Literature
首页医源资料库在线期刊分子药理学杂志2006年第68卷第10期

Inhibition of Ca2+ Influx Is Required for Mitochondrial Reactive Oxygen Species-Induced Endoplasmic Reticulum Ca2+ Depletion and Cell Death in Leukemia Cells

来源:《分子药理学杂志》
摘要:【关键词】InhibitionDisturbancesofendoplasmicreticulum(ER)Ca2+homeostasisorproteinprocessingcanleadtoERstress-inducedcelldeath。ToinvestigatetheroleofOSinERstress,wemeasuredOSinresponsetothreeERstressagents:econazole(Ec),whichstimulatesERCa2+release......

点击显示 收起

【关键词】  Inhibition

    Disturbances of endoplasmic reticulum (ER) Ca2+ homeostasis or protein processing can lead to ER stress-induced cell death. Increasing evidence suggests that oxidative stress (OS) plays an important role in a variety of cell death mechanisms. To investigate the role of OS in ER stress, we measured OS in response to three ER stress agents: econazole (Ec), which stimulates ER Ca2+ release and blocks Ca2+ influx; thapsigargin (Tg), a sarco(endo)plasmic reticulum Ca2+ ATPase inhibitor that releases ER Ca2+ and stimulates Ca2+ influx; and tunicamycin (Tu), a glycosylation inhibitor that causes protein accumulation in the ER. Ec, but not Tg or Tu, caused a rapid increase in OS. Reactive oxygen species (ROS) generation was observed within mitochondria immediately after exposure to Ec. Furthermore, Ec hyperpolarized the mitochondrial membrane and inhibited adenine nucleotide transport in cell-free mitochondria, suggesting a mitochondrial target. Antimycin A, an inhibitor of complex III in electron transport, reversed mitochondrial hyperpolarization, OS generation, ER Ca2+ depletion, and cell death by Ec, suggesting complex III dependence for these effects. Antioxidants butylated hydroxytoluene and N-Acetyl-L-cysteine prevented ER Ca2+ depletion and cell death by Ec. However, inhibition of Ca2+ influx by Ec was unaffected by either antimycin A or the antioxidants, suggesting that this target is distinct from the mitochondrial target of Ec. Atractyloside, an adenine nucleotide transport inhibitor, generated ROS and stimulated ER Ca2+ release, but it did not block Ca2+ influx, deplete the ER or induce cell death. Taken together, these results demonstrate that combined mitochondrial ROS generation and Ca2+ influx blockade by Ec is required for cell death.The endoplasmic reticulum (ER) is a major intracellular calcium store and the organelle responsible for the synthesis and post-translational modification of proteins destined for secretion or surface expression. These post-translational processes include protein folding, glycosylation, disulfide bond formation, and ER-Golgi protein trafficking. Disturbances of ER calcium homeostasis and protein processing cause accumulation of unfolded or misfolded proteins in the ER lumen and initiate the unfolded protein response (Kaufman, 1999; Ferri and Kroemer, 2001; Patil and Walter, 2001). Cells exhibit a variety of responses in their attempt to mitigate such ER stress. These include increased expression of ER-resident Ca2+-dependent molecular chaperones such as GRP78 (BiP) and GRP94 (Kozutsumi et al., 1988) and suppression of protein synthesis to reduce the unfolded protein load. Preconditioning with sublethal levels of ER stress has been shown to protect cells, in part through up-regulation of chaperones (Liu et al., 1998; Hung et al., 2003). However, sustained ER stress will eventually result in prolonged protein synthesis inhibition that leads to cell death (Soboloff and Berger, 2002; Zhang and Berger, 2004). Important mediators of ER stress-associated death include the cleavage and activation of the ER-associated caspase-12 (Szegezdi et al., 2003) and increased expression of CHOP/GADD153 (Wang et al., 1996), a transcription factor that sensitizes cells to apoptosis.

    Disruption of Ca2+ homeostasis in the ER, such as after treatment with the SERCA inhibitor thapsigargin (Tg) (Thastrup et al., 1990), can stimulate sustained Ca2+ influx from the extracellular milieu. In some cell types, this leads to both cytosolic and mitochondrial Ca2+ overload (Babich et al., 1994; Soboloff and Berger, 2002), triggering apoptosis (Orrenius et al., 2003). Econazole (Ec), an imidazole antifungal, also stimulates depletion of the Tg-sensitive ER calcium store. However unlike Tg, Ec additionally blocks Ca2+ influx, resulting in sustained ER Ca2+ depletion (Franzius et al., 1994; Jan et al., 1999; Soboloff and Berger, 2002). In previous studies, we have shown that the Ca2+ depletion caused by Ec induces activation enhanced cell death in leukemic cells, breast cancer cells, and murine bone marrow-derived mast cells through sustained inhibition of protein synthesis (Gommerman and Berger, 1998; Soboloff and Berger, 2002; Soboloff et al., 2002; Zhang et al., 2002; Zhang and Berger, 2004). However, the mechanism of how this compound affects the intracellular ER store and the subsequent fate of the cell remains unknown.

    Increasing evidence suggests that reactive oxygen species (ROS) and the oxidation-reduction (redox) state play important roles in a variety of cell death mechanisms induced by widely used antitumor drugs or by environmental toxic substances (Orrenius, 1985; Orrenius and Nicotera, 1987; Slater et al., 1995) (Feinendegen, 2002; Ueda et al., 2002). Because oxidative damage to the ER has been implicated in some forms of cell death (Hayashi et al., 2003; Lai et al., 2003; Watanabe et al., 2003), we investigated the possible role of oxidative stress (OS) in the induction of ER stress by different agents. Here, we show that ER Ca2+ depletion and cell death induced by Ec but not Tg or tunicamycin (Tu) is dependent on ROS production at the mitochondria, thus identifying a mediating role for ROS in communicating mitochondrial disruption to the ER.

    Cell Culture, Cell Death, and Caspase Assays. Human promyelocytic leukemia HL-60 cells were grown in RPMI 1640 medium containing 10% heat-inactivated fetal bovine serum (FBS) and 2.5 mM L-glutamine. HL-60 cells were treated with 15 µM Ec in the presence or absence of 125 nM AA, 5 µM BHT, or 1 mM Nac at 37°C for 2 h, in medium containing 1% FBS. Drugs were washed out, and the cells were recovered in 10% FBS drug-free RPMI 1640 medium for 6 h. Samples were costained with Annexin V-Cy5 (BioVision, Mountain View, CA) and propidium iodide (PI), and cell death was determined by flow cytometry. Early (Annexin V-positive, PI-negative) and late (Annexin V-positive, PI-positive) events were scored as dead cells. Caspase-3/7 activation was measured using the Vybrant 5-carboxyfluorescein caspase assay kit from Invitrogen (Carlsbad, CA) as described by the manufacturer.

