Tumor cell response to drug induced apoptosis is a function of intra cellular redox status 2

12 237 0
Tumor cell response to drug induced apoptosis is a function of intra cellular redox status 2

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

[CANCER RESEARCH 64, 7867–7878, November 1, 2004] Hydrogen Peroxide-Mediated Cytosolic Acidification Is a Signal for Mitochondrial Translocation of Bax during Drug-Induced Apoptosis of Tumor Cells Kashif A. Ahmad,1 Kartini B. Iskandar,1 Jayshree L. Hirpara,1 Marie-Veronique Clement,3,4 and Shazib Pervaiz1,2,4 Department of Physiology, 2Oncology Research Institute, 3Department of Biochemistry, Faculty of Medicine, and 4National University of Singapore (NUS) Graduate School for Integrative Sciences and Engineering, NUS, Singapore ABSTRACT Absence of the proapoptotic protein Bax renders tumor cells resistant to drug-induced apoptosis. We have shown that hydrogen peroxide (H2O2)-mediated cytosolic acidification is an effector mechanism during drug-induced apoptosis of tumor cells. Here, we report that Bax is critical in determining the sensitivity of tumor cells to H2O2-induced apoptosis. More importantly, exposure of colorectal carcinoma (HCT116) and leukemia cells (HL60 and CEM) to H2O2 or its intracellular production during drug-induced apoptosis is a signal for mitochondrial translocation of Bax. Furthermore, we provide evidence that drug-induced H2O2mediated Bax translocation in tumor cells is caspase independent but involves cytosolic acidification. Inhibiting cytosolic acidification prevents Bax translocation, and contrarily enforced acidification of the intracellular milieu results in mitochondrial recruitment of Bax, even in the absence of a trigger. These findings provide a novel mechanism for mitochondrial translocation of Bax and directly implicate H2O2-mediated cytosolic acidification in the recruitment of the mitochondrial pathway during druginduced apoptosis of tumor cells. INTRODUCTION Apoptotic execution is orchestrated by intricate networking between caspases and apoptogenic factors released from the mitochondria (1). A growing body of evidence seems to favor the involvement of intracellular reactive oxygen species at some point during apoptotic execution (2– 6). These observations become more important considering the critical role of the mitochondria during apoptosis and the fact that mitochondria have been implicated directly or indirectly as the prime source of reactive oxygen species during drug-induced apoptosis (2, 5, 7, 8). However, it is still unclear whether reactive oxygen species generation is a critical initial trigger or a downstream effect of caspase-mediated mitochondrial damage. To that end, our findings have highlighted the regulatory role of intracellular reactive oxygen species in the apoptotic pathway (6, –11). We showed that intracellular increase in H2O2 was a critical effector mechanism during drug-induced apoptosis of human tumor cells (5). This increase in H2O2 was responsible for early cytosolic acidification, thus creating an environment conducive for caspase activation. Although our data and a number of other reports describing H2O2-mediated apoptosis, e.g., the inhibitory effect of Bcl-2 overexpression on H2O2-induced death signaling (12–14) and the up-regulation of the proapoptotic protein Bax in some systems (15), point to the mitochondria as the target organelle, the upstream events leading to the engagement of the mitochondria remain clouded. The role of the proapoptotic protein Bax in the recruitment of the mitochondria has been well established (16 –18). A well-accepted model is the death receptor-mediated activation of caspase that triggers Bid processing (19 –21). Truncated Bid can then signal translocation of cytosolic Bax to the mitochondria (19), where it can form homo- or heterodimers with other Bcl-2 family members. The resultant conformational change in Bax can result in channel formation that could mediate egress of proteins, such as cytochrome c, from the mitochondrial intermembrane space (22–25), thereby activating downstream execution. Hence, translocation of Bax is a critical signal for the involvement of the mitochondrial death machinery, and redistribution of Bax has been reported with a variety of apoptotic stimuli (22, 23, 26 –28). The critical involvement of Bax during drug-induced apoptosis is further supported by the relative insensitivity of cells lacking Bax expression to drug treatment (29) and observation that BaxϪ/Ϫ cells when xenografted in mice are deficient in apoptotic signaling. In the light of these findings, the objectives of this study were 2-fold: (1) to decipher the role of Bax in H2O2-mediated apoptosis, and (2) to investigate whether the intracellular increase in H2O2 was a signal for Bax translocation during drug-induced apoptosis of tumor cells. Here, we report that H2O2-induced apoptosis is inhibited in colorectal carcinoma cells lacking Bax (HCT116 BaxϪ/Ϫ), unlike HCT116 Baxϩ/Ϫ cells. Exposure of HCT 116 Baxϩ/Ϫ or HL60 human leukemia cells to H2O2 or its intracellular generation during drug-induced apoptosis signals translocation of Bax to the mitochondria, which is mediated by cytosolic acidification. MATERIALS AND METHODS Cell Lines. The human leukemia HL60 and CEM cell lines were purchased from American Type Culture Collection (Rockville, MD) and maintained in RPMI 1640, supplemented with 10% fetal bovine serum, 1% L-glutamine, and 1% S-Penicillin. HCT116 Baxϩ/Ϫ and BaxϪ/Ϫ cell lines were generous gifts from Dr. Vogelstein at Johns Hopkins University (Baltimore, MD). HCT116 cell lines were maintained in McCoy’s 5A (Invitrogen Life Technologies, Inc., Carlsbad, CA, USA), supplemented with 10% fetal bovine serum, 1% L-glutamine, and 1% S-Penicillin. All cell lines were maintained in a 37°C incubator with 5% CO2. Apoptosis was induced by exposure of cells (1 ϫ 106/ mL) to H2O2 (100 –500 ␮mol/L) or the anticancer agent merodantoin (C1; 50 ␮g/mL; ref. 30) for to 18 hours. Cell survival was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay as described previously (31). Subcellular Fractionation. Subcellular fractions were obtained as described previously (5). Briefly, 20 ϫ 106 cells were washed with ice-chilled ϫ PBS at 1200 ϫ g. Cell pellets were resuspended in 500 ␮L of extraction buffer [200 mmol/L mannitol, 68 mmol/L sucrose, 50 mmol/L Pipes (pH 7.4), 50 mmol/L KCl, mmol/L EGTA, mmol/L MgCl2, mmol/L DTT, and protease inhibitor mixture in double distilled H2O] and incubated at 4°C for 20 minutes, followed by Dounce homogenization. The homogenate was centrifuged at 150 ϫ g for minutes at 4°C. The supernatant was additionally centrifuged at 14,000 ϫ g for 10 minutes (fraction enriched with intact Received 2/22/04; revised 8/23/04; accepted 8/31/04. mitochondria). The supernatant from the last centrifugation was used as the Grant support: The National Medical Research Council (R-185-000-032-213) and cytosolic fraction. Purity of the mitochondrial fractions was confirmed by Biomedical Research Council (R-185-000-048-305), Singapore. Western blot analysis using a monoclonal antibody that recognizes the mitoThe costs of publication of this article were defrayed in part by the payment of page chondrial-specific protein MnSOD. charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. Determination of Caspase 3, 8, and Activities. Caspase 3, 8, and Requests for reprints: Shazib Pervaiz, Department of Physiology and Oncology activities were assayed by using AFC-conjugated substrates supplied by BioResearch Institute, Faculty of Medicine, National University of Singapore, MD9, #03-06, Rad Laboratories (Hercules, CA). Cells (1 ϫ 106/mL) were exposed to H2O2 Singapore 117597. Phone: 65-6874-6602; Fax: 65-6778-8161; E-mail: phssp@nus.edu.sg. (100 –500 ␮mol/L) or C1 (50 ␮g/mL) to 24 hours, washed twice with ©2004 American Association for Cancer Research. 