Exposure of U937 human leukemic cells to the phorbol ester 12-O-tetradecanoylphorbol 13-acetate (TPA) induces their differentiation into monocyte/macrophage-like cells. This terminal differentiation is associated with a resistant phenotype to apoptosis induced by the topoisomerase II inhibitor etoposide. The inhibition occurs upstream of the mitochondrial release of cytochrome c and the activation of procaspase-2, -3, -6, -7, -8, and -9. By using cell-free systems, it was demonstrated that the mitochondrial pathway to cell death that involves mitochondrial membrane depolarization, cytochrome c release and cytosolic activation of procaspases by cytochrome c/dATP remains functional in TPA-differentiated U937 cells. Accordingly, 2 drugs recently shown to target the mitochondria, namely lonidamine and arsenic trioxide, bypass the resistance of TPA-differentiated U937 cells to classical anticancer drugs. Cell death induced by the 2 compounds is associated with mitochondrial membrane depolarization, release of cytochrome c and Smac/Diablo from the mitochondria, activation of caspases, poly(ADP-ribose) polymerase cleavage and internucleosomal DNA fragmentation. Moreover, the decreased glutathione content associated with the differentiation process amplifies the ability of arsenic trioxide to activate the mitochondrial pathway to cell death. Similar results were obtained by comparing undifferentiated and TPA-differentiated human HL60 leukemic cells. These data demonstrate that mitochondria-targeting agents bypass the resistance to classical anticancer drugs induced by TPA-mediated leukemic cell differentiation.

Chemotherapeutic drugs can kill cultured leukemic cell lines by inducing apoptosis.1 This mode of cell death may be clinically relevant because apoptotic blast cells have been detected in the peripheral blood of patients with acute leukemia who received chemotherapeutic drugs.2 Hallmarks of apoptosis in leukemic cells include characteristic morphologic changes such as cell shrinkage, membrane blebbing, and cell fragmentation into apoptotic bodies, internucleosomal DNA fragmentation, and limited proteolytic cleavage of selective intracellular proteins such as the nuclear enzyme poly(ADP-ribose)polymerase (PARP). This cell death phenotype is induced by activation of a constitutively expressed apoptotic machinery that involves a family of cysteine proteases known as caspases, organized in a branched proteolytic cascade. These enzymes are synthesized as inactive proenzymes (procaspases) whose proteolysis at internal aspartate residues generates large and small subunits.3 Heterodimerization of these subunits is required to form the active enzyme (caspase) that cleaves intracellular substrates, either a downstream procaspase or other cellular proteins.4 

Activation of the caspase cascade in response to specific damage induced by chemotherapeutic drugs is the consequence of a disruption of the mitochondrial membrane barrier function.5 Although the exact mechanism remains controversial, mitochondrial changes lead to the release of soluble apoptogenic proteins such as cytochrome c,6 apoptosis-inducing factor (AIF),7 and Smac/Diablo8,9 from the mitochondrial intermembrane space to the cytosol. Once in the cytosol, cytochrome c, in the presence of adenosine triphosphate (ATP), induces conformational changes of an adaptor molecule designated Apaf-1 that recruits and activates procaspase-9 molecules.10,11 In turn, caspase-9 activates the downstream caspase cascade that involves caspase-3 and other effector enzymes.12 Smac/Diablo was recently shown to suppress the inhibition of caspases by proteins known as inhibitor of apoptosis proteins (IAPs), thereby increasing their sensitivity to the cytochrome c/ATP pathway of activation.8,9 Once activated, effector caspases cleave a limited set of essential cellular proteins, leading to cell dismantling.13-15 Mitochondrial function during apoptosis is controlled by the Bcl-2 family of proteins that could prevent the opening of the permeability transition pore or stabilize the barrier function of the outer mitochondrial membrane and prevent the release of apoptogenic molecules.5,16 

How the specific damage induced by chemotherapeutic drugs leads to mitochondrial changes and activation of the caspase cascade under the control of Bcl-2–related proteins remains poorly understood. We have shown previously that differentiation of human myeloid leukemia cell lines along a macrophagic pathway induced by exposure to the phorbol ester 12-O-tetradecanoylphorbol 13-acetate (TPA) correlated with a resistance phenotype to apoptosis induced by a series of anticancer drugs with distinct primary intracellular targets.17,18 This inhibition occurs downstream of specific damage, measured as cleavage complexes, induced by topoisomerase poisons.17 In addition, TPA-differentiated cells are resistant to apoptosis induced by exposure to Fas-ligand, a specific pathway that was proposed as an amplifier system in drug-induced cell death.19 A resistance to drug-induced and TRAIL-mediated apoptosis was also reported in vitamin D3- and dimethyl sulfoxide (DMSO)-differentiated HL60 cells, respectively.20,21 

Identification of the molecular mechanisms that modulate cell sensitivity to chemotherapeutic drugs in differentiated cells could help to define strategies combining differentiating and cytotoxic agents for treating acute leukemias. The present study further explores the level of inhibition of drug-induced apoptosis in TPA-differentiated cells. By using cell-free systems, we demonstrate that the pathway involving mitochondria and the downstream caspase cascade remains functional in terminally differentiated cells. Based on this observation, we tested the ability of 2 chemotherapeutic drugs recently shown to target the mitochondria, namely the indazole-3-carboxylic acid derivative lonidamine and the metalloid arsenic trioxide, to induce apoptosis in these cells.

Drugs and chemical reagents

Etoposide (VP-16) and TPA were obtained from Sigma-Aldrich Laboratories (St Quentin Fallavier, France). Lonidamine was kindly provided by Dr M. F. Poupon (Institut Curie, Paris, France). Stock solutions were prepared by diluting these reagents in DMSO and storing them at −20°C. Further dilutions were made in culture medium just before use. The final concentration of DMSO in culture medium never exceed 0.1% (vol/vol), which was nontoxic to the cells. Arsenic trioxide was kindly provided by Dr G. Q. Chen (Shangai Institute of Hematology, China). [2-14C]-Thymidine (56 mCi/mM) was obtained from Amersham (Les Ulis, France). All other chemicals were of reagent grade and were purchased from local sources.

Antibodies

The tested mouse monoclonal antibodies (mAbs) included antihuman HSC70 from Santa Cruz Biotechnology (Santa Cruz, CA), cytochrome c, caspase-2, caspase-6, and caspase-8 from Pharmingen (San Diego, CA), caspase-3 and caspase-7 from Transduction Laboratories (Lexington, KY), and cytochrome oxidase subunit IV (COX IV) from Molecular Probes (Eugene, OR). We also used the polyclonal rabbit antibodies raised against human caspase-3-p17 (kindly provided by Donald W. Nicholson, Merk-Frost, Toronto, ON, Canada), caspase-9 (Pharmingen), caspase-7 (StressGen, Victoria, BC, Canada), PARP (Boehringer-Mannheim, Germany), and Smac/Diablo (kindly provided by Xiadong Wang, Howard Hughes Medical Institute, Dallas, TX).

