<<

[CANCER RESEARCH 61, 1038–1044, February 1, 2001] Anticancer Drugs Induce Increased Mitochondrial c Expression That Precedes Cell Death1

Jose´A.Sa´nchez-Alca´zar, Alexey Khodjakov, and Erasmus Schneider2 Wadsworth Center, New York State Department of Health [J. A. S-A., A. K., E. S.], and Department of Biomedical Sciences, School of Public Health, University at Albany [A. K., E. S.], Albany, New York 12201

ABSTRACT The goal of the present study was to characterize the biochemical and molecular events occurring in the various stages leading to cell Recent studies have demonstrated that cytochrome c plays an impor- death, using human breast epithelial cells as a model. To this end we tant role in cell death. In the present study, we report that teniposide and took advantage of the fact that, during , cells grown as various other chemotherapeutic agents induced a dose-dependent increase in the expression of the mitochondrial respiratory chain proteins cyto- monolayers detach from the surface of the culture flask and float in chrome c, subunits I and IV of , and the free radical the medium. The standard procedure of collecting the medium, scavenging manganous dismutase. The teniposide- trypsinizing the attached cells and then combining both populations induced increase of cytochrome c was inhibited by cycloheximide, indi- (floating cells and adherent cells) is often used for quantitative anal- cating new protein synthesis. Elevated cytochrome c levels were associated ysis of apoptosis. However, this approach may bias biochemical with enhanced cytochrome c oxidase-dependent uptake using studies carried out on the bulk cell population and may result in TMPD/ascorbate as the electron donor, suggesting that the newly synthe- misinterpretation of the real events occurring in the living population sized proteins were functional. Cytochrome c was released into the cyto- before death. Therefore, in the present study we separately collected plasm only after maximal levels had been reached in the mitochondria, and studied attached and floating cells to characterize the biochemi- but there was no concomitant decrease in mitochondrial membrane po- cal/molecular events, whereas we combined both populations to quan- tential or activation. Our results suggest that the increase in mitochondrial protein expression may play a role in the early cellular tify apoptosis. Using this approach, we showed that teniposide and defense against anticancer drugs. other chemotherapeutic agents increased the levels of proteins of the mitochondrial respiratory chain, such as cytochrome c, and subunits I and IV of COX. This up-regulation of mitochondrial proteins pre- INTRODUCTION ⌬⌿ ceded cytochrome c release, drop of m and caspase activation. Recent studies have shown that alterations of mitochondrial func- tions such as PT3 play a major role in the apoptotic process induced MATERIALS AND METHODS by chemotherapeutic agents (1, 2). Mitochondria undergoing PT re- Reagents. All reagents and drugs were obtained from Sigma (St. Louis, lease apoptogenic proteins such as cytochrome c or the apoptosis- MO), unless otherwise indicated. Antibodies against COX subunits I and IV inducing factor from the mitochondrial intermembrane space into the were from Molecular Probes (Eugene, OR); against cytochrome c (clone cytosol, where they can activate and endonucleases (3, 4). 7H8.2C12) and caspases 3 and 9 from PharMingen (San Diego, CA); against The release of mitochondrial cytochrome c in particular is a critical PARP from Roche Biochemicals (Indianapolis, IN); against GAPDH (clone step in the apoptotic as well as the necrotic process (5–7). However, 6C5) from Research Diagnostics, Inc. (Flanders, NJ); and against citrate the mechanism(s) by which cytochrome c is released from mitochon- synthase from Chemicon International, Inc. (Temecula, CA). Anti-VDAC dria remains unclear. It has been proposed that PT would allow the antibody was a gift from Dr. C.A. Mannella, (Wadsworth Center, Albany, NY) passive release of cytochrome c and other caspase-activating factor(s) and anti-manganous superoxide dismutase antibody was a gift from Dr. J. A. from the mitochondrial intermembrane space into the cytosol (8, 9). Melendez (Albany Medical College, Albany, NY). A complete cocktail of inhibitors was purchased from Roche Biochemicals (Indianapolis, IN). However, it has also been reported that in various cell types the Cell Culture. Human breast carcinoma MDA-MB-231 were cultured in release of cytochrome c occurs before or even in the absence of a IMEM medium supplemented with penicillin, streptomycin, glutamate (2 mM), change in mitochondrial permeability, suggesting that this release can and 10% fetal bovine serum at 37°C in a humidified atmosphere of 5% CO2 in air. occur by mechanisms other than the opening of the PT pore (10–13). Separation of Attached and Floating Cells. During the process of cell For example, overexpression of the pro-apoptotic protein Bax has death, MDA-MB-231 cells detached from the culture flask and floated in the been shown to trigger cytochrome c efflux from mitochondria and cell medium. We exploited this phenomenon as an easy way to separate attached/ death (14, 15), possibly via the formation of membrane channels, living cells from floating/dead cells. Thus, floating cells were collected from whereas the redistribution of cytochrome c during apoptosis can be the culture supernatant by centrifugation (5 min at 1200 rpm), whereas the prevented by overexpression of the anti-apoptotic protein Bcl-2 (10, attached cells were harvested by trysinization. Viability of floating and at- 11). Thus, the exact sequence of events leading to the onset of cell tached cells was determined by PI staining and microscopic evaluation. Only samples of attached cells with Ͼ85% of viable cells were used for additional death remains unclear and may differ in different systems. studies. The floating cell population was 100% PI-positive. DNA Fragmentation Analysis. For DNA fragmentation analysis, 3 ϫ 106 Received 3/20/00; accepted 11/21/00. MDA-MB-231 cells were exposed to teniposide (10 ␮M) for 24, 48, or 72 h. The costs of publication of this article were defrayed in part by the payment of page Attached and floating cells were harvested and combined, washed with cold charges. This article must therefore be hereby marked advertisement in accordance with ␮ 18 U.S.C. Section 1734 solely to indicate this fact. PBS, and resuspended in 400 l of hypotonic lysis buffer A [10 mM Tris/HCl 1 This study was supported by NIH Grants CA72455 and CA25933. (pH 7.5), 1 mM EDTA, and 0.2% Triton X-100]. The cell lysates were 2 To whom requests for reprints should be addressed, at Wadsworth Center, Empire centrifuged at 13,000 rpm for 15 min in a microcentrifuge. The supernatant State Plaza, Albany, NY 12201. Phone: (518) 474-2088; Fax: (518) 474-1850; E-mail: (350 ␮l) was then incubated with 106 ␮l of lysis buffer B [150 mM NaCl, 10 [email protected]. 3 The abbreviations used are: PT, permeability transition; COX, cytochrome c oxidase; mM Tris-HCl (pH 8.0), 40 mM EDTA, 1% SDS, and 0.2 mg/ml proteinase K, GAPDH, glyceraldehyde-3-phosphate dehydrogenase; TMPD, tetramethyl-p-phenylene- final concentrations] for4hat37°C. The DNA was extracted with phenol/ diamine; JC-1, 5,5Ј,6,6Ј-tetrachloro-1,1Ј,3,3Ј-tetraethylbenzimidazolecarbocyanine iodide; chloroform/isoamyl alcohol (25:25:1, v/v/v) and ethanol precipitated for 12–18 PARP, poly(ADP-ribose) polymerase; PI, propidium iodide; VDAC, voltage-dependent h. After centrifugation for 5 min at 13,000 rpm and 4°C, the DNA pellet was anion channel; DRB, 5,6-dichlorobenzimidazole riboside; IMEM, improved minimum ␮ ␮ essential medium (Richter’s modification); CCCP, carbonyl cyanide m-chlorophenyl- washed with 500 l of 70% ethanol and resuspended in 15 lof10mM hydrazone. Tris/HCl (pH 8.5), 1 mM EDTA containing 50 ␮g/ml RNase, and incubated for 1038