7867 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS ϫ PBS, resuspended in 50 ␮L of chilled cell lysis buffer (provided by the supplier), and incubated on ice for 10 minutes. Fifty ␮L of ϫ reaction buffer (10 mmol/L HEPES, mmol/L EDTA, 10 mmol/L KCl, 1.5 mmol/L MgCl2, and 10 mmol/L DTT) and ␮L of the fluorogenic caspase-specific substrate (DEVD-AFC for caspase 3, LETD-AFC for caspase 8, and LEHD-AFC for caspase 9) were added to each sample and incubated at 37°C for hour. Protease activity was determined by the relative fluorescence intensity at 505 nm after excitation at 400 nm using a spectrofluorimeter (TECAN Spectrofluor Plus, Maennedorf, Switzerland). Results are shown as fold increase (ϫ increase) in activity relative to untreated cells (1ϫ). Caspase activation was also assessed by staining cells with an affinity-purified antibody that recognizes the processed (active) form of caspase (tagged with phycoerythrin; BD PharMingen) and analyzed by flow cytometry. Propidium Iodide Staining for DNA Fragmentation. Briefly, ϫ 106 cells/mL were triggered with H2O2 or C1 for 24 hours, fixed with 70% EtOH, and stained with propidium iodide for DNA content analysis as described elsewhere (5). Events (Ն10,000) were analyzed by flow cytometry with the excitation set at 488 nm and emission at 610 nm. Data are shown as a percentage of cells with subdiploid DNA and are mean Ϯ SD of three independent observations. In addition, apoptosis on exposure of cells to H2O2 or C1 was also verified by assaying exposure of phosphatidylserine using the Apoalert Annexin-V kit (Clontech) and analyzed by flow cytometry using excitation and emission wavelengths at 488 and 525 nm, respectively. Flow Cytometric Analysis of Intracellular H2O2 Concentration. Cells were exposed to the apoptotic trigger for 15 minutes to 12 hours, loaded with 5-(and-6)-chloromethyl-2Ј,7Ј-dichlorofluorescin diacetate (5 ␮mol/L) at 37°C for 15 minutes, and analyzed by flow cytometry (Coulter EPICS Elite ESP) using an excitation wavelength of 488 nm, as described previously (32). In addition, intracellular reactive oxygen species production was assessed by a lucigenin-based chemiluminescence assay as described previously (16) or flow cytometry using the sensitive probe hydroethidine (2 ␮mol/L). Chemiluminescence was monitored for 60 seconds in a TD-20/20 Luminometer (Turner Designs, Sunnyvale, CA). Lucigenin is a widely used and validated chemiluminescent detector of intracellular O2Ϫ in biological systems (33, 34). Data are shown as relative light units (RLU) per microgram of protein (RLU ␮gϪ1 protein) Ϯ SD from three to six independent measurements. Protein concentration was determined using the Coomassie Plus protein assay reagent from Pierce (Pierce Chemical Co., Rockford, IL). Fig. 1. H2O2-induced apoptosis is Bax dependent. In A, lysates from HCT116 Baxϩ/Ϫ and BaxϪ/Ϫ cells (2 ϫ 106) were subjected to 10% SDS-PAGE, and the presence or absence of Bax was confirmed by Western blot analysis using monoclonal anti-Bax. In B, ϫ 106/mL HCT116Baxϩ/Ϫ (Œ) and BaxϪ/Ϫ (f) cells were exposed to 100 ␮mol/L H2O2 for to 24 hours, and cell survival was assessed by the MTT assay. In C, caspases 3, 8, and activities were determined in HCT116Baxϩ/Ϫ cells using fluorogenic substrates as described in Materials and Methods. Results are shown as fold increase (ϫ increase) in caspase activity over the untreated cells (1ϫ). In D, HCT116 Baxϩ/Ϫ cells were exposed to H2O2 for 12 hours and analyzed by flow cytometry for phosphatidylserine exposure and processed caspase as described in Materials and Methods. E, Western blot analysis of poly(ADP-ribose) polymerase cleavage using anti-poly(ADP-ribose) polymerase (clone C-2–10; PharMingen) in Baxϩ/Ϫ and BaxϪ/Ϫ cells after 24-hour incubation with 100 ␮mol/L H2O2. In F, Baxϩ/Ϫ and BaxϪ/Ϫ cells were analyzed for DNA fragmentation by PI staining after 24-hour treatment with 100 ␮mol/L H2O2 as described in Materials and Methods. Subdiploid population (Sub-G1 fraction) is shown as mean Ϯ SD from three independent experiments. G, Western blot analysis of cytochrome c (Cyt. C) in cytosols of Baxϩ/Ϫ and BaxϪ/Ϫ cells after 12-hour treatment with 100 ␮mol/L H2O2 in the presence or absence of 1000 units/mL catalase. Membrane was reprobed with anti-␤actin as a loading control. In H, HCT116BaxϪ/Ϫ cells were transiently transfected with Bax as described in Materials and Methods and exposed to 100 ␮mol/L H2O2 for 24 h. Cell survival was assessed by the ␤-gal survival assay described elsewhere (36). All data shown are mean Ϯ SD of at least three independent experiments [MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; PI, propidium iodide]. 7868 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS Detection of Cytochrome C Release from the Mitochondria. Cytochrome c release was assessed by Western blot analysis of cytosolic extracts from 30 ϫ 106 cells as described previously (5) using anti-cytochrome c (7H8.2C12, PharMingen, San Diego, CA). Signal was detected by the Super Signal Substrate Western Blotting kit (Pierce). Determination of Mitochondrial ⌬⌿m by Flow Cytometry. Potentialsensitive probe 3, 3Ј dihexyloxacarbocyanine iodide (DiOC6) was used to measure mitochondrial ⌬␺m as described previously (31). Briefly, ϫ 106 cells were incubated with 3,3ЈDiOC6 (40 nmol/L) for 15 minutes at 37°C and immediately analyzed in Epic Profile flow cytometer with excitation set at 488 nm. Data were analyzed for 10,000 events using the WinMDI software. Western Blot Analyses of Poly(ADP-Ribose) Polymerase Cleavage, Bax, and Bid. For analysis of poly(ADP-ribose) polymerase cleavage, lysates from ϫ 106 cells were prepared in sample buffer [50 mmol/L Tris/HCl (pH 6.8), mol/L urea, 3% SDS, 0.003% bromphenol blue, and 6% ␤-mercaptoethanol] and subjected to Western blot analysis using anti-poly(ADP-ribose) polymerase (clone C-2–10, PharMingen) as described (30). For Western blot analysis of Bax, cells (2 ϫ 106) were lysed by adding 100 ␮L of chilled ϫ radioimmunoprecipitation assay buffer lysis buffer, and 50 ␮g of protein were subjected to 15% PAGE and transferred to polyvinylidene difluoride as above. Alternatively for analysis of Bax dimerization/multimerization, cell lysates were subjected to 10% native gel electrophoresis. Membranes were exposed to 1:2,000 dilution of mouse monoclonal anti-Bax antibody (clone 6A7, BD Pharmigen, San Diego, CA) at 25°C for hours, followed by 1:5,000 dilution of goat antimouse IgG-horseradish peroxidase. The anti-Bax antibody (6A7) recognizes epitopes that are in the vicinity of the dimerization domains of Bax (AA 12–24; ref. 35). Western blot analysis for Bid cleavage was performed on whole cell lysates using a rabbit polyclonal anti-Bid IgG (Biovision Research Products, Paolo Alto, CA) that recognizes the Mr 22,000 full-length Bid. Chemiluminescence was detected as described above. Transient Transfection of HCT116 Bax؊/؊ Cells with pcDNA3-Bax. Transient transfection of HCT116 BaxϪ/Ϫ cells with Bax was performed using pcDNA3-Bax (generously provided by Prof. Stanley Korsmeyer, Boston, MA). HCT116 BaxϪ/Ϫ (2 ϫ 105) cells were seeded per six-well plate in mL of 10% McCoy’s Medium and incubated at 37°C for 24 hours. The medium was then removed, and cells were resuspended in 1.5 mL of normal growth medium. Cells were cotransfected with a ␤- galactosidase-containing plasmid (p-cytomegalovirus-␤ galactosidase) at a ratio of 3.5 ␮g of pCDNA3-Bax:0.5 ␮g of p-cytomegalovirus-␤-galactosidase as described previously (36). Luminescence was recorded with a Luminometer (TD20/20; Turner Designs). The relative light units were calculated per microgram of protein. Measurement and Manipulation of Cytosolic pH. For measurement of cytosolic pH, cells were loaded with 10 ␮mol/L BCECF-AM [2Ј,7Ј-bis(2carboxyethyl)-5,6-carboxyfluorescein; Sigma, St. Louis, MO], and the fluorescence ratio of 525:610 nm was used to derive cytosolic pH using a standard pH calibration curve as described previously (5). To manipulate cytosolic pH, cells were incubated with a known inhibitor of the Naϩ-Hϩ antiporter, methylamiloride (10 – 40 ␮mol/L), for hours before the measurements. Alternatively, cells were preincubated with to ␮mol/L resveratrol for hours before exposure to the apoptotic stimuli and pH. In our recent study (37), we have demonstrated the inhibitory effect of low doses of resveratrol on drug-induced acidification. RESULTS H2O2-Induced Apoptosis Is Blocked in Bax؊/؊ Cells. The sensitivity of human colon carcinoma HCT116 Baxϩ/Ϫ and HCT116 BaxϪ/Ϫ cells (Fig. 1A) to H2O2 (100 ␮mol/L) for to 24 hours was determined. Unlike HCT116 Baxϩ/Ϫ cells, BaxϪ/Ϫ cells were resistant to the effect of H2O2 (Fig. 1B). H2O2 triggered significant increases in caspases and activities (but no caspase activation or cleavage of Bid; Fig. 1C), increase in membrane phosphatidylserine exposure and processing of caspase (Fig. 1D), cleavage of caspase substrate poly(ADP-ribose) polymerase (Fig. 1E), and increase in subdiploid DNA fraction (Fig. 1E) in Baxϩ/Ϫ cells, whereas little effects on caspase activation (Fig. 1, C and E), and DNA fragmentation (Fig. 1F) were observed in BaxϪ/Ϫ cells. H2O2 also induced cytosolic translocation of cytochrome c in HCT 116 Baxϩ/Ϫ cells (inhibited by catalase) but not the BaxϪ/Ϫ variant (Fig. 1G). Furthermore, transient transfection of BaxϪ/Ϫ cells with a vector containing full-length Bax (pcDNA3-Bax) restored their sensitivity to H2O2 (Fig. 1H), thus highlighting the importance of Bax in H2O2induced apoptosis. Fig. Continued. 7869 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS H2O2 Influences Subcellular Localization of Bax. H2O2 has been shown to induce up-regulation of Bax and a shift in the intracellular Bax:Bcl-2 ratio in some systems (38). Although exposure of HCT116 Baxϩ/Ϫ cells to H2O2 did not significantly change the Bax:Bcl-2 ratio (Fig. 2A), analysis of subcellular fractions clearly showed translocation of Bax from the cytosol to the mitochondria (Fig. 2B). Mitochondrial localization of Bax could be detected hours after exposure to H2O2 (data not shown) and was maximal at 12 hours after treatment (Fig. 2B). Importantly, Bax translocation was inhibited by the H2O2 scavenger catalase (1000 units/mL), additionally supporting the role of H2O2 in mitochondrial translocation of Bax. Contrarily, preincubation with the general caspase inhibitor ZVADfmk (50 ␮mol/L) did not inhibit H2O2-induced Bax translocation (Fig. 2C). Similar to HCT116 cells, HL60 and CEM leukemia cells underwent apoptosis on exposure to H2O2, as indicated by the appearance of subdiploid DNA, increases in caspase and activities, processing of caspase 3, and decrease in cell survival (Fig. 3, A–D). In addition, the involvement of the mitochondrial death pathway was evident by the drop in ⌬⌿m of cells exposed to H2O2 (Fig. 4A) and cytosolic translocation of cytochrome c from the mitochondria (Fig. 4B) that was significantly inhibited by catalase. More importantly, cytosolic Bax was redistributed to the mitochondrial fractions in HL60 and CEM cells, which could be inhibited by catalase (Fig. 4, C and D) but not ZVAD-fmk (Fig. 4C). These data suggest a critical role for H2O2 Fig. 2. H2O2 triggers mitochondrial translocation of Bax. In A, lysates of HCT116Baxϩ/Ϫ cells (5 ϫ 106) after exposure to H2O2 for –24 hours were subjected to Western blot analysis using anti-Bax or anti-Bcl-2 antibodies as described in Materials and Methods. In B, HCT116Baxϩ/Ϫ cells (1 ϫ 107) were treated with H2O2 for 12 hours in the presence or absence of either 1,000 units/mL catalase or 100 ␮mol/L ZVAD-fmk (C), and cytosolic and mitochondrial fractions were subjected to Western blot analysis using monoclonal anti-Bax. Mitochondrial purity was verified by blotting with a monoclonal antibody against the mitochondrial protein MnSOD. in signaling Bax to the mitochondria but argue against the involvement of the caspase family in this pathway. H2O2 Is a Signal for Bax Translocation during Drug-Induced Apoptosis. Having demonstrated that exogenously added H2O2 triggered translocation of Bax in tumor cells, we next asked whether this was also the mechanism used by anticancer drugs that trigger apoptosis via intracellular H2O2 production. HL60 cells were exposed to the novel experimental anticancer agent C1, a small (Mr ϭ 242) synthetic compound, first isolated and purified on photo-oxidation of merocyanine 540. The chemical structure of this imidazole compound is N,NЈ-Dibutyl-thio-4,5-imidazolindion, and its apoptosis-inducing activity was reported previously by our group (30). Exposure of HL60 cells to 50 ␮g/mL C1 for hours resulted in a significant increase in intracellular H2O2 (Fig. 5A, panel a). The intracellular generation of reactive oxygen species was additionally confirmed by a lucigenin-based chemiluminescence assay and flow cytometry on loading cells with hydroethidine. Results clearly indicate that exposure of cells to C1 for to hours resulted in a significant increase (1.4 –2ϫ) in intracellular reactive oxygen species compared with the cells treated with the carrier solvent (Fig. 5, panel b). Similarly, a right shift in the fluorescence of C1-treated cells loaded with hydroethidine (Fig. 5, panel c; mean fluorescence intensity shifted from 2.9 to 3.25) provided additional proof for druginduced increases in intracellular reactive oxygen species generation. In addition, exposure to C1 resulted in a significant decrease in cell survival (Fig. 5B), H2O2-dependent increase in subdiploid DNA fraction, increased processing of caspase 3, and cleavage of the caspase substrate poly(ADP-ribose) polymerase (Fig. 5, C–E). In addition, our data clearly provide evidence that C1 induced apoptosis by targeting the mitochondria, as demonstrated by the drop in ⌬⌿m of cells, cytosolic translocation of cytochrome c, and increase in caspase activity (Fig. 6, A–C). More importantly, Bax translocated from the cytosol to the mitochondria on exposure to C1, which could be significantly inhibited by catalase but not the general caspase inhibitor ZVAD-fmk (Fig. 6D). In addition, native gel electrophoresis revealed that C1, similar to pure H2O2, triggered dimerization and multimerization of Bax (Fig. 6E). A careful look at the blots in Fig. 6, D and E, showed that the intensity of the bands was much weaker in the catalase pretreated lanes in both the cases. This was additionally reinforced by a stronger inhibition of the oligomeric form by catalase (Fig. 6E), thus corroborating the involvement of H2O2 in C1-induced Bax activation. The fact that preincubation of cells with H2O2 scavenger catalase significantly blocked all mitochondrial changes triggered by C1 in tumor cells (Fig. 6) underscores the critical role of H2O2 as the mediator of mitochondrial recruitment and apoptosis triggered by C1. H2O2-Induced Cytosolic Acidification Signals Mitochondrial Recruitment of Bax. We have demonstrated previously that H2O2 added exogenously