    Detection of Oxidative Stress and Mitochondrial ROS. To examine the generation of OS, HL-60 cells were incubated with the indicated concentrations of Ec, Tu, or Tg in RPMI 1640 containing 2% FBS at 37°C for 2 h and then loaded with the OS indicator 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate (8 µM CM-H2DCFDA; Invitrogen, Carlsbad, CA) at 37°C for 30 min. Fluorescence was measured by flow cytometry using excitation at 488 nm and emission at 530 nm at indicated intervals after treatment. Mitochondrial-specific ROS generation was measured using the MitoSox Red fluorescent dye (Invitrogen) as described by the manufacturer. This dye accumulates in the mitochondria, is oxidized by superoxide, and emits at 580 nm.

    Measurement of Cytosolic Ca2+ Concentration. Cytosolic Ca2+ measurements were performed by flow cytometry. HL-60 cells were serum-deprived for 2 h in Tyrode's buffer (10 mM HEPES, 100 mM NaCl, 5 mM KCl, 1.4 mM CaCl2, 1 mM MgCl2, 5.6 mM glucose, and 0.05% bovine serum albumin (0.05%) containing Ec, atractyloside (Atra), antioxidants, and/or hydrogen peroxide where indicated. Cells were then incubated in loading buffer (5 µM indo-1AM and 0.03% Pluronic F-127 in Tyrode's buffer; both from Invitrogen) at 37°C for 30 min and washed and incubated (15 min at room temperature) to allow for the complete conversion of indo-1AM to Ca2+-sensitive indo-1 through hydrolysis. Measurements were performed using a laser tuned to 338 nm while monitoring emissions at 405 and 450 nm. FloJo software (Treestar Software, Ashland, OR) was used to analyze the flow cytometric data. The concentration of intracellular free Ca2+ was calculated according to the following formula (Grynkiewcz et al., 1985): [Ca2+]i = Kd x (Fmin/Fmax) x (R - Rmin)/(Rmax - R), where R is the ratio of the fluorescence intensities measured at 405 and 450 nm during the experiments, and F is the fluorescence intensity measured at 450 nm. Rmin, Rmax, Fmin, and Fmax were determined from in situ calibration of viable cells using 4 µM ionomycin in the absence (Rmin and Fmin; 10 mM EGTA) and presence of (Rmax and Fmax) of Ca2+. Kd (250 nM) is the dissociation constant for indo-1 at 37°C. Rmin, Rmax, Fmin, and Fmax varied depending upon settings and were determined at the beginning of each experimental procedure and experimental condition.

    Potentiometric Measurement of Mitochondria Membrane Potential. MMP measurements were performed by flow cytometry. Cells (5 x 105 cells/ml) were growth factor-deprived for 2hin Tyrode's buffer and then incubated with 5 µg/ml JC-1 (Invitrogen) for 15 min at room temperature in Tyrode's buffer. Then, cells were washed (three times) to remove extracellular JC-1. Measurements were performed using a laser tuned to 488 nm while monitoring the emissions of JC-1 monomers at 530 nm and JC-1 aggregates at 585 nm. MMP measurements were normalized using a modification of a formula from Rottenberg and Wu (1998): MMP = (R - RFccp)/(R0 - RFccp) x 100, where R is the ratio of the fluorescence intensities measured at 530 and 585 nm during the experiments, R0 is the fluorescence ratio of untreated cells, and RFccp is the fluorescence intensity measured after the addition of 2 µM carbonyl cyanide p-(trifluomethoxy) phenylhydrazone (FCCP; Sigma-Aldrich, St. Louis, MO), a procedure that collapses the mitochondrial membrane potential and was performed at the beginning of each experiment.

    Adenosine Nucleotide Transfer-Dependent ADP Import Assay. Adenosine nucleotide transfer (ANT) across the mitochondrial membrane in ER-stressed cells was measured as described by Vander Heiden et al. (1999). In brief, 5 x 107 HL-60 cells were disrupted by mechanical lysis through homogenization in a mitochondria isolation buffer (200 mM mannitol, 70 mM sucrose, 10 mM HEPES, pH 7.4, and 1 mM EGTA). After centrifugation of the supernatant (750g for 10 min to remove debris followed by 10,000g for 10 min), mitochondrial pellets were resuspended in ADP import buffer (250 mM sucrose, 20 mM HEPES, pH 7.2, 10 mM KCl, 5 mM succinate, 3 mM KH2PO4, 1.5 mM MgCl2, 1 mM EGTA, and 5 µM rotenone) with or without the adenine nucleotide transporter inhibitor atractyloside (50 µM; Sigma-Aldrich). [14C]ADP (1 µCi; PerkinElmer Life and Analytical Sciences, Boston, MA) was added to the mitochondrial suspension and incubated for 10 min on ice. After washing two times in ADP import buffer, the samples were resuspended in scintillant (PerkinElmer) and quantified using a beta counter (PerkinElmer Wallac, Gaithersburg, MD). ANT-dependent ADP transport activity was determined by calculating the difference in counts between samples that were or were not preincubated with atractyloside.

    ATP Bioluminescence Assay. ATP levels were measured by using ATP bioluminescence assay kit CLS II (Roche Diagnostics, Indianapolis, IN). In brief, cells (5 x 105 cells/ml) were treated with 15 µM Ec, 125 nM antimycin A, or both at 37°C for 2 or6hin RPMI 1640 medium with 0.05% bovine serum albumin. After washing in chilled PBS, cells were resuspended in 100 µl of dilution buffer, added to 900 µl of boiling buffer (100 mM Tris and 4 mM EDTA, pH 7.5), boiled for 2 min, and centrifuged at 1000g for 1 min. The supernatants were transferred to a fresh tube and kept on ice until measurements. ATP concentration was measured with a luminometer (Luminoskan; Thermo Electron Corporation, Waltham, MA) by adding 100 µl of luciferase reagent to 100 µl of sample or ATP standards (10-9-10-4 M). ATP values were calculated from a log-log plot of the standard curve.

    Fig. 1. Generation of OS in ER stress agent-treated cells. HL-60 cells were incubated with ER stress agents Ec (15 µM) (A), Tg (1 µM) (B), or Tu (1 µg/ml) (C) for 2 h and then loaded with 8 µMH2DCFDA for 30 min at 37°C and rinsed with PBS. Fluorescence was monitored by flow cytometry at FL-1. The gray tracing is the untreated control. The data depicted are a representative experiment. D, comparison of DCF fluorescence in arbitrary units for Ec-, Tg-, or Tutreated cells compared with control. Reported values are averages of three independent determinations and are reported ± S.E.M. **, p < 0.01 compared with control. E, time course of DCF fluorescence in response to 15 µM Ec.

