lymphocyte response by immature and activated dendritic cells, in vitro.
Cancer Res.
63
:
5607
.
8
Coulie, P. G., V. Brichard, A. Van Pel, T. Wolfel, J. Schneider, C. Traversari, S. Mattei, E. De Plaen, C. Lurquin, J. P. Szikora, et al
1994
. A new gene coding for a differentiation antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas.
J. Exp. Med.
180
:
35
.
9
Kawakami, Y., S. Eliyahu, K. Sakaguchi, P. F. Robbins, L. Rivoltini, J. R. Yannelli, E. Appella, S. A. Rosenberg.
1994
. Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes.
J. Exp. Med.
180
:
347
.
10
Chakraborty, N. G., L. Li, J. R. Sporn, S. H. Kurtzman, M. T. Ergin, B. Mukherji.
1999
. Emergence of regulatory CD4+ T cell response to repetitive stimulation with antigen-presenting cells in vitro: implications in designing antigen-presenting cell-based tumor vaccines.
J. Immunol.
162
:
5576
.
11
Mukherji, B., A. Guha, N. G. Chakraborty, M. Sivanandham, A. L. Nashed, J. R. Sporn, M. T. Ergin.
1989
. Clonal analysis of cytotoxic and regulatory T cell responses against human melanoma.
J. Exp. Med.
169
:
1961
.
12
Lenardo, M. J..
1991
. Interleukin-2 programs mouse αβ T lymphocytes for apoptosis.
Nature
353
:
858
.
13
Waldmann, T. A., S. Dubois, Y. Tagaya.
2001
. Contrasting roles of IL-2 and IL-15 in the life and death of lymphocytes: implications for immunotherapy.
Immunity
14
:
105
.
14
Schluns, K. S., L. Lefrancois.
2003
. Cytokine control of memory T-cell development and survival.
Nat. Rev. Immunol.
3
:
269
.
15
Zhang, X., S. Sun, I. Hwang, D. F. Tough, J. Sprent.
1998
. Potent and selective stimulation of memory-phenotype CD8+ T cells in vivo by IL-15.
Immunity
8
:
591
.
16
Dong, C., R. J. Davis, R. A. Flavell.
2002
. MAP kinases in the immune response.
Annu. Rev. Immunol.
20
:
55
.
17
Rincon, M., G. Pedraza-Alva.
2003
. JNK and p38 MAP kinases in CD4+ and CD 8+ T cells.
Immunol. Rev.
192
:
131
.
18
Green, D. R., J. C. Reed.
1998
. Mitochondria and apoptosis.
Science
281
:
1309
.
19
Perl, A., P. Gergely, Jr, G. Nagy, A. Koncz, K. Banki.
2004
. Mitochondrial hyperpolarization: a checkpoint of T-cell life, death and autoimmunity.
Trends Immunol.
25
:
360
.
20
Nagy, G., A. Koncz, A. Perl.
2003
. T cell activation induced mitochondrial hyperpolarization is mediated by Ca2+ and redox dependent production of nitric oxide.
J. Immunol.
171
:
5188
.
21
Banki, K., E. Hutter, N. J. Gonchoroff, A. Perl.
1999
. Elevation of mitochondrial transmembrane potential and reactive oxygen intermediate levels are early events and occur independently from activation of caspases in Fas signaling.
J. Immunol.
162
:
1466
.
22
Adams, J. A., S. Cory.
1998
. The Bcl-2 protein family: arbiters of cell survival.
Science
281
:
1322
.
23
Murali-Krishna, K., J. D. Altman, M. Suresh, D. J. Sourdive, A. J. Zajac, J. D. Miller, J. Slansky, R. Ahmed.
1998
. Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection.
Immunity
8
:
177
.
24
van Parjis, L., A. K. Abbas.
1998
. Homeostasis and self-tolerance: turning lymphocytes off.
Science
280
:
243
.
25
Dhein, J., H. Walczak, C. Baumler, K. Debatin, P. Krammer.
1995
. Autocrine T-cell suicide mediated by Apo-1 (Fas/CD95).
Nature
373
:
438
.
26
Zheng, L., G. Fisher, R. E. Miller, J. Peschon, D. H. Lynch, M. J. Lenardo.
1995
. Induction of apoptosis in mature T cells by tumour necrosis factor.
Nature
377
:
348
.
27
Hildeman, D. A., Y. Zhu, T. C. Mitchell, J. Kappler, P. Marrack.
2002
. Molecular mechanisms of activated T cell death in vivo.