or triggered endogenously during drug-induced apoptosis is a stimulus for cytosolic acidification, thereby creating a permissive intracellular milieu for death execution (5, 6). In agreement with our earlier studies, here we show that C1 or H2O2 induced a significant drop in cytosolic pH, which could be inhibited by catalase (Fig. 7A). Therefore, we questioned whether H2O2-dependent cytosolic acidification could be the stimulus for Bax translocation during drug-induced apoptosis. To prove that, we exploited the ability of low doses of the polyphenolic compound resveratrol to block H2O2 and drug-induced pH drops in human leukemia cells (37). Corroborating our recent findings, preincubation of HL60 cells for hours with to ␮mol/L resveratrol inhibited acidification induced by C1 or H2O2 (Fig. 7B). More interestingly, resveratrol exposure blocked mitochondrial translocation of Bax induced by either of the stimuli (Fig. 7, C and D). The effect of resveratrol on C1-induced Bax 7870 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS Fig. 3. H2O2 induces apoptosis and in human leukemia cells. HL60 and CEM leukemia cells (1 ϫ 106/mL) were incubated with 100 ␮mol/L H2O2 for 12 hours in the presence or absence of 1,000 units/mL catalase. In A, DNA fragmentation was assessed by PI staining; in B, activities of caspases and were determined by fluorogenic assays as described in Materials and Methods. Data are shown as fold increase (ϫ increase) in activity over untreated cells (1ϫ) and mean Ϯ SD of three independent experiments. In C, active caspase was assayed by staining cells with an antibody that recognizes the processed form of caspase tagged with PE. Cells were then analyzed by flow cytometry. In D, cell survival on H2O2 treatment (100 ␮mol/L for 18 hours) was assessed by MTT assay as described in Materials and Methods [PI, propidium iodide; PE, phycoerythrin; MTT, 3-(4,5-dimethylthiazol2-yl)-2,5-diphenyltetrazolium bromide]. activation appears to be a function of its inhibitory effect on Bax dimerization (data not shown). It should be pointed out that resveratrol treatment for to 24 hours did not alter the overall expression of either Bax or Bcl-2 (Fig. 7E). Furthermore, we have reported previously that resveratrol did not elicit H2O2 scavenging activity at these low concentrations. Stimulated by these findings and to verify that drop in cytosolic pH was a signal for Bax translocation, we assessed the effect of enforced intracellular acidification on Bax translocation. To that end, we made use of the pharmacological inhibitor of the pH regulator Naϩ/Hϩ antiporter, methylamiloride. As expected, methylamiloride (10 – 40 ␮mol/L) induced a significant drop in cytosolic pH (Fig. 8A). Most interestingly, exposure of tumor cells to methylamiloride alone resulted in significant translocation of Bax to the mitochondria (Fig. 8B), which was followed by cell death (Fig. 8C). It should be pointed out that exposure of cells to methylamiloride for to 12 hours did not alter the intracellular expression of Bax or Bcl-2 (Fig. 8D). Indeed, enforced acidification of the milieu was a much stronger stimulus for translocation of Bax as shown in Fig. 8B (virtually all Bax translocated as compared with ϳ60% in case of C1), additionally consolidating the direct effect of a pH drop on mitochondrial recruitment of Bax. These data provide strong evidence to support cytosolic acidification, downstream of H2O2 production, as an effector mechanism for Bax translocation during drug-induced apoptosis of tumor cells. Translocation of Bax Triggers Additional Increase in Mitochondrial H2O2 Production. Current opinion holds that the principal source of reactive oxygen species, including H2O2, during apoptotic signaling is the mitochondria (2, 5, 12). How does one reconcile with these findings given our observations that H2O2 acts upstream of the mitochondria to signal mitochondrial localization of Bax? To address this, we asked whether mitochondrial translocation of Bax resulted in a second wave of intracellular H2O2. HCT116 Baxϩ/Ϫ and BaxϪ/Ϫ cells were exposed to 100 ␮mol/L H2O2, and the intracellular level of H2O2 was determined at 30 minutes, 1, 6, and 12 hours. The initial increase in fluorescence observed in both Baxϩ/Ϫ and BaxϪ/Ϫ cells is indicative of cellular uptake of exogenously added H2O2 (Fig. 9A, panel a). However, by hour, intracellular H2O2 returned to the baseline in BaxϪ/Ϫ cells and remained unchanged for Յ12 hours (Fig. 9A, panels b and c). On the contrary, Baxϩ/Ϫ cells showed a subsequent increase in intracellular H2O2 starting at hours and signif- 7871 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS Fig. 4. H2O2 triggers caspase-independent translocation of Bax to the mitochondria. In A, HL60 cells (1 ϫ 106/mL) were incubated with H2O2 (100 ␮mol/L for hours) or CICCP (100 ␮mol/L for hour), and the change in ⌬␺m was assessed by flow cytometry using DiOC6 as described in Materials and Methods. In B, cytosolic fractions from HL60 cells (2.5 ϫ 107) treated with 100 ␮mol/L H2O2 for 12 hours in the presence or absence of 1,000 units/mL catalase were subjected to Western blot analysis for cytochrome c. Anti-␤ actin was used as a loading control. In C, mitochondrial and cytosolic fractions from HL60 cells (2.5 ϫ 107), incubated with 100 ␮mol/L H2O2 for 12 hours in the presence or absence of catalase or ZVAD-fmk, were subjected to Western blot analysis using anti-Bax. Mitochondrial purity was confirmed by probing with anti-MnSOD. D, Western blot analysis using anti-Bax on fractions obtained from CEM cells after exposure to 100 ␮mol/L H2O2 with or without catalase. icantly detectable even after 12 hours of the initial exposure to H2O2 (Fig. 9A, panels b and c). Indeed, a summary of three independent experiments showing change (percentage of control cells) in mean fluorescence intensity of dichlorofluorescin clearly demonstrates a significant increase in mean fluorescence intensity at and 12 hours after exposure to H2O2 in Baxϩ/Ϫ cells, whereas no detectable difference is seen in BaxϪ/Ϫ cells (Fig. 9B). Intracellular reactive oxygen species production was further verified in Baxϩ/Ϫ and BaxϪ/Ϫ cell lines by hydroethidine staining and flow cytometry (Fig. 9C). It is important to point out that the translocation of Bax to the mitochondria is also detected first at hours after incubation with H2O2 with a maximum shift observed at 12-hour postincubation (Fig. 9D). Furthermore, to confirm that intracellular reactive oxygen species generation on exposure to the apoptosis-inducing agent C1 was, indeed, upstream of Bax, HCT116Baxϩ/Ϫ and BaxϪ/Ϫ cells were exposed to C1, and reactive oxygen species production was assessed by loading cells with DCHF-DA or hydroethidine and analyzed by flow cytometry. Results clearly indicate that C1 triggered reactive oxygen species production in both cell lines as early as 15 minutes after exposure (Fig. 10); however, similar to H2O2, the second increase in intracellular reactive oxygen species was only observed in Baxϩ/Ϫ cells (data not shown). These results suggest a scenario where exogenous H2O2 or intracellular H2O2 produced by drug exposure triggers Bax translocation (via inducing cytosolic acidification), which then acts as a signal for the second wave of H2O2 production from the mitochondria, thereby reinforcing the death signal. DISCUSSION H2O2: Signal for Mitochondrial Targeting of Bax. Our recent findings and many related studies strongly argue in favor of a signaling role for reactive oxygen species, such as H2O2 (5, 10, 11, 38, 39). Aside from the up-regulation of Bax on exposure of some