    Statistical Analysis and Reproducibility. Data were analyzed by analysis of variance with Bonferroni's post tests and are expressed as means ± S.E.M. All measurements were performed in triplicate and repeated at least twice with similar results (*p < 0.05, **p < 0.01, and ***p < 0.005).

    Oxidative Stress Generation by ER Stress Agents. To investigate the possibility that ER stress inducers generate ROS, we exposed HL-60 cells to the agents Ec, Tg, and Tu for 2 h and measured OS levels in the cells by flow cytometry using the generalized OS-sensitive fluorescent dye CM-H2DCFDA (Halliwell and Whiteman, 2004). As shown in Fig. 1, A to D, increased fluorescence levels, indicating increases in OS levels, was observed for cells treated with Ec but not Tg or Tu. We have previously demonstrated that all three agents induce ER stress within the chosen 2-h time period, as indicated by induction of eukaryotic translation initiation factor 2 subunit  phosphorylation and BiP expression, along with suppression of protein synthesis (Soboloff and Berger, 2002; Zhang and Berger, 2004). However, this is achieved differently for each agent. Thus, Tu is an inhibitor of glycosylation (Kuo and Lampen, 1974) and causes ER stress through the accumulation of protein in the ER (Dorner et al., 1990). Tg inhibits the SERCA pump responsible for refilling ER Ca2+ stores (Thastrup et al., 1990), leading to rapid depletion of ER Ca2+ stores, followed by influx of Ca2+ from the extracellular milieu. Although Ec is best known as an inhibitor of Ca2+ influx, it also promotes ER Ca2+ depletion (Jan et al., 1999), although the mechanism of this Ca2+ release has not previously been addressed.

    Fig. 2. Ec hyperpolarizes MMP, generates mitochondrial ROS, and inhibits ADP transport. A, JC-1 loaded HL-60 cells were exposed to 15 µM Ec, 1 µM Tg, 1 µg/ml Tu, or 2 µM FCCP, respectively, and the membrane potential was monitored by flow cytometry as described under Materials and Methods. B, HL-60 cells were exposed to 15 µM Ec, 2 µM PMA, or 50 µM atractyloside for 2 h and then loaded with 8 µMH2DCFDA for 30 min at 37°C and rinsed with PBS. The mean fluorescence intensity was measured by flow cytometry. C, HL-60 cells were treated with ROS-generating agents described as in B but loaded with 5 µM MitoSox for 30 min at 37°C and rinsed with PBS. The mean fluorescence intensity was measured by flow cytometry. D, time course of MitoSox-sensitive ROS in response to 15 µM Ec. E, cell-free mitochondria were prepared as described under Materials and Methods. Samples were incubated with ER stress agents for 10 min in the presence or absence of atractyloside followed by the addition of [14C]ADP. ANT-dependent ADP transport activity was determined by calculating the atractyloside inhabitable counts. ANT-dependent ADP transport was inhibited by 15 µM Ec but not by 1 µMTgor1 µg/ml Tu. Values are reported as mean ± S.E.M.

    To determine the kinetics of OS generation by Ec, we followed CM-H2DCFDA fluorescence over time. As shown in Fig. 1E, fluorescence changes were observed with a short delay after addition of Ec, suggesting a relatively rapid generation of reactivity.

    Hyperpolarization of Mitochondrial Membrane Potential by Ec. The most common mechanism of OS generation by toxic agents is the disruption of electron transport in mitochondria (Ueda et al., 2002; Eckert et al., 2003; Inoue et al., 2003; Kadenbach, 2003; Rego and Oliveira, 2003). To investigate the effect of Ec on mitochondrial function, we first measured mitochondrial membrane potential, a useful indicator of mitochondrial activity and status, after exposure to Ec. As shown in Fig. 2A, the addition of Ec to cells caused an immediate hyperpolarization of the mitochondrial membrane. In contrast, Tg and Tu had no effect, whereas the protonophore Fccp caused an immediate collapse of the potential. These observations suggested that Ec, but not Tg or Tu, had a mitochondrial target.

    To further investigate the possibility of a mitochondrial target for Ec-induced ROS generation, we used MitoSox Red dye (Invitrogen), a mitochondrial-specific superoxide indicator. To confirm that MitoSox measures ROS generated at the mitochondria, we used the phorbol ester PMA, which stimulates oxidase activity at the plasma membrane (Teufelhofer et al., 2003; Dooley et al., 2004) and atractyloside, which generates mitochondrial ROS via inhibition of adenine nucleotide activity (see below) as controls. As shown in Fig. 2B, the CM-H2DCFDA dye detects similar levels of fluorescence by Ec, PMA, and atractyloside over a 2-h time period. In contrast, Ec and atractyloside, but not PMA, generate the superoxide anion, as detected with MitoSox (Fig. 2C). It is noteworthy that a nearly 3.5-fold increase in superoxide accumulation was observed in the presence of Ec, a far greater change than observed when DCF fluorescence was assessed. Cells were then stained with MitoSox, and the effect of Ec addition was followed over time. As shown in Fig. 2D, Ec stimulated a rapid and sustained increase in MitoSox fluorescence in contrast to the somewhat slower change in total DCF fluorescence (Fig. 1E). These observations are consistent with DCF, indicating ROS generation and mitochondria as the source of Ec-induced ROS.

    Fig. 3. Antimycin A reverses mitochondrial membrane hyperpolarization by Ec. A, illustration of the sites of action of inhibitors of mitochondrial electron transport. B, JC-1-loaded cells were pretreated for 20 min with 10 µM rotenone, 10 µM 4,4,4-trifluoro-1-(2-thienyl)-1,3-butanedione, or 400 µM KCN. The arrow indicates where 15 µMEcor2 µM FCCP was added to the cells. C, effect of antimycin A on membrane hyperpolarization. Cells stimulated with antimycin A (125 nM), Ec, or FCCP. Mitochondrial membrane potential was recorded for 2 h.

    Ec Inhibits Adenine Nucleotide Transport. Adenine nucleotide transport, performed by the mitochondrial ANT, is one of the final steps in the generation of ATP, and its inhibition has been shown to cause mitochondrial membrane hyperpolarization (Vander Heiden et al., 1999). We therefore investigated the effect of Ec, Tg, and Tu on adenine nucleotide transport. Cell-free mitochondria were prepared, exposed to the ER stress agents, and adenine nucleotide transport was measured. As shown in Fig. 2E, at the concentrations used to treat cells, Ec is a very potent inhibitor of ANT function, whereas Tg or Tu have no effect. This observation suggests that Ec acts directly on mitochondria to inhibit ANT, either directly or indirectly.