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2001 American Association for Cancer Research. MITOCHONDRIAL CYTOCHROME c EXPRESSION PRECEDES CELL DEATH

⌬⌿ 1 h at 37°C. Each DNA sample was then analyzed on a 1% agarose gel for each analysis. The ratio of FL2 versus FL1 was used to analyze m. containing 0.1 ␮g/ml ethidium bromide. The same amount of DNA, as as- Forward scatter was used to differentiate live from dead cells. sessed by spectrophotometric measurement, was loaded in each lane. A mix- Immunostaining and Microscopy. For immunostaining, MDA-MB-231 ture of HaeIII-digested ⌽X174 DNA and HindIII-digested ␭DNA was run as cells were seeded onto 18 ϫ 18 mm no. 1 glass coverslips and grown for size markers. DNA fragmentation was also measured by quantitation of hyp- 24–48 h in IMEM supplemented with 10% FBS. In some experiments, cells oploid nuclei after DNA staining with PI. After teniposide treatment and were treated with teniposide for 48 h. To differentiate between live and dead harvesting as above, cells were fixed with 70% cold ethanol at 4°C overnight. cells, the “Dead Red” reagent (Molecular Probes, Eugene, OR) was added to After centrifugation, the fixed cells were resuspended in 1 ml PI staining the cells 10 min before fixation. For immunostaining, cells were fixed in 3.8% ␮ solution (5 g/ml PI in 0.1% sodium citrate and 0.1% Triton X-100) and paraformaldehyde for 5 min at room temperature, permeabilized in 0.1% incubated for 30 min at 4°C. Stained nuclei were analyzed on a FACScan saponin for 5 min, and stained with anti-cytochrome c antibody diluted at (Becton Dickinson Immunocytometry System, San Jose, CA). Hypoploid cells 1:100. FITC-conjugated goat antimouse secondary antibody (Sigma, St. Louis, appear as a sub-G peak. 1 MO) was used at 1:100. DNA was stained using Hoechst 33342 at 0.4 ␮g/ml. COX Activity in Whole Cells. COX activity was measured as described Preparations were mounted in FluoroGard antifade reagent (Bio-Rad, Hercu- (16). MDA-MB-231 cells were treated with teniposide (10 ␮M) for 24, 48, or 72 h. After the indicated time, the floating cells were removed. The attached les, CA) and analyzed using a Nikon Diaphot microscope, equipped with a QuadFluor epi-fluorescence attachment. Images were recorded with a Photo- cells were harvested and resuspended in respiration buffer [0.25 M sucrose, ϩ metrics PXL cooled charge-coupled device camera. 0.1% BSA, 10 mM MgCl2,10mM K HEPES, 5 mM KH2PO4 (pH 7.2)] at a final concentration of 4 ϫ 107 cells/ml. One-half ml of the cell suspension was Protein Determinations. Protein concentrations were determined by the injected into a chamber containing 3.5 ml of air-saturated respiration buffer Bradford assay (20). and1mM ADP at 37°C. The cells were permeabilized with digitonin (final concentration, 0.005%), and substrates and inhibitors were added in the fol- lowing order and final concentrations: (a) antimycin A, 50 nM;(b) ascorbate, 1mM; and (c) TMPD, 0.4 mM. Antimycin A was used to inhibit autologous mitochondrial electron transport. TMPD is an electron donor that reduces cytochrome c nonenzymatically. Therefore, when TMPD is used as a substrate,