cell types to H2O2, the inhibitory effect of Bcl-2 overexpression (12) supports the involvement of the mitochondrial death pathway in H2O2-induced apoptosis. However, as the mitochondria are a major source of intracellular reactive oxygen species, it is tempting to speculate that reactive oxygen species, such as H2O2, may function both upstream and downstream of the mitochondria. Tumor cells lacking Bax (BaxϪ/Ϫ) are resistant to the effect of some anti-cancer drugs (29). In agreement with that, we show here that HCT116 Bax(Ϫ/Ϫ) cells are resistant to death induced by apoptotic concentrations of H2O2. Furthermore, transient transfection of Bax in HCT116 Bax(Ϫ/Ϫ) cells restores their sensitivity to H2O2, thus underscoring the critical role of Bax in H2O2-induced apoptosis. One probable mechanism underlying the differential sensitivity of Baxϩ/Ϫ and BaxϪ/Ϫ cells to H2O2 could be that H2O2 increases the expression of Bax or alters the cellular Bax:Bcl-2 ratio in Baxϩ/Ϫ cells, as suggested by some studies. However, our results show that the Bax:Bcl-2 ratio is not significantly altered on 4- to 24-hour exposure to H2O2. Interestingly, analysis of subcellular distribution of Bax (in HCT116, HL60, and CEM cells) revealed that Bax redistributed to the mitochondrial fraction from the cytosol on exposure to H2O2, which could be significantly blocked by the H2O2 scavenger catalase. 7872 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS Fig. 5. Apoptosis and Bax translocation induced by C1 are mediated by intracellular H2O2. HL60 cells (1 ϫ 106/mL) were exposed to 50 ␮g/mL C1 in the presence or absence of 1000 units/mL catalase for to hours (for intracellular reactive oxygen species), 12 hours (for caspase activation and cytochrome c translocation), or 18 hours (for DNA fragmentation and cell survival). In A, intracellular reactive oxygen species was detected by flow cytometry using H2O2-specific probe DCHF-DA (a), lucigenin-based chemiluminescence assay (b), or flow cytometry using the fluorescent probe hydroethidine (c) as described in Materials and Methods. In B, cell survival was assessed by the MTT assay, and the mean Ϯ SD of three independent experiments is shown. Statistical analysis of data was performed using the paired t test. In C, PI staining was used to assess subdiploid DNA (sub-G1 fraction). In D, processed caspase was assayed in C1-treated cells by staining with antiactive caspase and analyzed by flow cytometry. In E, poly(ADP-ribose) polymerase cleavage in lysates was determined by Western blot using anti-poly(ADP-ribose) polymerase as described in Materials and Methods [MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; PI, propidium iodide]. One could always argue that the changes elicited on exposure of cells to exogenous H2O2 may not have physiologic relevance, because such high levels of intracellular H2O2 are rarely observed in living cells. Therefore, to present a more real-life situation, we exploited the ability of certain anticancer drugs to increase intracellular production of reactive oxygen species, specifically H2O2 (5). Indeed, exposure of HCT116 Baxϩ/Ϫ or HL60 cells to a novel anticancer compound C1 resulted in an increase in intracellular H2O2 and translocation of Bax to the mitochondria. This translocation of Bax was inhibited by catalase, thus establishing the critical role of intracellular H2O2 in mitochondrial recruitment during drug-induced apoptosis of tumor cells. Recruitment of Bax to the mitochondria during apoptotic signaling has been linked to the activation of upstream caspase and caspase 8-mediated cleavage of the proapoptotic protein Bid (40). This is particularly true on ligation of death receptors, such as CD95 (Apo1/ Fas). Incidentally, H2O2 and anticancer drugs have been shown to up-regulate the expression of the CD95 receptor or its ligand (CD95L) in some systems (41, 42). In these systems, blocking receptor signaling or downstream caspase activation could abrogate Bax translocation and, consequently, the death signaling circuitry downstream of the mitochondria. Interestingly, in our study, neither of the stimuli triggered up-regulation of the CD95 receptor (data not shown), and inhibition of caspase activation had no effect on the mitochondrial translocation of Bax. In addition, the relative lack of caspase activation and absence of downstream Bid cleavage provide additional evidence in favor of a mechanism for signaling Bax to the mitochondria that is either parallel to or independent of death receptor and/or caspase activation. A similar mechanism of caspase-independent conformational change of Bax on triggering apoptosis has been reported previously (43). Recent findings also implicate ceramide production in response to activation of the membrane sphingomyelinase in signaling Bax to the mitochondria (44, 45). Ceramide production is observed in a variety of apoptotic models, including drug-induced apoptosis (45, 46), and in response to H2O2 in tumor cells (3). However, inhibiting ceramide synthesis in our system did not affect the subcellular localization of Bax triggered by C1 or H2O2 (data not shown). Collectively, these data indicate that Bax translocation triggered in tumor cells during drug (C1)-induced apoptosis was a direct result of intracellular H2O2 production, independent of the upstream caspase or ceramide pathways. Corroborating these findings is a recent study suggesting glutathione-dependent activation of Bax by oxidative stress in HeLa cells stably transfected with the cystic fibrosis transmembrane conductance regulator (47). Drug-Induced Bax Translocation Is Dependent on H2O2-Mediated Cytosolic Acidification. Cytosolic acidification is an early event in apoptosis and provides an intracellular milieu permissive for efficient death execution. In this regard, exposure of cells to H2O2 or 7873 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS Fig. 6. H2O2-dependent Bax translocation and activation during drug-induced apoptosis in tumor cells. In A, HL60 cells (1 ϫ 106/mL) were exposed to 50 ␮g/mL C1 for and hours or CICCP (100 ␮mol/L for hour), and the change in ⌬␺m was assessed by flow cytometry using DiOC6 as described in Materials and Methods. In B, cytosolic fractions from cells after 12-hour incubation with C1 in the presence or absence of catalase were subjected to Western blot analysis for cytochrome c translocation. In C, caspase activity was assayed using a fluorogenic substrate (LEHD-fmk) and shown as fold increase (ϫ increase) over that of untreated cells. Data are mean Ϯ SD of three independent experiments, and the significance was calculated by the paired t test. D, mitochondrial and cytosolic fractions from HL60 cells (2.5 ϫ 107) after exposure to 50 ␮g/mL C1 for 12 hours in the presence or absence of 1,000 units/mL catalase or ZVAD (100 ␮mol/L) were subjected to Western blot analysis using anti-Bax as described in Materials and Methods. In E, Bax dimerization/multimerization in HL60 cell lysates was determined by 10% native gel electrophoresis and Western blot analysis using anti-Bax (6A7). ‫ ء‬and ‫ءء‬, dimers and multimers of Bax. drugs that trigger intracellular increase in H2O2 results in a significant drop in cytosolic pH (5). Accordingly, signals that inhibit apoptotic acidification impede death signaling as demonstrated in our recent study (37). Our results provide strong evidence that the link between H2O2 and Bax translocation could be the drop in cytosolic pH brought about by exposure of cells to exogenous H2O2 or endogenous production of H2O2 on drug exposure. Not only did inhibition of acidification triggered