    Antimycin A Reverses Mitochondrial Hyperpolarization by Ec. Because adenine nucleotide transport is coupled to electron transport, it is possible that a member of the electron transport chain is the direct target of Ec, leading to ROS production and suppression of ANT function. To further investigate this possibility, we determined the effect of various inhibitors of electron transport on the ability of Ec to hyperpolarize the mitochondrial membrane. The inhibitors used are illustrated in Fig. 3A. As shown in Fig. 3B, preexposure of cells to 4,4,4-trifluoro-1-(2-thienyl)-1,3-butanedione, rotenone, or KCN, inhibitors of electron transport complexes I, II, and IV, respectively, had no effect on the ability of Ec to hyperpolarize the mitochondrial membrane. In contrast, the complex III inhibitor antimycin A both partially reduced mitochondrial membrane potential on its own and completely neutralized mitochondrial membrane hyperpolarization by Ec (Fig. 3C). Furthermore, the ability of antimycin A to neutralize Ec-induced hyperpolarization persisted for 2 h. These results therefore suggest that Ec is targeting the electron transport chain in a complex III-dependent manner, resulting in ROS generation and inhibition of ANT function.

    Antimycin A Reverses ROS Generation by Ec. The fact that antimycin A neutralizes Ec-induced hyperpolarization seems somewhat paradoxical given that antimycin A on its own has been reported to generate reactive oxygen. To further investigate the interaction between Ec and antimycin A, we measured ROS production after treatment with Ec, antimycin A, or both compounds together. Cells were incubated with the two compounds for 2 h, and ROS content was measured as described above. As shown in Fig. 4A, antimycin A alone, at the concentration used in this study, did not have any effects on ROS levels. However, it clearly suppressed ROS generation by Ec. This result therefore correlates Ec-induced hyperpolarization with ROS production and provides further evidence that the ability of Ec to generate ROS is dependent on complex III activity.

    Fig. 4. Antimycin A inhibits and reverses OS generation induced by Ec and partially restores ATP production. A, cells were incubated with 15 µM Ec, 125 nM antimycin A, or both at 37°C for 2 h, and DCF fluorescence was measured as described under Materials and Methods. Data are shown as mean ± S.E.M. *, p < 0.05; **, p < 0.01. B and C, intracellular ATP was measured in cells (0.5-1.0 x 106 cells/ml) treated with 15 µM Ec, 125 mM antimycin A, or both, at 37°C for 2 h (B) or 6 h (C). Values shown are reported as mean ± S.E.M. *, p < 0.05; **, p < 0.01.

    ATP Depletion by Ec. Given the inhibition of adenine nucleotide transport by Ec and its other effects on electron transport, it was of interest to determine the effect of Ec treatment on ATP levels. We therefore exposed cells to Ec for 2 h, and measured total ATP content. As shown in Fig. 4B, exposure to Ec for 2 h resulted in a decline in ATP content of 50%. Although a 2-h exposure to antimycin A caused a similar depletion of ATP content, exposure to both agents simultaneously resulted in significantly less ATP depletion than either agent alone, Extending the incubation time to 6 h (Fig. 4C) resulted in even greater depletion of ATP by Ec or anti-mycin A. However, as with the 2-h endpoint, combined exposure to both agents resulted in less ATP depletion than with either agent alone. These observations suggest that Ec and antimycin A both target complex III, with distinct and opposing effects.

    Antioxidants Do Not Reverse Mitochondrial Membrane Hyperpolarization by Ec. The results described above show that Ec hyperpolarizes the mitochondrial membrane and generates ROS. Moreover, both of these endpoints could be reversed by the complex III inhibitor antimycin A. These results, however, do not address the causal nature of ROS production. In particular, are increased ROS levels required for Ec-induced changes in membrane potential? To address this question, we pretreated cells with the antioxidants BHT or Nac and then followed mitochondrial membrane potential after exposure to Ec. Control experiments demonstrated that both antioxidants suppressed ROS generation by Ec (data not shown). As shown in Fig. 5, Nac slightly lowered the resting mitochondrial membrane potential compared with untreated cells. However, neither Nac nor BHT prevented Ec from hyperpolarizing the membrane above the resting level. Furthermore, the hyperpolarizing effect of Ec persisted for more than 2 h. Therefore, it is likely that the generation of ROS occurs either in parallel with or down-stream of changes in mitochondrial membrane potential.

    Fig. 5. Antioxidants do not reverse MMP hyperpolarization by Ec. Cells were pretreated with or without 5 µM BHT or 1 mM Nac at 37°C for 20 min followed by JC-1 loading and flow cytometry procedures described under Materials and Methods. Mitochondrial potential was followed for 2 h after 15 µMEcor2 µM FCCP was added.

    ER Ca2+ Depletion by Ec Is Inhibited by Antimycin A and Antioxidants. We have previously documented that Ec, in addition to blocking Ca2+ influx, also causes Ca2+ depletion from the ER (Soboloff and Berger, 2002; Zhang and Berger, 2004). To determine the role of the mitochondrial activity of Ec in ER depletion, we treated cells with Ec or Ec plus antimycin A and then measured the amount of Ca2+ present in the ER by releasing it into the cytoplasm with Tg while blocking the influx of Ca2+ with the nonspecific Ca2+ channel blocker Ni2+. As shown in Fig. 6A, no Ca2+ was released into the cytoplasm by Tg after pretreatment of cells with Ec, demonstrating that Ec depletes the ER. Antimycin A alone had little effect on Tg-releasable Ca2+. However, Ec-induced depletion of the Tg-sensitive store was reversed by antimycin A, revealing a key role for the mitochondrial target of Ec in ER Ca2+ depletion. To assess the role of mitochondria on Ca2+ influx, experiments were then performed in the absence of Ni2+. The difference between the magnitude of Tg-induced Ca2+ influx in the presence of Ni2+ versus the absence of Ni2+ reflects Ca2+ influx. Because Ec blocks Ca2+ influx in addition to depleting the ER of Ca2+, no Tg-induced changes in Ca2+ concentration occur after Ec pretreatment, even when Ni2+ is not present (Fig. 6B). Antimycin A alone had no effect on Tg-induced Ca2+ release or influx. However, the Tg-induced Ca2+ concentration observed with both anti-mycin A and Ec present was comparable with that observed in experiments when Ni2+ was present (Fig. 6A). Hence, antimycin A neutralized Ec-induced ER Ca2+ depletion, but it had little or no effect on Ec-induced inhibition of Ca2+ influx.

    Fig. 6. Ec-generated OS targets ER Ca2+ stores. Cells were loaded with indo-1AM in Tyrode's buffer as described under Materials and Methods. Ca2+ concentration was monitored by flow cytometry. A, cells were preincubated in the absence (Ctl) or presence of 15 µM Ec or Ec + 125 nM AA for 2h.Ni2+ (5 mM) was added to the cells to nonspecifically prevent influx. Ca2+ released by 3 µM Tg into the cytoplasm was used as an indicator of ER Ca2+ content. B, as in A, however, Ni2+ was not added to cells before Tg. C, as in A, except 5 µM BHT was used. D, as in C, however, Ni2+ was not added before Tg. E, as in A, except 1 mM Nac was used. F, as in E, however, Ni2+ was not added before Tg.