changes in O2 uptake rates reflect changes in COX activity. Ascorbate was used to reduce TMPD. The oxygen concentration was calibrated with air- saturated buffer, assuming 390 ng-atoms of oxygen/ml of buffer. Rates of potassium cyanide-sensitive oxygen consumption are expressed as ng-atoms of oxygen/min/2 ϫ 107 cells. Western Blot Analysis of Cytosolic and Mitochondrial Fractions. Cy- tosolic and mitochondrial fractions were prepared as described (17). Floating and attached MDA-MB-231 cells were collected separately, washed twice with ice-cold PBS (pH 7.4), and resuspended in 600 ␮l of extraction buffer con- taining 220 mM mannitol, 68 mM sucrose, 50 mM PIPES-KOH (pH 7.4), 50 mM

KCl, 5 mM EGTA, 2 mM MgCl2,1mM DTT, and protease inhibitors. After 30 min incubation on ice, cells were homogenized with a Teflon homogenizer for 3 min at 300 rpm. Cell homogenates were centrifuged at 14,000 ϫ g for 15 min, and the cytosolic supernatants removed and stored at Ϫ70°C. The quality of the cytosolic fraction was routinely monitored by Western blotting for cytochrome oxidase subunit I as a marker of mitochondrial contamination, whereas the mitochondrial fraction was routinely monitored for dihydrofolate reductase as a marker of cytoplasmic contamination. The pellet containing the mitochondria was resuspended in extraction buffer and stored at Ϫ70°C. Twenty-five ␮g of cytosolic or mitochondrial proteins were separated on a 15% denaturing SDS-PAGE minigel. After protein transfer, the membrane was incubated with various primary antibodies as indicated for 1 h. Anti-cyto- chrome c, anti-COX subunits I and IV, anti-PARP, anti-caspases 3 and 9, anti-VDAC, and anti- antibodies were all diluted 1:1,000. Anti-GAPDH antibody was diluted 1:5,000. The membrane was then incu- bated with the appropriate secondary antibody coupled to horseradish perox- idase at 1:10,000 dilution. The specific protein complexes were identified by chemiluminescence using the “Supersignal” substrate reagent (Pierce, Rock- ford, IL). Measurement of Mitochondrial Membrane Potential by Flow Cytom- ⌬⌿ etry. Changes in the mitochondrial membrane potential m were analyzed using JC-1 (Molecular Probes, Inc., Eugene, OR). This cyanine dye accumu- ⌬⌿ lates in the mitochondrial matrix under the influence of the m and forms J-aggregates that have characteristic absorption and emission spectra (18). The ⌬⌿ JC-1 staining method is reported to provide more accurate estimates of m than DiOC6 (Refs. 3 and 19). Untreated controls and cells treated with teniposide (10 ␮M) for 24, 48, or 72 h were incubated in 0.4 ml of IMEM with Fig. 1. Time course of teniposide-induced DNA fragmentation in MDA-MB-231 cells. 0.5 ␮M JC-1 for 10 min. As a positive control for reduction of ⌬⌿ , control m Cells were treated with 10 ␮M teniposide and both attached and floating cells were cells were treated with the uncoupling agent CCCP (1 ␮M) before labeling with harvested together at 24, 48, or 72 h. A, cells with a hypodiploid DNA content (sub-G1) JC-1. Cell suspensions were prepared for flow cytometry, and the 488-nm line were detected by PI staining and flow cytometry. The numbers represent the means Ϯ SD of an argon ion laser was used for excitation. Red and green emitted fluores- from three independent experiments, whereas the histograms are representatives from one cence was collected through 585/42 (FL2) and 530/30-nm (FL1) bandpass of three experiments. B, internucleosomal DNA fragmentation was evaluated by agarose gel electrophoresis. Lane 1, DNA from the control cells. Lanes 2, 3, and 4, DNA from filters, respectively. Flow cytometry was performed on a Coulter Elite flow cells treated with teniposide for 24, 48, or 72 h, respectively. Lane 5, DNA molecular cytometer. After gating out small-sized debris, 10,000 events were collected weight markers. 1039