Cell culture and differentiation

The human leukemic cell lines U937 and HL60 (American Type Culture Collection, Rockville, MD) were grown in suspension in RPMI 1640 medium with glutamax-I (Gibco BRL, Life Technologies, Cergy Pontoise, France) supplemented with 10% (vol/vol) fetal bovine serum (BioWhittaker, Fontenay-sous-bois, France) in an atmosphere of 95% air and 5% CO2 at 37°C. Cell viability was determined by using a trypan blue dye exclusion assay. To ensure exponential growth, cells were resuspended at a density of 0.5 × 106cells/mL in fresh medium 24 hours before each treatment. To induce differentiation, cells were culture in the presence of 20 nM TPA for 24 hours (HL60) or 72 hours (U937) as previously described.17,18 After treatment, adherent cells were harvested using a cell scraper. U937 and HL60 cell clones containing the full-length human bcl-2 cDNA were kindly provided by Jacqueline Bréard (INSERM 461, Chatenay-Malabry, France) and Michele Allouche (CNRS-UPCM, CHU Purpan, Toulouse, France), respectively.

DNA fragmentation

DNA fragmentation was quantified by using a previously reported filter elution assay.22 Briefly, exponentially growing cells were incubated with 0.02 μCi/mL [2-14C]-thymidine and cultured at 37°C for 2 days. Then, cells were chased in isotope-free medium overnight, resuspended in fresh medium, and treated with different agents before or after TPA treatment. Approximately 1.0 × 106 treated or untreated labeled cells were loaded onto a protein-absorbing filter (polyvinylidene difluoride filters, 0.65 μm pore size, 25 mm diameter; Durapore membrane, Millipore, Saint-Quentin en Yvelines, France). Cells were then washed once with ice-cold phosphate-buffered saline (PBS) and subsequently lysed in 0.2% sodium sarkosyl, 2 M NaCl, 0.04 M EDTA, pH 10.0. Filters were washed with 0.02 M EDTA, pH 10.0. DNA was depurinated by incubation of filters in 1 M HCl at 65°C for 45 minutes, then released from the filters with 0.4 M NaOH for 45 minutes at room temperature. Radioactivity was counted by liquid scintillation spectrometry in each fraction (wash, lysis, EDTA wash, and filter). DNA fragmentation was measured as the fraction of disintegrations per minute (dpm) in the lysis fraction plus EDTA wash relative to the total intracellular dpm. For analyzing DNA fragmentation by agarose gel electrophoresis, cellular DNA was extracted by a previously described salting-out procedure,23 and electrophoresis was performed in 1.8% agarose gel in Tris-borate–EDTA buffer (pH 8.0) at 20 V for 14 hours. After electrophoresis, DNA was visualized by ethidium bromide staining.

Western blot analysis

After treatment, whole cell lysates were prepared by lysing the cells in boiling buffer (1% SDS, 1 mM Na-vanadate, 10 mM Tris pH 7.4) in the presence of protease inhibitors (0.1 mM phenylmethylsulfonyl fluoride [PMSF, 2.5 μg/mL pepstatin, 10 μg/mL aprotinin, 5 μg/mL leupeptin). Viscosity of the samples was reduced by several passages through a 26-gauge needle. Protein concentration was measured by the Bio-Rad DC protein assay kit (Ivry sur Seine, France). Thirty micrograms protein was incubated in loading buffer (125 mM Tris-HCl, pH 6.8, 10% β-mercaptoethanol, 4.6% SDS, 20% glycerol, and 0.003% bromophenol blue), separated by sodium dodecyl sulfate–polyacrylamide gel (SDS-PAGE), and electroblotted to polyvinylidene difluoride membrane (Bio-Rad). After blocking nonspecific binding sites overnight by 5% nonfat milk in TPBS (0.1% PBS-Tween 20), the membrane was incubated for 2 hours at room temperature with primary antibody. After 2 washes in TPBS, the membrane was incubated with horseradish peroxidase–conjugated goat antimouse or antirabbit antibodies (Jackson ImmunoResearch Laboratories, West Grove, PA) for 30 minutes at room temperature, then washed twice in TPBS. Immunoblot was revealed using enhanced chemiluminescence detection kit (Amersham) by autoradiography.

Cell fractionation

Cytosolic fractions were prepared by resuspending the cells in ice-cold buffer A (250 mM sucrose, 20 mM HEPES, 10 mM KCl, 1.5 mM MgCl2, 1 mM EDTA, 1 mM EGTA, 1 mM dithiothreitol, 17 μg/mL PMSF, 8 μg/mL aprotinin, 2 μg/mL leupeptin [pH 7.4]) before passing them through an ice-cold cell homogenizer. Unlysed cells and nuclei were pelleted by a 10-minute, 750g spin. This step was repeated twice. The supernatant was centrifuged at 10 000g for 25 minutes, and the resultant supernatant was further centrifuged at 100 000g for 1 hour. The supernatant (cytosolic fraction) was frozen at −80°C. Nuclei-free, mitochondria-free cytosolic extracts (cell-free extracts) were generated as previously described.24 Briefly, cells (1-2 × 108) were pelleted and washed twice with PBS, pH 7.2, followed by a single wash with 4 mL ice-cold cell extract buffer (20 mM HEPES, 10 mM KCl, 1.5 mM MgCl2, 1 mM EDTA, 1 mM EGTA, 1 mM dithiothreitol, 100 μM PMSF, 2 μg/mL aprotinin, 10 μg/mL leupeptin [pH 7.4]). Two volumes ice-cold extract buffer were added to 1 vol packed cell pellet before the cellular suspension was transferred to a 2-mL Dounce homogenizer. Cells were allowed to swell under the hypotonic condition for 20 minutes, then were disrupted with 30 strokes of a B-type. Lysis was confirmed with the trypan blue dye exclusion test before centrifugation of lysates at 16 000g for 15 minutes at 4°C. Caspase activation by the cytochrome c/dATP combination was tested in these clarified supernatants.

Purification of mitochondria

Purified mitochondria were isolated as previously described.24 Briefly, cells (1 × 108) were resuspended in buffer H (300 mM saccharose, 5 mM N-tris(hydroxymethyl)methyl-2-amino-ethanesulfonic acid, 200 μM EGTA, pH 6.9), homogenized in a Potter-Thomas homogenizer, then centrifuged for 10 minutes at 760g. The supernatant was recovered while the pellet was resuspended in H buffer and centrifuged to recover the supernatant as above. Both supernatants were mixed and centrifuged for 10 minutes at 8740g before the mitochondria pellet was resuspended in buffer H.

Activation of cell-free apoptosis

Cell-free extracts (10 μL, 5-10 mg/mL protein) were incubated at 37°C with 5 μM horse heart cytochrome c (Sigma) and 1 mM dATP (Pharmacia). Then caspase activation was determined by Western blot analysis. For cell-free reaction activated by atractyloside, purified mitochondria were exposed to 5 mM atractyloside for 30 minutes and pelleted by a 10 000g spin for 10 minutes before cytochrome c release was determined by Western blot in both the supernatant and the mitochondria pellet.