by H2O2 or C1 result in a significant reduction in mitochondrial localization of Bax, but also, most interestingly, clamping cytosolic pH to a more acidic range (ϳ7.1) directly induced Bax translocation in tumor cells. Because caspase inhibitors had no effect in our system, these results point to a direct effect of an acidic intracellular milieu in trafficking Bax from the cytosol to the mitochondria. It is interesting to note that one of the first studies to report a relationship between cytosolic pH and Bax translocation had concluded to the contrary, i.e., pH increase was shown to facilitate Bax translocation (48). However, in our system, the absence of any initial increase in cytosolic pH downstream of H2O2 or C1 even as early as 30 minutes after treatment (data not shown), fact that inhibition of pH drop abrogated Bax translocation, and direct effect of acidic intracellular milieu on Bax translocation provide strong evidence that acidic pH is a direct signal for Bax translocation even in the absence of a trigger. Furthermore, this phenomenon of H2O2-mediated, acidification-induced Bax translocation may not be exclusive to C1, because in our earlier study, we demonstrated the ability of commonly used chemotherapeutic drugs vincristine and daunorubicin to trigger an early increase in intracellular H2O2 (37). Interestingly, these compounds also induce cytosolic acidification and Bax-dependent apoptosis in tumor cells.5 Given these observations, it is highly likely that acidification-induced Bax activation may be a common mechanism during drug-induced apoptosis in tumor cells. Indeed, our findings linking an acidic milieu to Bax translocation are in agreement with a recent study demonstrating a relationship between cytosolic acidification and Bax translocation during staurosporine and tumor necrosis factor-induced apoptosis (49). Changes in pH affect the conformation of Bax, and a change in conformation facilitates Bax translocation to the mitochondria. In this regard, both positively and negatively charged residues contribute to the pH dependence of Bax conformation (48). It is plausible that similar to the effect of an alkaline pH, a considerable acidic shift in the pH could also induce a conformational change in Bax, thus making it more amenable for membrane insertion. 7874 S. Pervaiz et al., unpublished data. H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS Fig. 7. Drug-induced Bax translocation is dependent on H2O2-mediated intracellular acidification. In A, HL60 cells (1 ϫ 106) were exposed to 50 ␮g/mL or C1 or 100 ␮mol/L H2O2 for hours in the presence or absence of 1000 units/mL catalase, and cytosolic pH was measured with BCECF-AM as described in Materials and Methods. In C, cells were preincubated for hours with RSV (4 and ␮mol/L), followed by exposure to C1 and H2O2 for hours, and cytosolic pH was determined as described above. In C, mitochondrial and cytosolic fractions from HL60 (2.5 ϫ 107) or CEM (D) cells after exposure to 50 ␮g/mL of C1 or 100 ␮M H2O2 for 12 hours with or without preincubation with RSV (4 – ␮mol/L) were subjected to Western blot analysis using anti-Bax as described in Materials and Methods. In E, HL60 cells were exposed to ␮mol/L RSV for –24 hours, and lysates were analyzed by Western blotting using anti-Bcl-2 or anti-Bax antibodies. (RSV, resveratrol) Fig. 8. Enforced acidification triggers Bax translocation. HL60 cells were treated with – 40 ␮mol/L methylamiloride for (A), 12 (B), or 18 (C) hours. Cytosolic pH was determined by BCECF-AM loading, Bax translocation by Western blotting, and cell survival by the MTT assay, as described in Materials and Methods. In D, HL60 cells were treated with 25 ␮mol/L methylamiloride for to 12 hours, and lysates were analyzed by Western blotting using anti-Bcl-2 or anti-Bax antibodies [MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5diphenyltetrazolium bromide]. 7875 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS Fig. 9. Mitochondrial translocation of Bax results in additional increases in intracellular H2O2. In A, HCT116Baxϩ/Ϫ and BaxϪ/Ϫ cells (1 ϫ 106) were exposed to H2O2 (100 ␮mol/L), and intracellular levels of H2O2 were measured at 30 minutes, 1, 6, and 12 hours as described in Materials and Methods. In B, mean fluorescence intensity indicative of the mean intracellular H2O2 levels from three independent experiments is plotted to demonstrate the increase in intracellular H2O2 in Baxϩ/Ϫ cells. In C, HCT116 Baxϩ/Ϫ and BaxϪ/Ϫ cells were incubated with H2O2 for 15 minutes and loaded with the fluorescent probe hydroethidine. Events (Ն10,000) were analyzed by flow cytometry. In D, Baxϩ/Ϫ cells (1 ϫ 107) were incubated with 100 ␮mol/L H2O2 for 1, 6, and 12 hours, followed by cell fractionation and Western blot analysis using anti-Bax as described in Materials and Methods. Indeed, dimerization/multimerization of Bax in cells treated with C1 or H2O2 provide evidence to support conformational change in Bax in response to these apoptotic stimuli. Furthermore, considering the ability of drugs such as C1 to trigger early increases in intracellular H2O2, it is possible that the presence of scavenger catalase may have a more longer and sustained inhibitory effect on drug-induced Bax translocation. H2O2 Mitochondrial Bax- H2O2: Reactive Oxygen SpeciesDependent Reactive Oxygen Species Production. Our data provide a novel mechanism for drug-induced translocation of Bax and argue in Fig. 10. Intracellular H2O2 production by C1 is upstream of Bax.HCT116 Baxϩ/Ϫ (A). In B, BaxϪ/Ϫ cells were incubated with 50 ␮g/mL for 15 minutes, followed by loading of cells separately with DCHF-DA or hydroethidine for intracellular reactive oxygen species detection. Cells were analyzed by flow cytometry as described in Materials and Methods. 7876 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS favor of an upstream role for H2O2-mediated cytosolic acidification. Earlier studies have implicated superoxide anion and H2O2 as probable species responsible for mitochondrial damage, such as lipid peroxidation of the mitochondrial membranes and induction of mitochondrial permeability transition (MPT) (50), changes that could explain the increase in the outer membrane permeability and leakage of pro-apoptotic proteins to the cytosol. However, most of these studies seem to imply that the mitochondrial burst of H2O2 is likely to be a downstream effector mechanism for the execution signal. Our data argue strongly in favor of a two-hit mechanism for drug-induced increases in intracellular H2O2. The initial increase (first hit) in intracellular H2O2 on drug treatment of tumor cells leads to the targeting of Bax to the mitochondria in a caspase-independent manner, thus resulting in the recruitment of the mitochondrial death pathway. Bax translocation can then bring about mitochondrial changes, such as induction of MPT, formation of a functional channel in the outer membrane, and as our data suggest, serve as a stimulus for the next burst of H2O2 (second hit) from the mitochondria. This could lead to peroxidative damage of mitochondrial lipids, such as cardiolipin, and facilitate the egress of cytochrome c, Smac/Diablo, or apoptosis inducing factor (AIF). This phenomenon of reactive oxygen species-induced reactive oxygen species production has been described before as a probable mechanism for the induction of MPT, and more recently, in a model of nerve growth factor-deprived neuronal cell death, Bax insertion has been proposed to induce reactive oxygen species burst, strong enough to trigger the release of cytochrome c (51). It should be pointed out that the initial early increase (within 15 minutes of exposure) in intracellular H2O2 occurs independent of Bax (both Baxϩ/Ϫ and BaxϪ/Ϫ cells); however, more importantly, the subsequent (second) increase is entirely dependent on Bax and its translocation to the mitochondria. One probable mechanism for Baxinduced H2O2 production could be that Bax translocation leads to cytochrome c release, which depletes the mitochondrial electron transport chain with the resultant leakage of electrons to molecular oxygen to form O2Ϫ, which is then dismutated by the mitochondrial superoxide dismutase to H2O2. Taken together, our data demonstrate a novel mechanism for mitochondrial recruitment of the proapoptotic protein Bax and provide a mechanistic explanation to connect extramitochondrial death circuitry to the mitochondria, the point of convergence for effective druginduced death signals. REFERENCES 1. Green DR, Kroemer G. The pathophysiology of mitochondrial cell death. Science 2004;305:626 –9. 