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2001 American Association for Cancer Research. MITOCHONDRIAL CYTOCHROME c EXPRESSION PRECEDES CELL DEATH

with teniposide demonstrated that cell death was accompanied by a

significant increase in the percentage of sub-G1 cells with a subdiploid DNA content (Fig. 1A). Furthermore, internucleosomal DNA degra- dation was also demonstrated by the appearance of DNA ladders (Fig. 1B). Both of these features are considered typical of apoptosis, sug- gesting that teniposide-treated MDA-MB-231 cells underwent a form of cell death characterized by features common to the apoptotic pathway. Teniposide Induces Increased Mitochondrial Cytochrome c Expression. Recent work on apoptosis and necrosis revealed that cytochrome c is released from the mitochondria into the cytoplasm during cell death (4–7). To examine whether cytochrome c release

Fig. 3. Cytochrome c and COX subunits I and IV levels in attached versus floating cells. Western blot analysis of cytosolic and mitochondrial extracts from MDA-MB-231 cells treated with 10 ␮M teniposide for 24, 48, or 72 h. Attached and floating cells were harvested separately, and cytosolic and mitochondrial fractions were prepared as de- scribed in “Materials and Methods.” Membrane blots were incubated with antibodies against the various proteins as indicated. The last right lane contains 10 ng of horse heart cytochrome c as a standard. Pictures are representative from one of three studies. Fig. 2. Effect of teniposide on cytochrome c and COX subunits I and IV levels in Cytosolic and mitochondrial gels and blots were processed in parallel and exposed for the mitochondria and the cytoplasm. A, Western blot analysis of cytosolic and mitochondrial same amount of time onto the same film. fractions from attached MDA-MB-231 cells treated with 10 ␮M teniposide for 24, 48, or 72 h. Attached cells were harvested, cytosolic and mitochondrial fractions were prepared, and proteins were separated by SDS-PAGE as described in “Materials and Methods.” Membrane blots were incubated with antibodies against the various proteins as indicated. The last right lane contained 10 ng of horse heart cytochrome c as a standard. Pictures are representative from one of three studies. Cytosolic and mitochondrial gels and blots were processed in parallel and exposed for the same amount of time onto the same film. B, densitometric quantitation of cytochrome c (——ࡗ——) and COX subunits I (—— Œ——) and IV (——f——) levels in mitochondria and of cytochrome c (...... ) in the cytosol after teniposide treatment. Results are expressed as means Ϯ SD from three independent experiments. C, dose response of the increase in mitochondrial protein levels. Cells were treated with increasing concentrations of teniposide as indicated for 48 h. Attached cells were harvested, and mitochondrial fractions were prepared. The levels of cytochrome c, COX I, COX IV, and citrate synthase were determined by Western blot analysis as described in “Materials and Methods.” Pictures are representative from one of three studies.

Statistical Analysis. All results are expressed as means Ϯ SD unless stated otherwise. The unpaired Student’s t test was used to evaluate the significance of differences between groups, accepting P Ͻ 0.05 as the level of significance.

RESULTS Fig. 4. Mitochondrial cytochrome c up-regulation is attributable to new protein Characterization of Teniposide-induced Cell Death in MDA- synthesis. MDA-MB-231 cells were treated for 24 h with 10 ␮M teniposide in the absence MB-231 Cells. In agreement with previous reports demonstrating that and presence of cycloheximide (0.1 mM) or DRB (0.1 mM). Attached cells were harvested and mitochondrial fractions were prepared. The levels of cytochrome c and citrate teniposide and other anticancer drugs can induce apoptosis in various synthase were determined by Western blot analysis as described in “Materials and cell lines, flow cytometric analysis of MDA-MB-231 cells treated Methods.” The results shown are representative from one of three separate experiments. 1040

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2001 American Association for Cancer Research. MITOCHONDRIAL CYTOCHROME c EXPRESSION PRECEDES CELL DEATH