Analysis of mitochondrial membrane potential

Mitochondrial membrane depolarization was analyzed using the DePsipher kit (R&D Systems, Abington, United Kingdom) according to the manufacturer's instructions. Briefly, 106 cells were incubated for 20 minutes at 37°C in the presence of 5 μg/mL DepSipher (R&D Systems) solution and then washed 2 times in PBS. In some cases, cells were treated for 30 minutes with the uncoupling agent carbonyl cyanide m-chlorophenylhydrazone (100 μM; Sigma-Aldrich). Analysis was performed by the use of a FACscan cytometer (Becton Dickinson, Le Pont de Claix, France).

Quantification of intracellular glutathione and reactive oxygen species content by flow cytometry

The level of cellular glutathione (GSH) was determined by flow cytometry after staining with monochlorobimane (Molecular Probes) as previously described.25 Briefly, 200 μM monochlorobimane [syn-(ClCH2, CH3)-1,5-diazabicyclo-[3.3.0]-octa-3,6-dione-2,8-dione)] was added to cell suspensions for 30 minutes at 37°C. After 2 washes in PBS, cells were resuspended in PBS and analyzed on a Bio-Rad flow cytometer (Hercules, CA). For reactive oxygen species (ROS) measurements, cells were incubated for 15 minutes at 37°C in the presence of 6.6 μM dihydro-ethidium (Sigma-Aldrich) and analyzed by flow cytometry.

TPA-induced differentiation of U937 human leukemic cells prevents etoposide-induced apoptosis

U937 leukemic cells were induced to differentiate along a macrophagic pathway on exposure to 20 nM TPA. After a 48- to 72-hour exposure to TPA, more than 90% cells demonstrate a differentiated phenotype including adhesion to the culture flask and increased expression of the glycoprotein CD11b at the cell surface. On continuous exposure to 50 μM etoposide, most of the parental U937 cells undergo apoptosis in 6 hours, as demonstrated by staining nuclear chromatin with Hoechst 33342, whereas TPA-differentiated cells strongly resist drug-induced cell death (Figure1).

Fig. 1.

TPA-mediated differentiation of U937 cells delays etoposide-induced apoptosis.

Undifferentiated (▴) and TPA-differentiated (●) U937 cells were treated with 50 μM etoposide for indicated times. Percentages of apoptotic cells were determined by fluorescent microscopy after Hoechst staining. Each value is the mean ± SD of triplicate determinations.

Fig. 1.

TPA-mediated differentiation of U937 cells delays etoposide-induced apoptosis.

Undifferentiated (▴) and TPA-differentiated (●) U937 cells were treated with 50 μM etoposide for indicated times. Percentages of apoptotic cells were determined by fluorescent microscopy after Hoechst staining. Each value is the mean ± SD of triplicate determinations.

Close modal

To link the resistance phenotype to the differentiation process, we exposed the cells for 4.5 hours to 50 μM etoposide at various times after the beginning of TPA treatment (20 nM) (Figure2). Apoptosis was identified by Hoechst staining of nuclear chromatin (Figure 2A) and quantified by measuring nuclear DNA fragmentation (Figure 2B). Exposure to TPA provoked a time-dependent decrease of etoposide-induced morphologic changes and DNA fragmentation that were almost completely inhibited 48 to 72 hours after the beginning of TPA exposure. Accordingly, by using agarose gel electrophoresis, the characteristic nucleosome-sized ladder fragmentation of nuclear DNA observed in parental undifferentiated cells exposed for 4.5 hours to the drug was not detected in TPA-differentiated cells treated in the same conditions (Figure 2B, insert).

Fig. 2.

Inhibition of etoposide-induced apoptosis in U937 cells induced to differentiate by TPA.

U937 cells were untreated (TE) or preincubated for indicated times with 20 nM TPA, washed, then treated for 4.5 hours with 50 μM etoposide. (A) Nuclear morphologic changes associated with apoptosis were visualized by fluorescent microscopy after Hoechst staining. (B) (upper panel) Apoptotic DNA fragmentation was quantified using a filter elution assay. Values from 1 of 2 independent experiments are shown (mean ± SD of triplicate samples). (inset) Visualization of apoptotic DNA fragmentation by agarose gel electrophoresis. (lower panel) Percentage of differentiated cells according to CD11b expression measured before etoposide treatment. (C) Indicated caspases and PARP were analyzed in whole-cell extracts by immunoblot. Numbers are molecular weights in kilodaltons. “Procaspase” and “caspase” are used to design the studied enzymes, depending on whether the tested antibody does not or does detect the cleavage fragments (∗), respectively. A unique series of cell lysates was used for these experiments. Loading of each strip was checked by using an antihuman HSC70 mAb (one representative control is shown).

Fig. 2.

Inhibition of etoposide-induced apoptosis in U937 cells induced to differentiate by TPA.

U937 cells were untreated (TE) or preincubated for indicated times with 20 nM TPA, washed, then treated for 4.5 hours with 50 μM etoposide. (A) Nuclear morphologic changes associated with apoptosis were visualized by fluorescent microscopy after Hoechst staining. (B) (upper panel) Apoptotic DNA fragmentation was quantified using a filter elution assay. Values from 1 of 2 independent experiments are shown (mean ± SD of triplicate samples). (inset) Visualization of apoptotic DNA fragmentation by agarose gel electrophoresis. (lower panel) Percentage of differentiated cells according to CD11b expression measured before etoposide treatment. (C) Indicated caspases and PARP were analyzed in whole-cell extracts by immunoblot. Numbers are molecular weights in kilodaltons. “Procaspase” and “caspase” are used to design the studied enzymes, depending on whether the tested antibody does not or does detect the cleavage fragments (∗), respectively. A unique series of cell lysates was used for these experiments. Loading of each strip was checked by using an antihuman HSC70 mAb (one representative control is shown).

Close modal

Etoposide treatment of parental cells also induced a decrease of procaspase-2, -3, -6, -7, -8, and -9 expression, as determined by Western blot analysis (Figure 2C). By using antibodies reacting with the cleavage fragments that characterize active enzymes, decreased expression of the 48-kd procaspase-2, the 32-kd procaspase-3, the 35-kd procaspase-7, and the 48-kd procaspase-9 correlated with the appearance of a p35, a p19/p17, a p10, and a p37 active fragment, respectively. The 30-kd fragment identified with the anti–caspase-9 antibody might correspond to the previously described caspase-9b isoform.26 The significance of the 18-kd band remains unknown. All these etoposide-induced events were progressively inhibited as U937 cells progressed toward the differentiation phenotype on TPA exposure. This inhibition became identifiable between 6 and 12 hours after the beginning of TPA exposure, depending on the experiment. In addition, TPA-induced differentiation was associated with a progressive inhibition of the 116-kd DNA repair enzyme PARP cleavage into an 85-kd N-terminal fragment (Figure 2C).