2. Fleury C, Mignotte B, Vayssiere JL. Mitochondrial reactive oxygen species in cell death signaling. Biochimie 2002;84:131– 41. 3. Mansat-de Mas V, Bezombes C, Quillet-Mary A et al. Implication of radical oxygen species in ceramide generation, c-Jun N-terminal kinase activation and apoptosis induced by daunorubicin. Mol Pharmacol 1999;56:867–74. 4. Simizu S, Umezawa K, Takada M, Arber N, Imoto M. Induction of hydrogen peroxide production and Bax expression by caspase-3(-like) proteases in tyrosine kinase inhibitor-induced apoptosis in human small cell lung carcinoma cells. Exp Cell Res 1998;238:197–203. 5. Hirpara JL, Clement MV, Pervaiz S. Intracellular acidification triggered by mitochondrial-derived hydrogen peroxide is an effector mechanism for drug-induced apoptosis in tumor cells. J Biol Chem 2001;276:514 –21. 6. Clement MV, Ponton A, Pervaiz S. Apoptosis induced by hydrogen peroxide is mediated by decreased superoxide anion concentration and reduction of intracellular milieu. FEBS Lett 1998;440:13– 8. 7. Childs AC, Phaneuf SL, Dirks AJ, Phillips T, Leeuwenburgh C. Doxorubicin treatment in vivo causes cytochrome C release and cardiomyocyte apoptosis, as well as increased mitochondrial efficiency, superoxide dismutase activity, and Bcl-2:Bax ratio. Cancer Res 2002;62:4592– 8. 8. Quillet-Mary A, Jaffrezou JP, Mansat V, Bordier C, Naval J, Laurent G. Implication of mitochondrial hydrogen peroxide generation in ceramide-induced apoptosis. J Biol Chem 1997;272:21388 –95. 9. Clement MV, Pervaiz S. Reactive oxygen intermediates regulate cellular response to apoptotic stimuli: an hypothesis. Free Radic Res 1999;30:247–52. 10. Pervaiz S, Ramalingam JK, Hirpara JL, Clement MV. Superoxide anion inhibits drug-induced tumor cell death. FEBS Lett 1999;459:343– 8. 11. Clement MV, Pervaiz S. Intracellular superoxide and hydrogen peroxide concentrations: a critical balance that determines survival or death. Redox Rep 2001;6:211– 4. 12. Esposti MD, Hatzinisiriou I, McLennan H, Ralph S. Bcl-2 and mitochondrial oxygen radicals. New approaches with reactive oxygen species-sensitive probes. J Biol Chem 1999;274:29831–7. 13. Distelhorst CW, Lam M, McCormick TS. Bcl-2 inhibits hydrogen peroxide-induced ER Ca2ϩ pool depletion. Oncogene 1996;12:2051–5. 14. Yamakawa H, Ito Y, Naganawa T et al. Activation of caspase-9 and -3 during H2O2-induced apoptosis of PC12 cells independent of ceramide formation. Neurol Res 2000;22:556 – 64. 15. Naderi J, Hung M, Pandey S. Oxidative stress-induced apoptosis in dividing fibroblasts involves activation of p38 MAP kinase and over-expression of Bax: resistance of quiescent cells to oxidative stress. Apoptosis 2003;8:91–100. 16. Harris CA, Johnson EM Jr. BH3-only Bcl-2 family members are coordinately regulated by the JNK pathway and require Bax to induce apoptosis in neurons. J Biol Chem 2001;276:37754 – 60. 17. Zong WX, Lindsten T, Ross AJ, MacGregor GR, Thompson CB. BH3-only proteins that bind pro-survival Bcl-2 family members fail to induce apoptosis in the absence of Bax and Bak. Genes Dev 2001;15:1481– 6. 18. Simonen M, Keller H, Heim J. The BH3 domain of Bax is sufficient for interaction of Bax with itself and with other family members and it is required for induction of apoptosis. Eur J Biochem 1997;249:85–91. 19. Crompton M. Bax, Bid and the permeabilization of the mitochondrial outer membrane in apoptosis. Curr Opin Cell Biol 2000;12:414 –9. 20. Strasser A, O’Connor L, Dixit VM. Apoptosis signaling. Annu Rev Biochem 2000; 69:217– 45. 21. Scaffidi C, Kirchhoff S, Krammer PH, Peter ME. Apoptosis signaling in lymphocytes. Curr Opin Immunol 1999;11:277– 85. 22. Murphy KM, Streips UN, Lock RB. Bax membrane insertion during Fas(CD95)induced apoptosis precedes cytochrome c release and is inhibited by Bcl-2. Oncogene 1999;18:5991–9. 23. Nechushtan A, Smith CL, Lamensdorf I, Yoon SH, Youle RJ. Bax and Bak coalesce into novel mitochondria-associated clusters during apoptosis. J Cell Biol 2001;153: 1265–76. 24. Marzo I, Brenner C, Zamzami N et al. Bax and adenine nucleotide translocator cooperate in the mitochondrial control of apoptosis. Science 1998;281:2027–31. 25. De Giorgi F, Lartigue L, Bauer MK et al. The permeability transition pore signals apoptosis by directing Bax translocation and multimerization. FASEB J 2002;16: 607–9. 26. Jia L, Patwari Y, Srinivasula SM et al. Bax translocation is crucial for the sensitivity of leukaemic cells to etoposide-induced apoptosis. Oncogene 2001;20:4817–26. 27. Putcha GV, Deshmukh M, Johnson EM Jr. BAX translocation is a critical event in neuronal apoptosis: regulation by neuroprotectants, BCL-2, and caspases. J Neurosci 1999;19:7476 – 85. 28. Kobayashi T, Sawa H, Morikawa J, Zhang W, Shiku H. Bax induction activates apoptotic cascade via mitochondrial cytochrome c release and Bax overexpression enhances apoptosis induced by chemotherapeutic agents in DLD-1 colon cancer cells. Jpn J Cancer Res 2000;91:1264 – 8. 29. Zhang L, Yu J, Park BH, Kinzler KW, Vogelstein B. Role of BAX in the apoptotic response to anticancer agents. Science 2000;290:989 –92. 30. Pervaiz S, Seyed MA, Hirpara JL, Clement MV, Loh KW. Purified photoproducts of merocyanine 540 trigger cytochrome C release and caspase 8-dependent apoptosis in human leukemia and melanoma cells. Blood 1999;93:4096 –108. 31. Hirpara JL, Seyed MA, Loh KW, Dong H, Kini RM, Pervaiz S. Induction of mitochondrial permeability transition and cytochrome C release in the absence of caspase activation is insufficient for effective apoptosis in human leukemia cells. Blood 2000;95:1773– 80. 32. Pervaiz S, Clement MV. Hydrogen peroxide-induced apoptosis: oxidative or reductive stress? Methods Enzymol 2002;352:150 –9. 33. Gyllenhammar H. Lucigenin chemiluminescence in the assessment of neutrophil superoxide production. J Immunol Meth 1987;97:209 –20. 34. Li Y, Kuppusamy P, Roubaud V, Zweier JL, Trush MA. Validation of lucigenin (bis-N-methylacridinium) as a chemilumigenic probe for detectiing superoxide anion radical production by enzymatic and cellular systems. J Biol Chem 1998;273: 2015–23. 35. Soucie EL, Annis MG, Sedivy J et al. Myc potentiates apoptosis by stimulating Bax activity at the mitochondria. Mol Cell Biol 2001;21:4725–36. 36. Pervaiz S, Cao J, Chao OS, Chin YY, Clement MV. Activation of the RacGTPase inhibits apoptosis in human tumor cells. Oncogene 2001;20:6263– 8. 37. Ahmad KA, Clement MV, Hanif IM, Pervaiz S. Resveratrol inhibits drug-induced apoptosis in human leukemia cells by creating an intracellular milieu nonpermissive for death execution. Cancer Res 2004;64:1452–9. 