    We also assessed the ability of the antioxidants BHT and Nac to prevent Ec-mediated ER Ca2+ depletion and/or Ca2+ influx. As shown in Fig. 6C, in the presence of Ni2+, BHT alone had no effect on Tg-releasable Ca2+. Similar to anti-mycin A, however, BHT prevented Ec from fully depleting the Tg-sensitive store. Moreover, BHT did not alter Ec-induced inhibition of Ca2+ influx, as illustrated in Fig. 6D, where, in the presence of BHT and Ec, Tg-releasable Ca2+ was similar in magnitude to that released by Tg in the presence of Ni2+ (Fig. 6C). Similar results were observed with the antioxidant Nac (Fig. 6, E and F). These results indicate that Ec-induced ER Ca2+ depletion is probably due to its ROS production, presumably via its action on complex III within the mitochondria. They further demonstrate that Ec-induced ER Ca2+ depletion is a distinct phenomenon from Ec-induced block of Ca2+ influx, which can be separated via the actions of antimycin A, BHT, and Nac.

    Antimycin A and Antioxidants Protect Cells from Ec-Induced Cell Death. Because antimycin A and the antioxidants BHT and Nac prevent ER Ca2+ depletion by Ec, we tested its effect on Ec-induced toxicity. Cells were exposed to Ec plus antimycin A or the antioxidants for 2 h. The agents were then removed, and the cells were incubated in medium for an additional 6 h. Antimycin A, BHT, and Nac were nontoxic at the concentrations used under these conditions (data not shown). The cells were then evaluated by flow cytometry using Annexin V staining. As shown in Fig. 7A, antimycin A, BHT, and Nac provided significant protection from Ec-induced cell death, revealing a key role for mitochondrially generated ROS and the release of ER Ca2+ in Ec toxicity.

    Fig. 7. Ec-induced cell death and caspase activation are suppressed by antimycin A, BHT, and Nac. Cells were treated with 15 µMEcinthe presence or absence of 125 nM AA, 5 µM BHT, or 1 mM Nac at 37°C for 2 h, in medium containing 1% FBS. Drugs were washed out, and the cells were recovered in RPMI 1640 medium plus 10% FBS for 6 h (A). Samples were costained with Annexin V-Cy5 and propidium iodide, and cell death was determined by flow cytometry. Values include both early- and late-stage apoptotic cells. **, p < 0.01; ***, p < 0.001 compared with Ec. B, cells were treated as described above, except the recovery time was extended to18 h. Cells were then incubated with the fluorochrome inhibitor of caspases reagent (Invitrogen) to detect active caspases-3 or-7 and analyzed by flow cytometry. Reported values are percentage of cells that are positive for activated caspase-3/7 and are averages of two repeat measurements with standard errors.

    Antimycin A and Antioxidants Prevent Caspase Activation in Response to Ec. We have demonstrated in other cell types that Ec-induced cell death was associated with a variety of apoptotic indicators, including poly(ADP-ribose) polymerase cleavage and protection from cell death by caspase inhibitors (Gommerman and Berger, 1998). However, we have also observed that some cells such as human MCF-7 breast cancer cells undergo a nonapoptotic form of cell death (Zhang et al., 2002) associated with a lack of DNA fragmentation and a necrotic phenotype. We previously observed that Ec-treated HL-60 cells display classic apoptotic endpoints, including membrane blebbing and nuclear condensation (Zhang and Berger, 2004). However, caspase activation in these cells was not investigated. We therefore examined the degree of caspase activation in these cells in response to Ec, focusing on caspase-3/7, the major effectors of apoptotic cell death. Initial experiments indicated that caspase-3/7 were not activated after exposure to the drug for 2 h followed by a 6-h incubation in medium (data not shown). However, as shown in Fig. 7B, extensive caspase-3/7 activation was observed if the incubation in medium was extended to 18 h. Furthermore, the number of cells displaying caspase activation could be reduced significantly by antimycin A or the antioxidants. These observations indicate that caspase activation is a relatively late event that is at least partially dependent on the mitochondrial activity of Ec.

    Hydrogen Peroxide and Atractyloside Mobilize Ca2+ from the ER without Depleting the Store. Other studies have shown that ROS stimulate Ca2+ release from the ER (Downey, 1990). Consistent with these observations, we observed that H2O2 caused a slow, but significant Ca2+ transient in HL-60 cells (Fig. 8A). When cells were exposed to Ec, followed by H2O2, we observed that H2O2 failed to stimulate additional release of Ca2+ (Fig. 8B). This observation suggests that Ca2+ released by Ec and H2O2 are probably from the same intracellular store. It is noteworthy that when cells were treated for 2 h with H2O2, we observed that significant levels of Ca2+ could still be mobilized from the ER with Tg (Fig. 8C). This observation suggests that, unlike Ec (Fig. 6), ROS alone stimulate Ca2+ release without fully depleting the ER of Ca2+, presumably because they do not block the refilling process.

    Fig. 8. Ec and H2O2 both target the Tg-releasable Ca2+ store. A, 200 µM H2O2-induced changes in cytoplasmic Ca2+ concentration. Indo-1AM-loaded cells were exposed to H2O2, and cytoplasmic Ca2+ was followed over time. B, as described in A, except cells were preincubated in 15 µM Ec. C, Tg-induced release of ER Ca2+ content can still be detected in cells pretreated with 200 µM H2O2 for 2 h. Ni2+ (5 mM) was used to demonstrate that this was release of Ca2+ from intracellular stores and not Ca2+ entry.

    Atra is an inhibitor of the adenine nucleotide transporter (Robinson et al., 1975). Our results with Ec predict that Atra should also generate ROS, which would stimulate ER Ca2+ mobilization. To test this possibility, we exposed cells to Atra and measured ROS content. As shown in Fig. 9A, increased ROS levels were observed after Atra treatment. Furthermore, we observed that Atra could stimulate Ca2+ mobilization in HL-60 cells (Fig. 9B). However, unlike Ec, extended incubation with Atra failed to fully deplete the Tg-sensitive store (Fig. 9C). Furthermore, despite generating ROS and stimulating Ca2+ release from the ER, Atra did not induce cell death in HL-60 cells (data not shown), consistent with the concept that the cell death associated with ER Ca2+ release requires ER Ca2+ depletion.