Caspases remain able to be activated in cell-free extracts from TPA-induced differentiated cells

Exposure of undifferentiated U937 cells to 50 μM etoposide for 4.5 hours triggered the appearance of cytochrome c in the cytosol. This event was prevented by TPA-induced differentiation (Figure3A). To determine whether the cytochrome c–mediated activation of the caspase cascade was influenced by the differentiation process, we used a previously described cell-free system.24 Exposure of cell-free extracts from undifferentiated U937 cells to 5 μM cytochrome c and 1 mM dATP induced a time-dependent decrease of the expression of procaspase-2, -3, -6, -7, and -9, suggesting their cleavage into active fragments. Accordingly, immunoblot experiments detected the time-dependent appearance of the 35-kd active fragment of caspase-2, the 19- and 17-kd fragments of caspase-3, the 11-kd fragment of caspase-6, the 30-kd fragment of caspase-7, and the 37-kd fragment of caspase-9 (Figure 3B). In this time-course, procaspase-8 expression remained unchanged. TPA-mediated differentiation of U937 cells induced some changes in the subcellular localization of procaspases, mainly the disappearance of procaspase-2 from the cytosol (O.S. et al, manuscript in preparation). The other procaspases remained sensitive to activation by the cytochrome c–dATP combination with kinetics similar to that observed in cytosolic extracts from undifferentiated cells (Figure 3B). Thus, TPA-induced differentiation does not significantly influence procaspase activation by cytochrome c/dATP.

Fig. 3.

Cytochrome c-mediated activation of caspases in cell-free extracts.

(A) Western blot analysis of cytochrome c in cytosolic fractions from undifferentiated (U937) and TPA-differentiated (U-TPA) U937 cells treated (+) or not treated (−) with 50 μM etoposide for 4.5 hours. (B) Cell-free extracts from U937 and U-TPA cells were left untreated for 2 hours (TE) or were incubated for indicated times (minutes) in the presence of 5 μM cytochrome c and 1 mM dATP. Processing of indicated caspases was analyzed by Western blot. Numbers are molecular weights in kilodaltons. *Cleavage products. Loading was checked with the use of an antihuman HSC70 mAb.

Fig. 3.

Cytochrome c-mediated activation of caspases in cell-free extracts.

(A) Western blot analysis of cytochrome c in cytosolic fractions from undifferentiated (U937) and TPA-differentiated (U-TPA) U937 cells treated (+) or not treated (−) with 50 μM etoposide for 4.5 hours. (B) Cell-free extracts from U937 and U-TPA cells were left untreated for 2 hours (TE) or were incubated for indicated times (minutes) in the presence of 5 μM cytochrome c and 1 mM dATP. Processing of indicated caspases was analyzed by Western blot. Numbers are molecular weights in kilodaltons. *Cleavage products. Loading was checked with the use of an antihuman HSC70 mAb.

Close modal

Mitochondria from TPA-induced differentiated U937 cells remain reactive

The mechanisms that account for the mitochondrial release of cytochrome c are still controversial. One of the proposed mechanisms involves a decrease of the mitochondrial membrane potential Δψm.5 To determine whether TPA-induced differentiation was associated with a decreased reactivity of mitochondria to uncoupling agents, we exposed undifferentiated and TPA-differentiated U937 cells for 30 minutes to 100 μM of the uncoupling agent carbonyl cyanide m-chlorophenylhydrazone (mCICCP). This treatment induced a switch from the mitochondrial aggregate (FL2-H) to the cytosolic monomer (FL1-H) that was similar in mitochondria from both undifferentiated and TPA-differentiated cells indicating mitochondrial depolarization (Figure 4A). A mechanism proposed to account for the mitochondrial release of cytochrome c was the opening of the megachannel that can be induced by exposing purified mitochondria to atractyloside (5 mM, 30 minutes). This compound induced the release of cytochrome c from mitochondria of undifferentiated and differentiated U937 cells with a similar efficacy (Figure 4B). In the presence of cell-free extracts from undifferentiated U937 cells, the effect of atractyloside on purified mitochondria resulted in cleavage of the DEVD-AFC substrate, suggesting caspase activation (data not shown). Together these results suggested that the mitochondrial pathway to cell death may remain functional in TPA-differentiated U937 cells.

Fig. 4.

TPA-induced differentiation of U937 cells does not alter mitochondrial reactivity.

(A) Flow cytometry analysis of mitochondrial potential in undifferentiated (U937) and TPA-differentiated (U-TPA) U937 cells, treated (+) or not treated (−) with 100 μM m-chlorophenylhydrazone for 30 minutes. Mitochondrial membrane depolarization is indicated by the decreased mitochondrial aggregate (FL2-H) and the increased cytosolic monomer (FL1-H). Dashed lines indicate the controls. (B) Purified mitochondria isolated from U937 and U-TPA cells were treated (+) or not treated (−) with 5 mM atractyloside for 30 minutes. Cytochrome c expression was analyzed by Western blot in mitochondrial (Mit) and supernatant (Buffer) fractions. Lack of supernatant contamination by mitochondria extracts was checked by using an antihuman cytochrome oxidase subunit IV (COX IV) mAb.

Fig. 4.

TPA-induced differentiation of U937 cells does not alter mitochondrial reactivity.

(A) Flow cytometry analysis of mitochondrial potential in undifferentiated (U937) and TPA-differentiated (U-TPA) U937 cells, treated (+) or not treated (−) with 100 μM m-chlorophenylhydrazone for 30 minutes. Mitochondrial membrane depolarization is indicated by the decreased mitochondrial aggregate (FL2-H) and the increased cytosolic monomer (FL1-H). Dashed lines indicate the controls. (B) Purified mitochondria isolated from U937 and U-TPA cells were treated (+) or not treated (−) with 5 mM atractyloside for 30 minutes. Cytochrome c expression was analyzed by Western blot in mitochondrial (Mit) and supernatant (Buffer) fractions. Lack of supernatant contamination by mitochondria extracts was checked by using an antihuman cytochrome oxidase subunit IV (COX IV) mAb.

Close modal

Mitochondria-targeting agent lonidamine induced apoptosis of TPA-differentiated cells

The previous observations suggested that drugs shown to directly target the mitochondria might overcome the resistance of TPA-differentiated cells to etoposide and various other classical anticancer drugs.18 We first tested the mitochondria-targeting drug lonidamine.27 The ability of this compound to induce apoptosis of TPA-differentiated cells was only slightly delayed in TPA-differentiated compared to undifferentiated U937 cells, as demonstrated by quantifying DNA fragmentation (Figure5A) and by counting the percentage of cells demonstrating nuclear chromatin condensation after a 24- to 48-hour exposure to 200 μM lonidamine (Table1). Lonidamine-induced apoptotic DNA fragmentation and chromatin condensation were completely inhibited by stable overexpression of Bcl-2 in U937 cells (Figure 5A; Table 1). In undifferentiated and TPA-differentiated cells, lonidamine induced mitochondrial depolarization (Figure 5C), cytochrome c release in the cytosol (Figure 5B), procaspase-3 cleavage in its active fragments, and PARP cleavage (Figure 5D). All these events were prevented by Bcl-2 overexpression. The recently described Smac/Diablo protein8,9 was only slightly released in the cytosol on lonidamine exposure, an effect that was not prevented by Bcl-2 overexpression (Figure 5B).