that the extensive release of cytochrome c was associated with mito- chondrial disintegration. Because the data in the attached cells suggested that there was a net increase in total cellular cytochrome c protein, we tested whether the increase in cytochrome c was transcriptional or translational. Cells were treated with teniposide in the absence or presence of cyclohex- imide (a protein synthesis inhibitor) or DRB (a transcription inhibitor; Fig. 4). The results showed that cycloheximide, but not DRB, pre- vented the teniposide-induced increase in cytochrome c levels in the mitochondrial fraction. These data suggested that this phenomenon was dependent on de novo protein synthesis. Taken together, these results indicate that teniposide induced a dose- and protein synthesis-dependent increase in respiratory chain proteins before cells died. Essentially the same results were obtained with HeLa cells treated with teniposide (data not shown) and Jurkat cells treated with camptothecin (21), suggesting that this is a more general phenomenon. Immunofluorescence Microscopy. To confirm the finding that cytochrome c is released into the cytosol in living cells, the cellular localization of cytochrome c protein was examined by immunofluo- rescence microscopy in MDA-MB-231 cells treated with teniposide for 48 h. At this stage we found live and dead cells, which allowed us to compare the different cytochrome c expression patterns in both populations. To distinguish between live and dead cells, we used the Dead Red dye, which is a cell-impermeant red fluorescent nucleic acid stain that labels only dead cells (22). By using this stain, viability staining can take place before para-formaldehyde treatment (fixation) without disrupting the distinctive immunofluorescence-staining pat- tern. Untreated cells demonstrated a punctate pattern for cytochrome c consistent with its mitochondrial localization (Fig. 5A). Upon teni- poside treatment, a brighter punctate, as well as a somewhat diffuse, staining was seen throughout the live cells (Fig. 5B). Together with the Western blot data, these results support the conclusion that there Fig. 5. Immunolocalization of cytochrome c. Teniposide-treated (48 h) and untreated was an increased amount of cytochrome c in the mitochondria and MDA-MB-231 cells were stained for cytochrome c (cyto-c) by indirect immunofluores- some release of cytochrome c from the mitochondria to the cytosol cence as described in “Materials and Methods.” Staining with Hoechst 33343 was used to before cell death. In contrast, the staining in dead cells was far more visualize the nucleus, whereas the Dead Red dye was used to identify dead cells. A representative field from both untreated (A) and treated (B) cells is shown. diffuse and mostly without a punctate pattern (see short arrow in Fig. 5B), which is in agreement with our observation by Western blot that mitochondria are depleted of cytochrome c after cell death. Similar also occurred after teniposide treatment, we analyzed cytosolic and results were also obtained in teniposide-treated HeLa cells (data not mitochondrial fractions from attached and floating cells separately by shown). Western blot for the presence of cytochrome c. As shown in Fig. 2A, The Teniposide-induced Increase in Cytochrome c Is Func- increasing amounts of cytochrome c were detected in the cytosol from tional. To test if the teniposide-induced up-regulation of mitochon- attached MDA-MB-231 cells in a time-dependent manner after treat- drial respiratory chain protein expression was accompanied by a ment with teniposide. Surprisingly, however, the analysis of the corresponding mitochondrial fractions from the same cells did not show a depletion of cytochrome c as one might expect if this protein were released into the cytoplasm. Rather, the levels of cytochrome c increased almost 10-fold within 24 h after treatment with teniposide (Fig. 2B). Similarly, levels of cytochrome oxidase subunits I and IV, two other respiratory chain proteins, also increased, though to a somewhat smaller extent. The increase of these proteins was dose- dependent and reached a maximum at 10 ␮M teniposide (Fig. 2C). In contrast, no changes were detected in either the mitochondrial matrix protein citrate synthase or the outer membrane protein VDAC. These results suggested that the effect of teniposide on mitochondrial pro- teins might be specific. Furthermore, as the data in Fig. 2B show, the increase in mitochondrial cytochrome c occurred 24 h before that in the cytosol, suggesting a possible cause-and-effect relationship. Fig. 6. Effect of teniposide treatment on COX-dependent oxygen uptake in MDA- In contrast with what was seen with attached cells, in floating cells MB-231. MDA-MB-231 cells were treated with 10 ␮M teniposide for 24, 48, or 72 h. cytochrome c disappeared from the mitochondrial fraction as it ap- Attached cells were harvested and placed into an oxygen electrode cuvette. Oxygen consumption was measured using ascorbate/TMPD as the electron donor. Values are P Ͻ 0.001 between control and ,ء .peared in the cytosolic fraction (Fig. 3). Similarly, there also was a given as means Ϯ SD of three independent experiments loss of citrate synthase from the mitochondrial fraction, suggesting treated cells. 1041

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2001 American Association for Cancer Research. MITOCHONDRIAL CYTOCHROME c EXPRESSION PRECEDES CELL DEATH ⌬⌿ Teniposide Did Not Induce m Reduction in Attached Cells. Previous studies have suggested that a decline of the mitochondrial ⌬⌿ membrane potential m may be an early event in the process of cell ⌬⌿ death. Therefore, we determined m at various times after tenipo- side treatment in MDA-MB-231 cells. For this purpose we used the membrane potential-sensitive probe JC-1, which forms monomers (green fluorescence) at low membrane potential and J-aggregates (red fluorescence) at higher membrane potential. The ratio between the red ⌬⌿ and the green signals is indicative of the m. JC-1 fluorescence ratios remained essentially unchanged during the entire incubation time in the attached cell population (Fig. 8). In contrast, there was a dramatic drop of the red fluorescence in the floating cells, indicating ⌬⌿ a loss of m. To confirm that the JC-1 dye was sensitive to mitochondrial transmembrane depolarization, control MDA-MB-231 cells were treated with the mitochondrial uncoupling agent CCCP (1 ␮M). Within 10 min, essentially the entire population exhibited a ⌬⌿ decline of red fluorescence, indicative of a loss of m. These results indicate that the mitochondrial membrane potential remained intact in the attached population. Caspase Activation and PARP Cleavage Occurred in Floating, but not in Attached, Cells. Recent studies have demonstrated that Fig. 7. Effect of various anticancer agents on mitochondrial protein levels. MDA-MB- the release of cytochrome c from mitochondria leads to activation of 231 cells were treated for 24 h with teniposide, doxorubicin, taxol, camptothecin, vin- the caspase cascade in the cytosol (3, 10, 11, 23). To assess whether cristine, and methotrexate, as indicated. Attached cells were harvested and mitochondrial fractions were prepared. The levels of cytochrome c, COX I, COX IV, and citrate synthase the appearance of cytochrome c in the cytosol of teniposide-treated were determined by Western blot analysis as described in “Materials and Methods.” MDA-MB-231 cells led to the activation of caspases 9 and 3, cyto- Pictures are representative from one of three studies. solic fractions from both attached and floating cells were analyzed by immunoblotting with anti-caspases 9 and 3 antibodies (Fig. 9). Sig- nificant amounts of both caspases were detected in the cytoplasm, but functional increase in the transport of electrons through the respiratory there was no apparent activation of either caspase in the attached cell chain, we examined COX-dependent oxygen uptake in teniposide- population. In contrast, however, clear activation of both caspases was treated MDA-MB-231 cells (Fig. 6). The results showed a significant detected in the cytosol of the floating cells. Furthermore, PARP increase in COX activity, consistent with the observed up-regulation cleavage was observed in the cytosol of the floating, but not the of components of the respiratory chain, and suggested that the newly attached, cells as indicated by the appearance of the Mr 85,000 synthesized proteins were functional. fragment and consistent with the activation pattern of the caspases. Various Chemotherapeutic Drugs Induced Increased Levels of Thus, it appeared that caspase activation was associated with cell Cytochrome c and Cytochrome Oxidase Subunits I and IV. To death but did not correlate with the release of cytochrome c into the assess if the teniposide-mediated increase in mitochondrial respiratory cytoplasm of attached cells. chain protein levels was specific to this drug, we also treated MDA- MB-231 cells with various other anticancer drugs for 24 h, and DISCUSSION determined levels of cytochrome c and subunits I and IV of COX proteins in the mitochondrial fraction. As seen in Fig. 7, all of the In the present study we demonstrated that anticancer drug treatment drugs tested had a similar effect on the levels of these proteins, resulted in an early increase in mitochondrial respiratory chain protein suggesting that it might be a more general response to drug treatment. levels, in particular cytochrome c, that preceded cytochrome c release