    Fig. 9. Atractyloside-generated ROS targets ER Ca2+ stores. A, HL-60 cells were incubated with 50 µM atractyloside for 2 h and then loaded with 8 µMH2DCFDA for 30 min at 37°C. H2DCFDA fluorescence was measured by flow cytometry. B, indo-1-loaded HL-60 cells were treated with 50 µM atractyloside and cytoplasmic Ca2+ followed by flow cytometry. C, cells were pretreated with 50 µM atractyloside for 2 h. ER Ca2+ content was measured by releasing ER Ca2+ into the cytoplasm with 3 µM Tg in the presence of Ni2+ to prevent Ca2+ influx.

    High levels of ROS have multiple effects on proteins, lipids, and nucleic acids in key organelles such as the mitochondria, ER, and nucleus. Emerging evidence also supports the concept that subtoxic levels of ROS act as endogenous signaling messengers, especially in calcium signaling and protein phosphorylation (Nathan, 2003). In the course of investigating the role of OS in ER stress, we observed that Ec, an imidazole that blocks Ca2+ influx and stimulates ER Ca2+ depletion, increased OS levels in cells. In contrast, Tg or Tu, two other agents that also induce ER stress-related cell death, did not generate increased OS. Ec was also observed to cause rapid hyperpolarization of the mitochondrial membrane. This hyperpolarization was associated with inhibition of adenine nucleotide transport, observed in cell-free mitochondria. The complex III inhibitor antimycin A neutralized Ec-induced hyperpolarization of the mitochondrial membrane, suppressed ROS increases by Ec, partially reversed ATP depletion by Ec, prevented ER Ca2+ depletion, and protected cells from Ec-induced cell death. The antioxidants BHT and Nac also blocked Ca2+ mobilization from the ER by Ec and protected from Ec-induced cell death, consistent with a role for ROS in ER depletion. Taken together, these results suggest that Ec decouples complex III from downstream steps in electron transport, resulting in relatively rapid ROS generation, inhibition of adenine nucleotide transport, and ATP depletion. The reactive oxygen produced is responsible for stimulating Ca2+ release from the ER. The combination of ROS generation stimulating ER Ca2+ mobilization with Ca2+ influx blockade results in the sustained ER Ca2+ depletion associated with cell death. This model is illustrated in Fig. 10.

    Fig. 10. Model for how Ec-stimulated mitochondrial ROS contributes to ER stress. 1) Ec targets complex III in the electron transport chain resulting in hyperpolarization of the mitochondrial membrane; 2) decoupling of electron transport from adenine nucleotide transport and ATP production; 3) inhibition of ANT and ROS generation; 4) ROS stimulates ER Ca2+ release; and 5) Ec also blocks Ca2+ influx, resulting in an ER stress response.

    Because antimycin A reverses Ec-induced hyperpolarization, ROS generation, ER Ca2+ depletion, and cell death, complex III activity is probably required for ROS generation by Ec. Furthermore, the inhibition of ANT activity by Ec suggests that electron transport does not continue down the chain to the transporter. Taken together, these observations suggest that Ec decouples complex III from complex IV, presumably by providing an alternative outlet for complex III electrons (directly or indirectly). Although further work will be required to identify how Ec targets complex III causing ROS generation, our observations show that the mitochondrial target is critical for Ec-induced cell death. We have demonstrated that cancer and leukemia cells are highly sensitive to Ec-induced cell death (Gommerman and Berger, 1998; Soboloff et al., 2002; Zhang et al., 2002). Our identification of a mitochondrial target for Ec may prove to be useful in further identifying the cellular factors associated with sensitivity to this compound.

    In addition to its inhibitory effects on complex III function, antimycin A has also been reported to bind to and inhibit bcl-2 and related family members (Tzung et al., 2001). However, we suspect that this activity is unrelated to the ability of antimycin A to inhibit Ec function, because the concentration of the drug required for bcl-2 inhibition is much higher than that used in the current study. As well, we previously reported that cell death induced by Ec was unaffected by high levels of bcl-2 in cells (Gommerman and Berger, 1998), suggesting that bcl-2 does not affect the mechanism of action of Ec. Nevertheless, given the importance of bcl-2 family members and their effects on mitochondrial function, it would be of interest to further investigate a potential role for these proteins in ROS generation and Ca2+ depletion.

    We previously demonstrated that exposure to Ec for 2 h was sufficient to initiate an irreversible cell death process. It is of interest to note that after 2 h of exposure, the mitochondrial membrane remains hyperpolarized and ATP content is reduced, but only by approximately 50%. Antimycin A exposure also results in a similarly reduced ATP level, whereas the combination of the two agents results in a slightly increased ATP content. This increase may be due to reduced ATP consumption associated with decreased ROS levels rather than a restoration of ATP production.

    Cells exposed to lethal agents generally undergo variations of necrotic or apoptotic cell death (Orrenius et al., 2003). After exposure to Ec, HL-60 cells exhibit clear endpoints associated with apoptosis, such as membrane blebbing, Annexin positivity, and nuclear condensation (Zhang and Berger, 2004); however, these endpoints occur late in the cell death process. In the present study, caspase activation was observed at 18 h after Ec exposure (Fig. 7B), but not at 6 h. The inability to detect caspase activation early may be partly due to inactivation of caspases by ROS as has been documented in other cell systems (Hampton et al., 1998; Samali et al., 1999). On the other hand, apoptosis may be a secondary or indirect consequence of the ER stress process and is probably not absolutely required for cell death. We have observed, in a variety of cell systems, that 2-h exposure to Ec optimizes the difference in sensitivity between caner or leukemia cells and normal cells (Soboloff et al., 2002; Zhang et al., 2002). We have also found that ER Ca2+ release is stimulated at lower Ec concentrations than those required for blocking influx (Soboloff and Berger, 2002). Taken together, these observations suggest that longer term exposure to lower doses of Ec may result in a qualitatively different form of cell death that is more dependent on mitochondrial dysfunction than ER Ca2+ depletion.

    The ER Ca2+ store is a dynamic entity requiring constant energy and SERCA activity to maintain the large concentration gradient across the ER membrane. It is probable that as cells undergo various forms of cell death, depleted energy levels of the cell will be insufficient to maintain the Ca2+ gradient resulting in indirect ER Ca2+ depletion (Nicotera and Orrenius, 1998). In contrast, agents such as the ANT inhibitor atractyloside and others (Le Bras et al., 2005) generate ROS and cause Ca2+ release from the ER rapidly, well before energy levels are depleted. If the cell can maintain sufficient energy levels, then these agents will not be toxic. However, if the oxidative burden is too high, eventually, as energy levels deplete, the cell will undergo cell death (Richter et al., 1996).