Fig. 5.

Lonidamine induces apoptosis in TPA-differentiated U937 cells.

Undifferentiated (▴, U937), TPA-differentiated (●, U-TPA), and Bcl-2–overexpressing U937 cells (▪, U-BCL2) were treated for indicated times with 200 μM lonidamine. (A) Apoptotic DNA fragmentation was quantified using a filter elution assay. Values from 1 of 2 independent experiments are shown (mean ± SD of triplicate samples). (B) Western blot analysis of cytochrome c and Smac/Diablo expression in cytosolic fractions from U937, U-TPA, and U-BCL2 cells treated (+) or not treated (−) with 200 μM lonidamine for 48 hours. (C) Mitochondrial membrane depolarization was visualized by flow cytometry in U937, U-TPA, and U-BCL2 cells treated with 200 μM lonidamine for indicated times. Mitochondrial depolarization was identified by an increased level of cytosolic green monomer (FL1-H; dashed lines indicate the controls). (D) Western blot analysis of procaspase-3 and caspase-3 active fragments and PARP expression in whole-cell extracts from U937, U-TPA, and U-BCL2 cells treated (+) or not treated (−) with 200 μM lonidamine for 48 hours. Numbers are molecular weights in kilodaltons. *Cleavage products. Loading was checked with the use of an antihuman HSC70 mAb.

Fig. 5.

Lonidamine induces apoptosis in TPA-differentiated U937 cells.

Undifferentiated (▴, U937), TPA-differentiated (●, U-TPA), and Bcl-2–overexpressing U937 cells (▪, U-BCL2) were treated for indicated times with 200 μM lonidamine. (A) Apoptotic DNA fragmentation was quantified using a filter elution assay. Values from 1 of 2 independent experiments are shown (mean ± SD of triplicate samples). (B) Western blot analysis of cytochrome c and Smac/Diablo expression in cytosolic fractions from U937, U-TPA, and U-BCL2 cells treated (+) or not treated (−) with 200 μM lonidamine for 48 hours. (C) Mitochondrial membrane depolarization was visualized by flow cytometry in U937, U-TPA, and U-BCL2 cells treated with 200 μM lonidamine for indicated times. Mitochondrial depolarization was identified by an increased level of cytosolic green monomer (FL1-H; dashed lines indicate the controls). (D) Western blot analysis of procaspase-3 and caspase-3 active fragments and PARP expression in whole-cell extracts from U937, U-TPA, and U-BCL2 cells treated (+) or not treated (−) with 200 μM lonidamine for 48 hours. Numbers are molecular weights in kilodaltons. *Cleavage products. Loading was checked with the use of an antihuman HSC70 mAb.

Close modal
Table 1.

Effects of phorbol ester 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced differentiation and Bcl-2 overexpression on etoposide, lonidamine, and arsenic trioxide–induced apoptosis in U937 and HL60 human leukemic cells

U937U-TPAU-BCL2HL60HL-TPAHL-BCL2
Control 2.8 ± 1.6 1.0 ± 0.6 0.8 ± 0.7 0.8 ± 0.7 1.3 ± 1.2 0.3 ± 0.7 
VP16       
  6 h 71.7 ± 6.7 4.3 ± 2.1 — 46.3 ± 1.2 2.3 ± 2.6 — 
 24 h 94.0 ± 1.6 22.3 ± 3.1 — 98.7 ± 1.2 9.5 ± 0.9 — 
LND       
 24 h 39.3 ± 3.5 16.3 ± 3.3 2.8 ± 1.1 79.7 ± 4.1 25.2 ± 5.2 3.3 ± 1.2 
 48 h 93.3 ± 2.7 81.3 ± 5.5 4.5 ± 1.4 98.7 ± 0.7 67.5 ± 5.4 13.2 ± 2.6 
As2O3       
 24 h 12.7 ± 2.1 26.8 ± 5.8 2.3 ± 2.0 8.0 ± 1.6 20.3 ± 3.7 0.3 ± 0.5 
 48 h 30.3 ± 2.5 79.5 ± 8.0 2.7 ± 0.5 13.0 ± 5.5 60.7 ± 10.5 0.6 ± 0.5 
U937U-TPAU-BCL2HL60HL-TPAHL-BCL2
Control 2.8 ± 1.6 1.0 ± 0.6 0.8 ± 0.7 0.8 ± 0.7 1.3 ± 1.2 0.3 ± 0.7 
VP16       
  6 h 71.7 ± 6.7 4.3 ± 2.1 — 46.3 ± 1.2 2.3 ± 2.6 — 
 24 h 94.0 ± 1.6 22.3 ± 3.1 — 98.7 ± 1.2 9.5 ± 0.9 — 
LND       
 24 h 39.3 ± 3.5 16.3 ± 3.3 2.8 ± 1.1 79.7 ± 4.1 25.2 ± 5.2 3.3 ± 1.2 
 48 h 93.3 ± 2.7 81.3 ± 5.5 4.5 ± 1.4 98.7 ± 0.7 67.5 ± 5.4 13.2 ± 2.6 
As2O3       
 24 h 12.7 ± 2.1 26.8 ± 5.8 2.3 ± 2.0 8.0 ± 1.6 20.3 ± 3.7 0.3 ± 0.5 
 48 h 30.3 ± 2.5 79.5 ± 8.0 2.7 ± 0.5 13.0 ± 5.5 60.7 ± 10.5 0.6 ± 0.5 

Undifferentiated (U937, HL60), TPA-differentiated (U-TPA, HL-TPA), and Bcl-2 overexpressing (U-BCL2, HL-BCL2) U937 and HL60 cells were left untreated (control) or treated for indicated times with 50 μM etoposide (VP16), 200 μM lonidamine (LND), or 4 μM arsenic trioxide (As2O3). Results are expressed as percentages of apoptotic cells (mean ± SD of triplicate determinations).

TPA-induced differentiation sensitizes U937 cells to arsenic trioxide–induced apoptosis

As2O3 is another member of this novel class of chemotherapeutic agents that induce apoptosis, at least in part, by acting on the mitochondria.28,29As2O3 induced apoptosis of undifferentiated U937 cells in a dose- and time-dependent manner (Figure6A; Table 1). TPA-induced differentiation did not prevent, and even sensitized, U937 cells to As2O3-induced apoptosis. The differentiation process also increased the ability of As2O3 to induce mitochondrial depolarization (Figure 6C), cytochrome c release from the mitochondria to the cytosol (Figure 6B), procaspase-3 cleavage in its active fragments and PARP cleavage (Figure 6D). All these events were prevented by Bcl-2 overexpression in U937 cells (Figure 6A-D; Table 1). Smac/Diablo protein was released in the cytosol on As2O3 treatment more efficiently than on lonidamine exposure. Again, this effect was not prevented by Bcl-2 overexpression (Figure 6B). Because As2O3-induced apoptosis has been shown to be modulated by the cellular GSH redox system, with increased intracellular levels of reduced GSH having an inhibitory effect,30 we measured the influence of TPA-induced differentiation on cellular GSH and ROS content in untreated and As2O3-treated cells. By using flow cytometry methods, we observed that the differentiation process was associated with a decrease in the basal level of GSH (Figure7A) and an increase in the basal ROS content in the cells (Figure 7B). Arsenic exposure decreased GSH content and increased ROS level in parental cells. These effects were prevented by Bcl-2 overexpression. In differentiated cells, As2O3 treatment further decreased GSH content without increasing the cellular ROS content, as measured by using dihydro-ethidium (Figure 7A-B).