Curr. Opin. Immunol.
14
:
354
.
28
Jaattela, M., J. Tschopp.
2003
. Caspase-independent cell death in T lymphocytes.
Nat. Immunol.
4
:
416
.
29
Hildeman, D. A., T. Mitchell, T. K. Teague, P. Henson, B. J. Day, J. Kappler, P. C. Marrack.
1999
. Reactive oxygen species regulate activation-induced T cell apoptosis.
Immunity
10
:
735
.
30
Grayson, J. M., N. G. Laniewski, J. G. Lanier, R. Ahmed.
2003
. Mitochondrial potential and reactive oxygen intermediates in antigen-specific CD 8+ T cells during viral infection.
J. Immunol.
170
:
4745
.
31
Badovinac, V. P., B. B. Poter, J. T. Harty.
2003
. Programmed contraction of CD8+ T cells after infection.
Nat. Immunol.
3
:
619
.
32
Michel, J., S. Pauly, J. Langstein, P. H. Krammer, H. Schwarz.
1999
. CD137-induced apoptosis is independent of CD95.
Immunology
98
:
42
.
33
Cooper, D., P. Bansal-Pakala, M. Croft.
2002
. 4-1BB (CD137) controls the clonal expansion and survival of CD8 T cells in vivo but does not contribute to the development of cytotoxicity.
Eur. J. Immunol.
32
:
521
.
34
Cannons, J. L., K. P. Hoeflich, J. R. Woodgett, T. H. Watts.
1999
. Role of the stress kinase pathway in signaling via the T cell costimulatory receptor 4-1BB.
J. Immunol.
163
:
2990
.
35
Manning, A. M., R. J. Davis.
2003
. Targeting JNK for therapeutic benefit: from junk to gold?.
Nat. Rev. Drug Discov.
2
:
554
.
36
Zhu, L., X. Yu, Y. Akatsuka, J. A. Cooper, C. Anasetti.
1999
. Role of mitogen-activated protein kinases in activation-induced apoptosis of T cells.
Immunology
97
:
26
.
37
Merritt, C., H. Enslen, N. Diehl, D. Conze, R. J. Davis, M. Rincon.
2000
. Activation of p38 mitogen-activated protein kinase in vivo selectively induces apoptosis of CD8+ but not CD4+ T cells.
Mol. Cell. Biol.
20
:
936
.
38
Sabapathy, K., T. Kallunki, J. P. David, I. Fraef, M. Karn, E. F. Wagner.
2001
. c-jun NH2-terminal kinase and JNK2 have similar and stage-dependent roles in regulating T cell apoptosis and proliferation.
J. Exp. Med.
193
:
317
.
39
Conze, D., T. Krahl, N. Kennedy, L. Weiss, J. Lumsden, P. Hess, R. Flavell, G. Le Gros, R. J. Davis, M. Rincon.
2002
. c jun NH2-terminal kinase(JNK)1 and JNK2 have distinct roles in CD8+ T cell activation.
J. Exp. Med.
195
:
811
.
40
Arbour, N., D. Naniche, D. Homann, R. J. Davis, R. A. Flavell, M. B. Oldstone.
2002
. c-Jun NH(2)-terminal kinase (JNK)1 and JNK2 signaling pathways have divergent roles in CD8+ T cell-mediated antiviral immunity.
J. Exp. Med.
195
:
801
.
41
Tournier, C., P. Hess, D. D. Yang, J. Xu, T. K. Turner, A. Nimnual, D. Bar-Sagi, S. N. Jones, R. A. Flavell, R. J. Davis.
2000
. Requirement of JNK for stress-induced activation of the cytochrome c-mediated death pathway.
Science
288
:
870
.
42
Yamamoto, K., H. Ichijo, S. J. Korsmeyer.
1999
. BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-terminal protein kinase pathway normally activated at G2/M.
Mol. Cell. Biol.
19
:
8469
.
43
Blagosklonny, M. V., P. Giannakakou, W. S. el-Deiry, D. G. Kingston, P. I. Higgs, L. Neckers, T. Fojo.
1997
. Raf-1/bcl-2 phosphorylation: a step from microtubule damage to cell death.
Cancer Res.
57
:
130
.
44
Opferman, J. T., A. Letai, C. Beard, M. D. Sorcinelli, C. C. Ong, S. J. Korsmeyer.
2003
. Development and maintenance of B and T lymphocytes requires antiapoptotic MCL-1.
Nature
426
:
671
.