⌬⌿ Fig. 8. Teniposide did not induce m reduction in at- tached cells. Cells were treated with 10 ␮M teniposide for 24, 48, or 72 h. Untreated controls and teniposide-treated cells were then stained with JC-1 and analyzed by flow cytometry as described in “Materials and Methods.” Bivariate plots of red (FL2) versus green (FL1) fluorescence were used as an ⌬⌿ estimate of mitochondrial membrane potential ( m). Live cells were gated on the basis of their forward/side scatter characteristics. n ϭ 10,000 cells/analysis. Three separate experiments were conducted that produced results similar ⌬⌿ to those shown here. As a control, m was measured in the presence of CCCP, an uncoupling agent that reduces ⌬⌿ m. Floating cells were collected at 48 h and analyzed the same way.

1042

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2001 American Association for Cancer Research. MITOCHONDRIAL CYTOCHROME c EXPRESSION PRECEDES CELL DEATH

reactive oxygen species in the cell. The fact that teniposide also up-regulated the mitochondrial expression of manganous superoxide dismutase suggested that free radicals are involved in the pathological changes observed. In this context, therefore, up-regulation of cyto- chrome c levels in mitochondria may play a protective role during the initial response of cells to drug treatment. Different models have been proposed to explain the mechanism of cytochrome c release from the intermembrane space of mitochondria during apoptosis or necrosis. Swelling and subsequent rupture of the outer mitochondrial membrane have been proposed as a mechanism for the release of cytochrome c into the cytosol (26), events which are usually associated with the mitochondrial PT (27, 28) and with a loss ⌬⌿ of m (28). However, cytochrome c appeared in the cytoplasm of ⌬⌿ teniposide-treated cells with a normal m. Furthermore, using elec- tron microscopy, predominantly hyperdense and condensed mitochon- Fig. 9. Caspase activation and PARP cleavage in MDA-MB-231 cells after teniposide dria were observed after teniposide treatment of MDA-MB231 cells treatment. Western blot analysis of cytosolic extracts from attached and floating MDA- (data not shown), suggesting that the swelling of mitochondria is MB-231 cells treated with 10 ␮M teniposide for 24, 48, or 72 h. The blots were probed with antibodies against caspase-9, caspase-3, and PARP as indicated. Activation of unlikely to be the primary mechanism of cytochrome c release. pro-caspase 9 and pro-caspase 3 is seen as a loss of their proforms or as the detection of Together, these data suggest that mitochondrial depolarization was not their active form as indicated by arrows. Pictures are representative from one of three required for cytochrome c release, a conclusion that is also consistent studies. with previous results reported by us and by others (17, 29). In conclusion, the present work demonstrated that teniposide and into the cytosol. In mitochondria, cytochrome c is required as an other chemotherapeutic drugs can induce an increase in cytochrome c electron carrier during oxidative phosphorylation, where cytochrome and other mitochondrial respiratory chain proteins. We have also c shuttles electrons from complex III to complex IV of the respiratory shown that cytochrome c release from mitochondria to the cytosol is ⌬⌿ chain (24). The electron transport between these complexes generates an early event preceding the drop of m and caspase activation. We a proton gradient across the inner mitochondrial membrane, which is propose that mitochondrial cytochrome c enrichment may play a ⌬⌿ required to maintain m. Thus, the release of cytochrome c from the critical role in the initial defense response of a cell and precedes the ⌬⌿ is expected to result in impairment of the final events leading to extensive cytochrome c release, a drop of m, ⌬⌿ electron flow and a decrease in m. The data in the present study caspase activation, plasma membrane disruption, and eventually to indicate, however, that even when cytochrome c enters the cytosol, cell death. ⌬⌿ m is maintained for a substantial time afterward. In this context, our observation that teniposide treatment also induced an increase in ACKNOWLEDGMENTS several other mitochondrial respiratory proteins, in particular subunits I and IV of COX, may help us to understand how cells can maintain We thank Renji Song and Robert Dilwith for their help with the flow ⌬⌿ cytometry and Dr. Jeff Ault for help with the electronmicroscopy. We ac- a high m despite the release of cytochrome c from the mitochon- dria. New synthesis of mitochondrial cytochrome c may serve to knowledge the videomicroscopy, cellular and molecular immunology, and prevent its levels from falling below a critical threshold required to electron microscopy core facilities of the Wadsworth Center, as well as Jan ⌬⌿ Galligan from the photography department for help with the figures. maintain m. Although the exact signal that leads to cytochrome c protein synthesis is not known, one possible hypothesis is that the initial release of cytochrome c itself functions as the feedback signal REFERENCES that triggers new synthesis of mitochondrial proteins in a kind of 1. Kroemer, G., Zamzami, N., and Susin, S. A. Mitochondrial control of apoptosis. compensatory reaction. Alternatively, it is possible that mitochondrial Immunol. Today, 18: 44–51, 1997. 2. Decaudin, D., Geley, S., Hirsch, T., Castedo, M., Marchetti, P., Macho, A., Kofler, R., cytochrome c synthesis and enrichment is the primary event and and Kroemer, G. Bcl-2 and Bcl-XL antagonize the mitochondrial dysfunction pre- cytochrome c release (or leakage) is simply a consequence of the ceding nuclear apoptosis induced by chemotherapeutic agents. Cancer Res., 57: 62–67, 1997. higher levels of this protein. The fact that only a small amount of 3. Liu, X., Kim, C. N., Yang, J., Jemmerson, R., and Wang, X. Induction of apoptotic cytochrome c was detected in the cytosol at 24 h, a time when program in cell-free extracts: requirement for dATP and cytochrome c. Cell, 86: mitochondria were already maximally enriched with cytochrome c 147–157, 1996. 4. Li, P., Nijhawan, D., Budihardjo, I., Srinivasula, S. M., Ahmad, M., Alnemri, E. S., (Fig. 2B), suggests that up-regulation of cytochrome c expression is and Wang, X. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 the primary event. The physiological and/or pathological significance complex initiates an apoptotic protease cascade. Cell, 91: 479–489, 1997. of this effect in anticancer drug-treated cells is currently unknown. 5. Kroemer, G., Dallaporta, B., and Resche-Rigon, M. The mitochondrial death/life regulator in apoptosis and necrosis. Annu. Rev. Physiol., 60: 619–642, 1998. However, this effect does not seem to be unique to the present study’s 6. Li, Y. Z., Li, C. J., Pinto, A. V., and Pardee, A. B. Release of mitochondrial model system, because increased cytochrome c levels were also cytochrome C in both apoptosis and necrosis induced by ␤-lapachone in human carcinoma cells. Mol. Med., 5: 232–239, 1999. observed within 1–2 h of staurosporine treatment of HeLa cells (data 7. Samali, A., Nordgren, H., Zhivotovsky, B., Peterson, E., and Orrenius, S. A compar- not shown) or camptothecin treatment of Jurkat cells (21). Recently, ative study of apoptosis and necrosis in HepG2 cells: oxidant- induced caspase Skulachev (25) has proposed that the role of cytochrome c in apo- inactivation leads to necrosis. Biochem. Biophys. Res. Commun., 255: 6–11, 1999. 8. Marchetti, P., Castedo, M., Susin, S. A., Zamzami, N., Hirsch, T., Macho, A., ptosis might represent one of the anti-oxidant functions inherent in Haeffner, A., Hirsch, F., Geuskens, M., and Kroemer, G. Mitochondrial permeability this protein. First, cytochrome c can operate as an enzyme, oxidizing transition is a central coordinating event of apoptosis. J. Exp. Med., 184: 1155–1160, free oxygen radicals (O Ϫ.) back to O . The reduced cytochrome c is 1996. 2 2 9. Susin, S. A., Zamzami, N., Castedo, M., Hirsch, T., Marchetti, P., Macho, A., Daugas, then reoxidized by COX. Second, the intermembrane cytochrome c E., Geuskens, M., and Kroemer, G. Bcl-2 inhibits the mitochondrial release of an can activate the electron transport chain in the outer mitochondrial apoptogenic protease. J. Exp. Med., 184: 1331–1341, 1996. 10. Kluck, R. M., Bossy-Wetzel, E., Green, D. R., and Newmeyer, D. D. The release of membrane. This bypasses the initial and middle part of the main cytochrome c from mitochondria: a primary site for Bcl-2 regulation of apoptosis. respiratory chain, which produces, as a rule, the major portion of Science (Washington DC), 275: 1132–1136, 1997. 1043

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2001 American Association for Cancer Research. MITOCHONDRIAL CYTOCHROME c EXPRESSION PRECEDES CELL DEATH