Fig. 6.

TPA-induced differentiation sensitizes U937 cells to arsenic trioxide (As2O3)-induced differentiation.

(A) Undifferentiated (▴), TPA-differentiated (●), and Bcl-2 overexpressing U937 cells (▪, U-BCL2) were treated with increasing concentrations of As2O3 for 72 hours (left panel) or with 4 μM As2O3 for indicated times (right panel). Apoptotic DNA fragmentation was quantified with the use of a filter elution assay. Values from 1 of 2 independent experiments are shown (mean ± SD of triplicate samples). (B) Western blot analysis of cytochrome c and Smac/Diablo expression in cytosolic fractions from undifferentiated (U937) and TPA-differentiated (U-TPA) U937 cells and U937 cells overexpressing Bcl-2 (U-BCL2) treated with 4 μM As2O3 for indicated times. (C) Mitochondrial membrane depolarization was visualized by flow cytometry in U937, U-TPA, and U-BCL2 cells treated with 4 μM As2O3 for indicated times. Mitochondrial depolarization was identified by an increased level of cytosolic green monomer (FL1-H; dashed lines indicate the controls). (D) Western blot analysis of procaspase-3 and caspase-3 active fragments and PARP expression in whole-cell extracts from U937, U-TPA, and U-BCL2 cells treated with 4 μM As2O3 for indicated times. Numbers are molecular weights in kilodaltons. *Cleavage products. Loading was checked with the use of an antihuman HSC70 mAb.

Fig. 6.

TPA-induced differentiation sensitizes U937 cells to arsenic trioxide (As2O3)-induced differentiation.

(A) Undifferentiated (▴), TPA-differentiated (●), and Bcl-2 overexpressing U937 cells (▪, U-BCL2) were treated with increasing concentrations of As2O3 for 72 hours (left panel) or with 4 μM As2O3 for indicated times (right panel). Apoptotic DNA fragmentation was quantified with the use of a filter elution assay. Values from 1 of 2 independent experiments are shown (mean ± SD of triplicate samples). (B) Western blot analysis of cytochrome c and Smac/Diablo expression in cytosolic fractions from undifferentiated (U937) and TPA-differentiated (U-TPA) U937 cells and U937 cells overexpressing Bcl-2 (U-BCL2) treated with 4 μM As2O3 for indicated times. (C) Mitochondrial membrane depolarization was visualized by flow cytometry in U937, U-TPA, and U-BCL2 cells treated with 4 μM As2O3 for indicated times. Mitochondrial depolarization was identified by an increased level of cytosolic green monomer (FL1-H; dashed lines indicate the controls). (D) Western blot analysis of procaspase-3 and caspase-3 active fragments and PARP expression in whole-cell extracts from U937, U-TPA, and U-BCL2 cells treated with 4 μM As2O3 for indicated times. Numbers are molecular weights in kilodaltons. *Cleavage products. Loading was checked with the use of an antihuman HSC70 mAb.

Close modal
Fig. 7.

Intracellular GST and ROS levels.

Analysis by flow cytometry of (A) intracellular GST and (B) ROS levels of undifferentiated (U937), TPA-differentiated (U-TPA), and Bcl-2 overexpressing (U-BCL2) U937 cells treated with 4 μM As2O3 for indicated times.

Fig. 7.

Intracellular GST and ROS levels.

Analysis by flow cytometry of (A) intracellular GST and (B) ROS levels of undifferentiated (U937), TPA-differentiated (U-TPA), and Bcl-2 overexpressing (U-BCL2) U937 cells treated with 4 μM As2O3 for indicated times.

Close modal

Lonidamine and arsenic trioxide also bypass the resistance of TPA-differentiated HL60 cells to classical chemotherapeutic drugs

The results obtained in U937 cells were reproduced in HL60 cells whose exposure to TPA also induces a macrophagic-like differentiation process with a resistant phenotype to classical anticancer drugs.17 TPA-differentiated HL60 cells, which were resistant to etoposide-induced apoptosis, demonstrated a sensitivity to lonidamine that was only slightly delayed compared to that of parental cells, whereas their sensitivity to As2O3-induced cell death was greatly increased (Figure 8; Table 1).

Fig. 8.

Lonidamine and arsenic trioxide bypass the resistance of TPA-differentiated HL60 cells to etoposide.

Undifferentiated (▴, HL60), TPA-differentiated (●, HL-TPA), and Bcl-2 overexpressing (▪, HL-BCL2) HL60 cells were treated with 50 μM etoposide (VP16), 200 μM lonidamine (LND), or 4 μM arsenic trioxide (As2O3) for indicated times. (A) Apoptotic DNA fragmentation was quantified with the use of a filter elution assay. Values from 1 of 2 independent experiments are shown (mean ± SD of triplicate samples). (B) Western blot analysis of PARP cleavage in whole-cell extracts from HL60, HL-TPA, and HL-BCL2 cells, either untreated (−) or treated (+) with 50 μM VP16 for 6 hours, 200 μM LND for 48 hours, or 4 μM As2O3 for 48 hours. Numbers are molecular weights in kilodaltons. *Cleavage product. Loading was checked with the use of an antihuman HSC70 mAb.

Fig. 8.

Lonidamine and arsenic trioxide bypass the resistance of TPA-differentiated HL60 cells to etoposide.

Undifferentiated (▴, HL60), TPA-differentiated (●, HL-TPA), and Bcl-2 overexpressing (▪, HL-BCL2) HL60 cells were treated with 50 μM etoposide (VP16), 200 μM lonidamine (LND), or 4 μM arsenic trioxide (As2O3) for indicated times. (A) Apoptotic DNA fragmentation was quantified with the use of a filter elution assay. Values from 1 of 2 independent experiments are shown (mean ± SD of triplicate samples). (B) Western blot analysis of PARP cleavage in whole-cell extracts from HL60, HL-TPA, and HL-BCL2 cells, either untreated (−) or treated (+) with 50 μM VP16 for 6 hours, 200 μM LND for 48 hours, or 4 μM As2O3 for 48 hours. Numbers are molecular weights in kilodaltons. *Cleavage product. Loading was checked with the use of an antihuman HSC70 mAb.