    Our observations are consistent with a general role for ROS generated at the mitochondria in stimulating ER Ca2+ release. The full spectrum of ROS targets in the ER remains unknown. However, one likely target is the SERCA, because ROS have been reported to inhibit SERCA function (Barnes et al., 2000). Our observations that Ec and H2O2 both targeted the Tg-sensitive store are consistent with the concept that SERCA is the ROS target responsible for ER Ca2+ release.

    Both antimycin A and the antioxidants neutralized the ability of Ec to deplete the ER with no discernible effect on Ec-induced inhibition of Ca2+ influx. Furthermore, antimycin A and the antioxidants provided significant protection from cell death and caspase activation induced by Ec. Likewise, although atractyloside caused Ca2+ release from the ER, this did not lead to ER Ca2+ depletion or significant cell death. These observations are in agreement with our model that sustained Ca2+ depletion of the ER is required for lethality (Soboloff and Berger, 2002; Zhang and Berger, 2004). This would seem to suggest that although Ec releases ER Ca2+ content via ROS just like atractyloside, blocking store-operated Ca2+ is required for its toxicity and conversely, blocking Ca2+ entry is only fully toxic when ER Ca2+ content is depleted. This is consistent with our previous observation in mast cells and breast cancer cells, where Ec-induced cell death was strongly enhanced by the addition of agents that stimulate Ca2+ release (Gommerman and Berger, 1998; Soboloff and Berger, 2002; Zhang et al., 2002).

    Large increases in cytoplasmic Ca2+ caused by ER depletion coupled with influx can result in rapid increases in mitochondrial Ca2+. Excessive mitochondrial Ca2+ has been linked to activation of the mitochondrial permeability transition and induction of apoptosis (Orrenius et al., 2003). Our results suggest that ROS can perform the opposite function, namely, communicating mitochondrial dysfunction to the ER through the stimulation of Ca2+ release.

    ABBREVIATIONS: ER, endoplasmic reticulum; SERCA, sarco(endo)plasmic reticulum Ca2+ ATPase; Tg, thapsigargin; Ec, econazole; Atra, atractyloside; ROS, reactive oxygen species; OS, oxidative stress; Tu, tunicamycin; FBS, fetal bovine serum; AA, antimycin A; BHT, butylated hydroxytoluene; Nac, N-Acetyl-L-cysteine; PI, propidium iodide; CM-H2DCFDA, 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate; AM, acetoxymethyl ester; MMP, mitochondrial membrane potential; JC-1, 5,5'6,6'-tetraethylbenzimidazolcarbocyanine iodide; FCCP, carbonyl cyanide p-(trifluomethoxy) phenylhydrazone; ANT, adenine nucleotide transporter; PBS, phosphate-buffered saline; PMA, phorbol 12-myristate 13-acetate; DCF, 2',7'-dichlorofluorescein.

    1 Current affiliation: Department of Hematology, Tongji Hospital, Tongji Medical College, and Huazhong University of Science and Technology, Wuhan China, 430030.

    2 Current affiliation: Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201.

【参考文献】
  Babich LG, Shlykov SG, Borisova LA, and Kosterin SA (1994) Energy-dependent Ca2+-transport in intracellular smooth muscle structures. Biokhimiia 59: 1218-1229.

Barnes KA, Samson SE, and Grover AK (2000) Sarco/endoplasmic reticulum Ca2+-pump isoform SERCA3a is more resistant to superoxide damage than SERCA2b. Mol Cell Biochem 203: 17-21.

Dooley CT, Dore TM, Hanson GT, Jackson WC, Remington SJ, and Tsien RY (2004) Imaging dynamic redox changes in mammalian cells with green fluorescent protein indicators. J Biol Chem 279: 22284-22293.[Abstract/Free Full Text]

Dorner AJ, Wasley LC, Raney P, Haugejorden S, Green M, and Kaufman RJ (1990) The stress response in Chinese hamster ovary cells. Regulation of ERp72 and protein disulfide isomerase expression and secretion. J Biol Chem 265: 22029-22034.[Abstract/Free Full Text]

Downey JM (1990) Free radicals and their involvement during long-term myocardial ischemia and reperfusion. Annu Rev Physiol 52: 487-504.

Eckert A, Keil U, Marques CA, Bonert A, Frey C, Schussel K, and Muller WE (2003) Mitochondrial dysfunction, apoptotic cell death, and Alzheimer's disease. Biochem Pharmacol 66: 1627-1634.

Feinendegen LE (2002) Reactive oxygen species in cell responses to toxic agents. Hum Exp Toxicol 21: 85-90.[Abstract/Free Full Text]

Ferri KF and Kroemer G (2001) Organelle-specific initiation of cell death pathways. Nat Cell Biol 3: E255-E263.

Franzius D, Hoth M, and Penner R (1994) Non-specific effects of calcium entry antagonists in mast cells. Pflueg Arch Eur J Physiol 428: 433-438.

Gommerman JL and Berger SA (1998) Protection from apoptosis by steel factor but not interleukin-3 is reversed through blockade of calcium influx. Blood 91: 1891-1900.[Abstract/Free Full Text]

Grynkiewcz G, Poenie M, and Tsien RY (1985) A new generation of Ca2+ indicators with greatly improved fluorescence properties. J Biol Chem 260: 3440-3450.[Abstract/Free Full Text]

Halliwell B and Whiteman M (2004) Measuring reactive species and oxidative damage in vivo and in cell culture: how should you do it and what do the results mean? Br J Pharmacol 142: 231-255.

Hampton MB, Fadeel B, and Orrenius S (1998) Redox regulation of the caspases during apoptosis. Ann NY Acad Sci 854: 328-335.

Hayashi T, Saito A, Okuno S, Ferrand-Drake M, Dodd RL, Nishi T, Maier CM, Kinouchi H, and Chan PH (2003) Oxidative damage to the endoplasmic reticulum is implicated in ischemic neuronal cell death. J Cereb Blood Flow Metab 23: 1117-1128.

Hung CC, Ichimura T, Stevens JL, and Bonventre JV (2003) Protection of renal epithelial cells against oxidative injury by endoplasmic reticulum stress preconditioning is mediated by ERK1/2 activation. J Biol Chem 278: 29317-29326.[Abstract/Free Full Text]

Inoue M, Sato EF, Nishikawa M, Park AM, Kira Y, Imada I, and Utsumi K (2003) Mitochondrial generation of reactive oxygen species and its role in aerobic life. Curr Med Chem 10: 2495-2505.

Jan CR, Ho CM, Wu SN, and Tseng CJ (1999) Multiple effects of econazole on calcium signaling: depletion of thapsigargin-sensitive calcium store, activation of extracellular calcium influx, and inhibition of capacitative calcium entry. Biochim Biophys Acta 1448: 533-542.

Kadenbach B (2003) Intrinsic and extrinsic uncoupling of oxidative phosphorylation. Biochim Biophys Acta 1604: 77-94.