11. Yang, J., Liu, X., Bhalla, K., Kim, C. N., Ibrado, A. M., Cai, J., Peng, T. I., Jones, implications for studies on mitochondrial functionality during apoptosis. FEBS Lett., D. P., and Wang, X. Prevention of apoptosis by Bcl-2: release of cytochrome c from 411: 77–82, 1997. mitochondria blocked. Science (Washington DC), 275: 1129–1132, 1997. 20. Bradford, M. M. A rapid and sensitive method for the quantitation of microgram 12. Bossy-Wetzel, E., Newmeyer, D. D., and Green, D. R. Mitochondrial cytochrome c quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem., release in apoptosis occurs upstream of DEVD-specific caspase activation and inde- 72: 248–254, 1976. pendently of mitochondrial membrane depolarization. EMBO J., 17: 37–49, 1998. 21. Sa´nchez-Alca´zar, J. A., Khodjakov, A., Ault, J., and Schneider, E. Increased mito- 13. Green, D. R., and Reed, J. C. Mitochondria and apoptosis. Science (Washington DC), chondrial cytochrome c levels and mitochondrial hyperpolarization precede campto- 281: 1309–1312, 1998. thecin-induced apoptosis in Jurkat cells. Cell Death Differ., 7: 1090–1100, 2000. 14. Eskes, R., Antonsson, B., Osen-Sand, A., Montessuit, S., Richter, C., Sadoul, R., 22. Molecular Probes Product Information Sheet MP 7013 for LIVE/DEAD Kit L-7013. Mazzei, G., Nichols, A., and Martinou, J. C. Bax-induced cytochrome C release from 1996. mitochondria is independent of the permeability transition pore but highly dependent 23. Kluck, R. M., Martin, S. J., Hoffman, B. M., Zhou, J. S., Green, D. R., and ϩ on Mg2 ions. J. Cell Biol., 143: 217–224, 1998. Newmeyer, D. D. Cytochrome c activation of CPP32-like proteolysis plays a critical 15. Rosse, T., Olivier, R., Monney, L., Rager, M., Conus, S., Fellay, I., Jansen, B., and role in a Xenopus cell-free apoptosis system. EMBO J., 16: 4639–4649, 1997. Borner, C. Bcl-2 prolongs cell survival after Bax-induced release of cytochrome c. 24. Hatefi, Y. The mitochondrial electron transport and oxidative phosphorylation sys- Nature (Lond.), 391: 496–499, 1998. tem. Annu. Rev. Biochem., 54: 1015–1069, 1985. 16. Krippner, A., Matsuno-Yagi, A., Gottlieb, R. A., and Babior, B. M. Loss of function 25. Skulachev, V. P. Cytochrome c in the apoptotic and antioxidant cascades. FEBS Lett., of cytochrome c in Jurkat cells undergoing fas-mediated apoptosis. J. Biol. Chem., 271: 21629–21636, 1996. 423: 275–280, 1998. 17. Sa´nchez-Alca´zar, J. A., Schneider, E., Martı´nez, M. A., Carmona, P., Herna´ndez- 26. Vander Heiden, M. G., Chandel, N. S., Williamson, E. K., Schumacker, P. T., and Mun˜oz, I., Siles, E., De la Torre, P., Ruı´z-Cabello, J., Garcı´a, I., and Solı´s-Herruzo, Thompson, C. B. Bcl-xL regulates the membrane potential and volume homeostasis J. A. -␣ increases the steady-state reduction of cytochrome b of of mitochondria. Cell, 91: 627–637, 1997. the mitochondrial respiratory chain in metabolically inhibited L929 cells. J. Biol. 27. Petit, P. X., Susin, S. A., Zamzami, N., Mignotte, B., and Kroemer, G. Mitochondria Chem., 275: 13353–13361, 2000. and programmed cell death: back to the future. FEBS Lett., 396: 7–13, 1996. 18. Smiley, S. T., Reers, M., Mottola-Hartshorn, C., Lin, M., Chen, A., Smith, T. W., 28. Reed, J. C. Cytochrome c: can’t live with it–can’t live without it. Cell, 91: 559–562, Steele, G. D., Jr., and Chen, L. B. Intracellular heterogeneity in mitochondrial 1997. membrane potentials revealed by a J-aggregate-forming lipophilic cation JC-1. Proc. 29. Dinsdale, D., Zhuang, J., and Cohen, G. M. Redistribution of cytochrome c precedes Natl. Acad. Sci. USA, 88: 3671–3675, 1991. the caspase-dependent formation of ultracondensed mitochondria, with a reduced 19. Salvioli, S., Ardizzoni, A., Franceschi, C., and Cossarizza, A. JC-1, but not DiOC6(3) inner membrane potential, in apoptotic monocytes. Am. J. Pathol., 155: 607–618, or rhodamine 123, is a reliable fluorescent probe to assess ␦␲changes in intact cells: 1999.

1044

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2001 American Association for Cancer Research. Anticancer Drugs Induce Increased Mitochondrial Cytochrome c Expression That Precedes Cell Death

José A. Sánchez-Alcázar, Alexey Khodjakov and Erasmus Schneider

Cancer Res 2001;61:1038-1044.

Updated version Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/61/3/1038

Cited articles This article cites 28 articles, 12 of which you can access for free at: http://cancerres.aacrjournals.org/content/61/3/1038.full#ref-list-1

Citing articles This article has been cited by 2 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/61/3/1038.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/61/3/1038. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2001 American Association for Cancer Research.