Close modal

Classical chemotherapeutic drugs induce apoptosis by compromising mitochondrial function in an indirect fashion—for example, by inducing DNA damage that triggers p53 activation and interferes with cell cycle progression31 by stimulating the Fas-dependent pathway,19,32 by triggering ceramide production,33 and by inducing shifts in redox potentials.1 A series of new chemotherapeutic drugs, many of them currently tested in clinical trials, have been shown to act, at least in part, by directly targeting the mitochondria, as demonstrated by their ability to activate the permeability transition pore complex and to trigger the release of soluble intermembrane proteins from purified mitochondria in vitro.16 This observation led to the theoretical suggestion that these compounds may bypass the resistance to anticancer drugs induced by alterations of indirect death-inducing pathways. The current study demonstrates that TPA-mediated differentiation of leukemic cells reproduces a situation in which the pathway to cell death, triggered by a classical anticancer drug such as etoposide, is inhibited upstream of the mitochondrial events. Analysis of this model system confirms the hypothesis that mitochondria-targeting drugs can overcome such a resistant phenotype.

TPA-mediated inhibition of etoposide-induced apoptosis increased as leukemic cells progressed toward the macrophagic differentiation phenotype. Short-term effects of TPA are mainly the activation of several protein kinase C (PKC) isoenzymes.34-36 The differentiation process involves several events downstream of the transient activation of PKCs.37 Thus, the differentiation-related resistance of leukemic cells to etoposide-induced cell death might be distinguished from the direct consequences of PKC activation. A differentiation-related resistance to apoptosis induced by various stimuli has been described in leukemic cells exposed to either 1,25-dihydroxyvitamin D338,39 or DMSO.21 A resistance to growth factor deprivation–induced cell death was also identified in differentiated myocytes.40 Thus, several differentiation processes in different cell types were associated with a decreased sensitivity to apoptosis induced by a series of distinct stimuli.

Most pathways to apoptosis converge on the mitochondrial release of apoptogenic molecules to the cytosol and the activation of the caspase cascade.1,4,5 The current study confirms that etoposide activates a variety of caspases while inducing apoptosis of undifferentiated U937 cells. Various mechanisms negatively interfere with this final pathway, including overexpression of antiapoptotic proteins of the Bcl-2 family that prevents the cytosolic release of mitochondrial soluble molecules7,41 and overexpression of heat-shock proteins such as HSP27 and HSP70 that prevent cytochrome c–mediated activation of the caspase cascade by interacting with cytochrome c24 and the Apaf-1 adaptor molecule,42 respectively. Despite the fact that the TPA-induced differentiation process induces some changes in the subcellular localization of procaspases (O.S. et al, manuscript in preparation) and modulates the expression of inhibitory proteins—for example, by inducing transient changes in the expression of the Bcl-2–related protein Mcl-118,43—our data, obtained by using cell-free systems, demonstrate that the differentiation process does not significantly influence the mitochondrial pathway to cell death.

Lonidamine (1-[(2,4-dichlorophenylmethyl) methyl]-1H-indazole-3-carboxylic acid]) is an antineoplastic drug derived from indazole-3-carboxylic acid. This compound, which was shown to exacerbate the response of human tumor cells to cisplatin, irradiation, doxorubicin, or cyclophosphamide, has been tested in phase II and III trials of metastatic breast cancer and ovarian cancer.44-48 Although several distinct mechanisms were proposed to account for lonidamine cytotoxic effects,49,50recent data suggest that mitochondria could be the main subcellular target of this compound. Lonidamine directly acts on the permeability transition pore complex when tested on purified organelles, an effect inhibited by Bcl-2.27 Since we have shown that the mitochondrial pathway to cell death remains unchanged in TPA-differentiated cells, lonidamine may have demonstrated a similar efficacy on undifferentiated and TPA-differentiated leukemic cells. Actually, TPA-induced differentiation slightly, but reproducibly, delays lonidamine-induced apoptosis in U937 and HL60 cells, suggesting a negative effect of the differentiation process on some other potential consequences of lonidamine treatment, such as cytosolic calcium increase and intracellular lactate accumulation. This effect of the differentiation process on lonidamine-induced cell death remains limited compared with the strong inhibition of apoptosis triggered by classical anticancer drugs.

As2O3 is another therapeutic compound that was suggested to directly target the mitochondria.28,29Although As2O3-induced apoptosis was initially described in acute promyelocytic cell lines,51,52 this compound induces the death of many other leukemia cell types.53-56 The hypothesis that As2O3 directly targets the mitochondria is based on the observation that exposure of purified organelles to arsenic triggers opening of the permeability transition pore and the release of soluble intermembrane proteins.28 In whole cells, As2O3 induces disruption of the mitochondrial transmembrane potential Δψm51 and caspase activation,56,57 2 events that are inhibited by Bcl-2 overexpression.28As2O3-induced apoptosis was also related to Bcl-2 down-regulation,52,56,58 Bax up-regulation,59 suppression of the apoptosis inhibitory Ras/MAP kinase cascade,60,61 and enhanced translocation of PML or PML/RARα chimeric proteins to nuclear bodies,62but these events depend on As2O3 dose, cell type, and cellular environment. We show here that TPA-induced differentiation of leukemic cells increases the ability of As2O3 to target the mitochondria, to induce the release of cytochrome c and Smac/Diablo, to activate downstream caspases, and to induce apoptotic cell death. A similar synergy was described between As2O3 and retinoic acid, another differentiating agent, in an arsenic-resistant NB4 subline.63,64 The differentiation-induced modulation of leukemic cell sensitivity to As2O3-induced apoptosis could account for the conflicting reports concerning the sensitivity of cell lines such as HL-60. Arsenic toxicity, including its mitochondrial effects, presumably results from forming reversible bonds with the thiol groups of proteins.65 Intracellular level of GSH, which potentially titrates intracellular arsenic by formation of transient As(GS)3 complexes, is a major determinant of As2O3-triggered cell death system, with increased intracellular levels of GSH having an inhibitory effect.30,66 The proton donor GSH prevents arsenic from attacking its targets and increases the GSH peroxidase-catalyzed breakdown of H2O2.67 Thus, the differentiation-related decrease in GSH cellular content may contribute to the sensitization of leukemic cells to As2O3-mediated cell death.

In conclusion, we have shown that the TPA-mediated differentiation of human myeloid leukemic cell lines induces a resistance phenotype to classical anticancer drugs such as etoposide without inhibiting the mitochondrial pathway to cell death. The differentiation process has a limited influence on lonidamine-induced cell death and sensitizes leukemic cells to As2O3-mediated apoptosis (Figure 9). Although the As2O3 doses required for inducing apoptosis in differentiated cells remain high compared with clinically achieved concentrations,68 these observations suggest that chemotherapeutic drugs that directly or rapidly target the mitochondria in vitro warrant further clinical investigation in the treatment of acute leukemias. These drugs may represent an effective strategy to bypass the resistance of leukemic cells to classical anticancer drugs, or they may be used in combination with differentiating agents.

Fig. 9.

Influence of TPA-induced differentiation on drug-induced cell death.

TPA-mediated differentiation of leukemic cells strongly decreases the ability of classical anticancer drugs, such as those inducing DNA damage, to activate the mitochondrial pathway to cell death. In contrast, the differentiation process has limited influence on cell death induced by the mitochondria-targeting agent lonidamine and even sensitizes the cells to arsenic-mediated activation of the mitochondrial pathway.

Fig. 9.