Kaufman RJ (1999) Stress signaling from the lumen of the endoplasmic reticulum: coordination of gene transcriptional and translational controls. Genes Dev 13: 1211-1233.[Free Full Text]

Kozutsumi Y, Segal M, Normington K, Gething MJ, and Sambrook J (1988) The presence of malfolded proteins in the endoplasmic reticulum signals the induction of glucose-regulated proteins. Nature (Lond) 332: 462-464.

Kuo SC and Lampen JO (1974) Tunicamycin-an inhibitor of yeast glycoprotein synthesis. Biochem Biophys Res Commun 58: 287-295.

Lai MT, Huang KL, Chang WM, and Lai YK (2003) Geldanamycin induction of grp78 requires activation of reactive oxygen species via ER stress responsive elements in 9L rat brain tumour cells. Cell Signal 15: 585-595.

Le Bras M, Clement MV, Pervaiz S, and Brenner C (2005) Reactive oxygen species and the mitochondrial signaling pathway of cell death. Histol Histopathol 20: 205-219.

Liu H, Miller E, van de Water B, and Stevens JL (1998) Endoplasmic reticulum stress proteins block oxidant-induced Ca2+ increases and cell death. J Biol Chem 273: 12858-12862.[Abstract/Free Full Text]

Nathan C (2003) Specificity of a third kind: reactive oxygen and nitrogen intermediates in cell signaling. J Clin Investig 111: 769-778.

Nicotera P and Orrenius S (1998) The role of calcium in apoptosis. Cell Calcium 23: 173-180.

Orrenius S (1985) Oxidative stress studied in intact mammalian cells. Philos Trans R Soc Lond B Biol Sci 311: 673-677.[Abstract/Free Full Text]

Orrenius S and Nicotera P (1987) Biochemical mechanisms of oxidative liver cell injury. Bull Eur Physiopathol Respir 23: 291-295.

Orrenius S, Zhivotovsky B, and Nicotera P (2003) Regulation of cell death: the calcium-apoptosis link. Nat Rev Mol Cell Biol 4: 552-565.

Patil C and Walter P (2001) Intracellular signaling from the endoplasmic reticulum to the nucleus: the unfolded protein response in yeast and mammals. Curr Opin Cell Biol 13: 349-355.

Rego AC and Oliveira CR (2003) Mitochondrial dysfunction and reactive oxygen species in excitotoxicity and apoptosis: implications for the pathogenesis of neurodegenerative diseases. Neurochem Res 28: 1563-1574.

Richter C, Schweizer M, Cossarizza A, and Franceschi C (1996) Control of apoptosis by the cellular ATP level. FEBS Lett 378: 107-110.

Robinson BH, Cheema-Dhadli S, and Halperin ML (1975) The effect of adenosine triphosphate on the tricarboxylate transporting system of rat liver mitochondria. J Biol Chem 250: 3639-3643.[Abstract/Free Full Text]

Rottenberg H and Wu S (1998) Quantitative assay by flow cytometry of the mitochondrial membrane potential in intact cells. Biochim Biophys Acta 1404: 393-404.

Samali A, Nordgren H, Zhivotovsky B, Peterson E, and Orrenius S (1999) A comparative study of apoptosis and necrosis in HepG2 cells: oxidant-induced caspase inactivation leads to necrosis. Biochem Biophys Res Commun 255: 6-11.

Slater AF, Stefan C, Nobel I, van den Dobbelsteen DJ, and Orrenius S (1995) Signalling mechanisms and oxidative stress in apoptosis. Toxicol Lett 82-83: 149-153.

Soboloff J and Berger SA (2002) Sustained ER Ca2+ depletion suppresses protein synthesis and induces activation-enhanced cell death in mast cells. J Biol Chem 277: 13812-13820.[Abstract/Free Full Text]

Soboloff J, Zhang Y, Minden M, and Berger SA (2002) Sensitivity of myeloid leukemia cells to calcium influx blockade: application to bone marrow purging. Exp Hematol 30: 1219-1226.

Szegezdi E, Fitzgerald U, and Samali A (2003) Caspase-12 and ER-stress-mediated apoptosis: the story so far. Ann NY Acad Sci 1010: 186-194.

Teufelhofer O, Weiss RM, Parzefall W, Schulte-Hermann R, Micksche M, Berger W, and Elbling L (2003) Promyelocytic HL60 cells express NADPH oxidase and are excellent targets in a rapid spectrophotometric microplate assay for extracellular superoxide. Toxicol Sci 76: 376-383.[Abstract/Free Full Text]

Thastrup O, Cullen PJ, Drobak BK, Hanley MR, and Dawson AP (1990) Thapsigargin, a tumor promoter, discharges intracellular Ca2+ stores by specific inhibition of the endoplasmic reticulum Ca2(+)-ATPase. Proc Natl Acad Sci USA 87: 2466-2470.[Abstract/Free Full Text]

Tzung SP, Kim KM, Basanez G, Giedt CD, Simon J, Zimmerberg J, Zhang KY, and Hockenbery DM (2001) Antimycin A mimics a cell-death-inducing Bcl-2 homology domain 3. Nat Cell Biol 3: 183-191.

Ueda S, Masutani H, Nakamura H, Tanaka T, Ueno M, and Yodoi J (2002) Redox control of cell death. Antioxid Redox Signal 4: 405-414.

Vander Heiden MG, Chandel NS, Schumacker PT, and Thompson CB (1999) Bcl-xL prevents cell death following growth factor withdrawal by facilitating mitochondrial ATP/ADP exchange. Mol Cell 3: 159-167.

Wang XZ, Lawson B, Brewer JW, Zinszner H, Sanjay A, Mi LJ, Boorstein R, Kreibich G, Hendershot LM, and Ron D (1996) Signals from the stressed endoplasmic reticulum induce C/EBP-homologous protein (CHOP/GADD153). Mol Cell Biol 16: 4273-4280.

Watanabe Y, Suzuki O, Haruyama T, and Akaike T (2003) Interferon-gamma induces reactive oxygen species and endoplasmic reticulum stress at the hepatic apoptosis. J Cell Biochem 89: 244-253.

Zhang Y and Berger SA (2004) Increased calcium influx and ribosomal content correlate with resistance to endoplasmic reticulum stress-induced cell death in mutant leukemia cell lines. J Biol Chem 279: 6507-6516.[Abstract/Free Full Text]

Zhang Y, Crump M, and Berger SA (2002) Purging of contaminating breast cancer cells from hematopoietic progenitor cell preparations using activation enhanced cell death. Breast Cancer Res Treat 72: 265-278.


作者单位:Arthritis and Immune Disorder Research Centre, University Health Network and Department of Immunology, University of Toronto; Toronto, Ontario, Canada

作者: 2009-8-25
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具