38. Nakamura T, Sakamoto K. Reactive oxygen species up-regulates cyclooxygenase-2, p53, and Bax mRNA expression in bovine luteal cells. Biochem Biophys Res Commun 2001;284:203–10. 39. Burdon RH. Superoxide and hydrogen peroxide in relation to mammalian cell proliferation. Free Radic Biol Med 1995;18:775–94. 40. Desagher S, Osen-Sand A, Nichols A et al. Bid-induced conformational change of Bax is responsible for mitochondrial cytochrome c release during apoptosis. J Cell Biol 1999;144:891–901. 7877 H2O2 SIGNALS BAX TRANSLOCATION IN TUMOR CELLS 41. Hug H, Strand S, Grambihler A et al. Reactive oxygen intermediates are involved in the induction of CD95 ligand mRNA expression by cytostatic drugs in hepatoma cells. J Biol Chem 1997;272:28191–3. 42. Suhara T, Fukuo K, Sugimoto T et al. Hydrogen peroxide induces up-regulation of Fas in human endothelial cells. J Immunol 1998;160:4042–7. 43. Bellosillo B, Villamor N, Lopez-Guillermo A et al. Spontaneous and drug-induced apoptosis is mediated by conformational changes of Bax and Bak in B-cell chronic lymphocytic leukemia. Blood 2002;100:1810 – 6. 44. von Haefen C, Wieder T, Gillissen B et al Ceramide induces mitochondrial activation and apoptosis via a Bax-dependent pathway in human carcinoma cells. Oncogene 2002;21:4009 –19. 45. Sawada M, Nakashima S, Banno Y et al. Ordering of ceramide formation, caspase activation, and Bax/Bcl-2 expression during etoposide-induced apoptosis in C6 glioma cells. Cell Death Differ 2000;7:761–72. 46. Delpy E, Hatem SN, Andrieu N et al. Doxorubicin induces slow ceramide accumulation and late apoptosis in cultured adult rat ventricular myocytes. Cardiovasc Res 1999;43:398 – 407. 47. Jungas T, Motta I, Duffieux F, Fanen P, Stoven V, Ojcius DM. Glutathione levels and BAX activation during apoptosis due to oxidative stress in cells expressing wild-type and mutant cystic fibrosis transmembrane conductance regulator. J Biol Chem 2002; 277:27912– 8. 48. Khaled AR, Kim K, Hofmeister R, Muegge K, Durum SK. Withdrawal of IL-7 induces Bax translocation from cytosol to mitochondria through a rise in intracellular pH. Proc Natl Acad Sci USA 1999;96:14476 – 81. 49. Tafani M, Cohn JA, Karpinich NO, Rothman RJ, Russo MA, Farber JL. Regulation of intracellular pH mediates Bax activation in HeLa cells treated with staurosporine or tumor necrosis factor-alpha. J Biol Chem 2002;277:49569 –76. 50. Asumendi A, Morales MC, Alvarez A, Arechaga J, Perez-Yarza G. Implication of mitochondria-derived ROS and cardiolipin peroxidation in N-(4-hydroxyphenyl)retinamide-induced apoptosis. Br J Cancer 2002;86:1951– 6. 51. Kirkland RA, Windelborn JA, Kasprzak JM, Franklin JL. A Bax-induced pro-oxidant state is critical for cytochrome c release during programmed neuronal death. J Neurosci 2002;22:6480 –90. 7878 [...]... Spontaneous and drug- induced apoptosis is mediated by conformational changes of Bax and Bak in B -cell chronic lymphocytic leukemia Blood 20 02; 100:1810 – 6 44 von Haefen C, Wieder T, Gillissen B et al Ceramide induces mitochondrial activation and apoptosis via a Bax-dependent pathway in human carcinoma cells Oncogene 20 02; 21:4009 –19 45 Sawada M, Nakashima S, Banno Y et al Ordering of ceramide formation,... signals apoptosis by directing Bax translocation and multimerization FASEB J 20 02; 16: 607–9 26 Jia L, Patwari Y, Srinivasula SM et al Bax translocation is crucial for the sensitivity of leukaemic cells to etoposide -induced apoptosis Oncogene 20 01 ;20 :4817 26 27 Putcha GV, Deshmukh M, Johnson EM Jr BAX translocation is a critical event in neuronal apoptosis: regulation by neuroprotectants, BCL -2, and caspases... membrane permeability and leakage of pro-apoptotic proteins to the cytosol However, most of these studies seem to imply that the mitochondrial burst of H2O2 is likely to be a downstream effector mechanism for the execution signal Our data argue strongly in favor of a two-hit mechanism for drug- induced increases in intracellular H2O2 The initial increase (first hit) in intracellular H2O2 on drug treatment... caspase-3(-like) proteases in tyrosine kinase inhibitor -induced apoptosis in human small cell lung carcinoma cells Exp Cell Res 1998 ;23 8:197 20 3 5 Hirpara JL, Clement MV, Pervaiz S Intracellular acidification triggered by mitochondrial-derived hydrogen peroxide is an effector mechanism for drug- induced apoptosis in tumor cells J Biol Chem 20 01 ;27 6:514 21 6 Clement MV, Ponton A, Pervaiz S Apoptosis. .. NO, Rothman RJ, Russo MA, Farber JL Regulation of intracellular pH mediates Bax activation in HeLa cells treated with staurosporine or tumor necrosis factor-alpha J Biol Chem 20 02; 277:49569 –76 50 Asumendi A, Morales MC, Alvarez A, Arechaga J, Perez-Yarza G Implication of mitochondria-derived ROS and cardiolipin peroxidation in N-(4-hydroxyphenyl)retinamide -induced apoptosis Br J Cancer 20 02; 86:1951–... Activation of the RacGTPase inhibits apoptosis in human tumor cells Oncogene 20 01 ;20 : 626 3– 8 37 Ahmad KA, Clement MV, Hanif IM, Pervaiz S Resveratrol inhibits drug- induced apoptosis in human leukemia cells by creating an intracellular milieu nonpermissive for death execution Cancer Res 20 04;64:14 52 9 38 Nakamura T, Sakamoto K Reactive oxygen species up-regulates cyclooxygenase -2, p53, and Bax mRNA expression... 1999 ;27 4 :29 831–7 13 Distelhorst CW, Lam M, McCormick TS Bcl -2 inhibits hydrogen peroxide -induced ER Ca2ϩ pool depletion Oncogene 1996; 12: 2051–5 14 Yamakawa H, Ito Y, Naganawa T et al Activation of caspase-9 and -3 during H2O2 -induced apoptosis of PC 12 cells independent of ceramide formation Neurol Res 20 00 ;22 :556 – 64 15 Naderi J, Hung M, Pandey S Oxidative stress -induced apoptosis in dividing fibroblasts... activation during apoptosis due to oxidative stress in cells expressing wild-type and mutant cystic fibrosis transmembrane conductance regulator J Biol Chem 20 02; 27 7 :27 9 12 8 48 Khaled AR, Kim K, Hofmeister R, Muegge K, Durum SK Withdrawal of IL-7 induces Bax translocation from cytosol to mitochondria through a rise in intracellular pH Proc Natl Acad Sci USA 1999;96:14476 – 81 49 Tafani M, Cohn JA, Karpinich... 1999;18:5991–9 23 Nechushtan A, Smith CL, Lamensdorf I, Yoon SH, Youle RJ Bax and Bak coalesce into novel mitochondria-associated clusters during apoptosis J Cell Biol 20 01;153: 126 5–76 24 Marzo I, Brenner C, Zamzami N et al Bax and adenine nucleotide translocator cooperate in the mitochondrial control of apoptosis Science 1998 ;28 1 :20 27–31 25 De Giorgi F, Lartigue L, Bauer MK et al The permeability transition... Cancer Res 20 02; 62: 45 92 8 8 Quillet-Mary A, Jaffrezou JP, Mansat V, Bordier C, Naval J, Laurent G Implication of mitochondrial hydrogen peroxide generation in ceramide -induced apoptosis J Biol Chem 1997 ;27 2 :21 388 –95 9 Clement MV, Pervaiz S Reactive oxygen intermediates regulate cellular response to apoptotic stimuli: an hypothesis Free Radic Res 1999;30 :24 7– 52 10 Pervaiz S, Ramalingam JK, Hirpara JL, . in intracellular H 2 O 2 and translocation of Bax to the mitochondria. This translocation of Bax was inhibited by catalase, thus establishing the critical role of intracellular H 2 O 2 in mito - chondrial. Bax in H 2 O 2 -mediated apoptosis, and (2) to investigate whether the intracellular increase in H 2 O 2 was a signal for Bax translocation during drug- induced apoptosis of tumor cells. Here,. in favor of a two-hit mechanism for drug- induced increases in intracellular H 2 O 2 . The initial increase (first hit) in intracellular H 2 O 2 on drug treatment of tumor cells leads to the targeting

Ngày đăng: 16/09/2015, 17:11

Tài liệu cùng người dùng

  • Đang cập nhật ...

Tài liệu liên quan