Influence of TPA-induced differentiation on drug-induced cell death.

TPA-mediated differentiation of leukemic cells strongly decreases the ability of classical anticancer drugs, such as those inducing DNA damage, to activate the mitochondrial pathway to cell death. In contrast, the differentiation process has limited influence on cell death induced by the mitochondria-targeting agent lonidamine and even sensitizes the cells to arsenic-mediated activation of the mitochondrial pathway.

Close modal

We thank M. F. Poupon, X. Wang, and D. G. Chen for providing lonidamine, anti-Smac antibody, and As2O3, respectively.

Supported by grants from the Ligue National Contre le Cancer (Equipe labelisée “la ligue”) and the Association Régionale pour l'Enseignement et la Recherche Scientifique. O.S. is the recipient of a grant from the Fondation pour la Recherche Médicale.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Solary
E
Droin
N
Bettaieb
A
Corcos
L
Dimanche-Boitrel
MT
Garrido
C
Positive and negative regulation of apoptotic pathways by cytotoxic agents in hematological malignancies.
Leukemia.
14
2000
1833
1849
2
Schuler
D
Szende
B
Borsi
JD
et al
Apoptosis as a possible way of destruction of lymphoblasts after glucocorticoid treatment of children with acute lymphoblastic leukemia.
Pediatr Hematol Oncol.
11
1994
641
649
3
Nicholson
DW
Caspase structure, proteolytic substrates, and function during apoptotic cell death.
Cell Death Differ.
6
1999
1028
1042
4
Slee
EA
Adrain
C
Martin
SJ
Serial killers: ordering caspase activation events in apoptosis.
Cell Death Differ.
6
1999
1067
1074
5
Kroemer
G
Reed
JC
Mitochondrial control of cell death.
Nat Med.
6
2000
513
519
6
Reed
JC
Cytochrome c: can't live with it—can't live without it.
Cell.
91
1997
559
562
7
Susin
SA
Lorenzo
HK
Zamzami
N
et al
Molecular characterization of mitochondrial apoptosis-inducing factor.
Nature.
397
1999
441
446
8
Verhagen
AM
Ekert
PG
Pakusch
M
et al
Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins.
Cell.
102
2000
43
53
9
Du
C
Fang
M
Li
Y
Li
L
Wang
X
Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition.
Cell.
102
2000
33
42
10
Li
P
Nijhawan
D
Budihardjo
I
et al
Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade.
Cell.
91
1997
479
489
11
Hu
Y
Benedict
MA
Ding
L
Nunez
G
Role of cytochrome c and dATP/ATP hydrolysis in Apaf-1–mediated caspase-9 activation and apoptosis.
EMBO J.
18
1999
3586
3595
12
Slee
EA
Harte
MT
Kluck
RM
et al
Ordering the cytochrome c-initiated caspase cascade: hierarchical activation of caspases-2, -3, -6, -7, -8, and -10 in a caspase-9–dependent manner.
J Cell Biol.
144
1999
281
292
13
Ghayur
T
Hugunin
M
Talanian
RV
et al
Proteolytic activation of protein kinase C delta by an ICE/CED 3-like protease induces characteristics of apoptosis.
J Exp Med.
184
1996
2399
2404
14
Liu
X
Zou
H
Slaughter
C
Wang
X
DFF, a heterodimeric protein that functions downstream of caspase-3 to trigger DNA fragmentation during apoptosis.
Cell.
89
1997
175
184
15
Eymin
B
Sordet
O
Droin
N
et al
Caspase-induced proteolysis of the cyclin-dependent kinase inhibitor p27Kip1 mediates its antiapoptotic activity.
Oncogene.
18
1999
4839
4847
16
Costantini
P
Jacotot
E
Decaudin
D
Kroemer
G
Mitochondrion as a novel target of anticancer compounds.
J Natl Cancer Inst.
5
2000
1042
1053
17
Solary
E
Bertrand
R
Kohn
K
Pommier
Y
Differential induction of apoptosis in undifferentiated and differentiated HL60 cells by DNA topoisomerase I and II inhibitors.
Blood.
81
1993
1359
1368
18
Sordet
O
Bettaieb
A
Bruey
JM
et al
Selective inhibition of apoptosis by TPA-induced differentiation of U937 leukemic cells.
Cell Death Differ.
6
1999
351
361
19
Micheau
O
Solary
E
Hammann
A
Dimanche-Boitrel
MT
Fas ligand-independent, FADD-mediated activation of the Fas death pathway by anticancer drugs.
J Biol Chem.
274
1999
7987
7992
20
Xu
HM
Tepper
CG
Jones
JB
Fernandez
CE
Studzinski
GP
1,25-Dihydroxyvitamin D3 protects HL-60 cells against apoptosis but down-regulates the expression of the bcl-2 gene.
Exp Cell Res.
209
1993
367
374
21
Shiiki
K
Yoshikawa
H
Kinoshita
H
et al
Potential mechanisms of resistance to TRAIL/Apo2L-induced apoptosis in human promyelocytic leukemia HL-60 cells during granulocytic differentiation.
Cell Death Differ.
7
2000
939
946
22
Bertrand
R
Kohn
KW
Solary
E
Pommier
Y
Detection of apoptosis-associated DNA fragmentation using a rapid and quantitative filter binding assay.
Drug Dev Res.
34
1995
138
144
23
Miller
SA
Dykes
DD
Polesky
HF
A simple salting out procedure for extracting DNA from human nucleated cells [abstract].
Nucleic Acids Res.
16
1988
1215
24
Bruey
JM
Ducasse
C
Bonniaud
P
et al
Hsp27 negatively regulates cell death by interacting with cytochrome c.
Nat Cell Biol.
2
2000
645
652
25
Lizard
G
Gueldry
S
Sordet
O
et al
Glutathione is implied in the control of 7-ketocholesterol– induced apoptosis, which is associated with radical oxygen species production.
FASEB J.
12
1998
1651
1663
26
Srinivasula
SM
Ahmad
M
Guo
Y
et al
Identification of an endogenous dominant-negative short isoform of caspase-9 that can regulate apoptosis.
Cancer Res.
59
1999
999
1002
27
Ravagnan
L
Marzo
I
Costantini
P
et al
Lonidamine triggers apoptosis via a direct, Bcl-2–inhibited effect on the mitochondrial permeability transition pore.
Oncogene.
18
1999
2537
2546
28
Larochette
N
Decaudin
D
Jacotot
E
et al
Arsenite induces apoptosis via a direct effect on the mitochondrial permeability transition pore.
Exp Cell Res.
249
1999
413
421
29
Kroemer
G
de The
H
Arsenic trioxide, a novel mitochondriotoxic anticancer agent?
J Natl Cancer Inst.
91
1999
743
745
30
Dai
J
Weinberg
RS
Waxman
S
Jing
Y
Malignant cells can be sensitized to undergo growth inhibition and apoptosis by arsenic trioxide through modulation of the glutathione redox system.
Blood.
93
1999
268
277
31
Dubrez
L
Goldwasser
F
Genne
P
Pommier
Y
Solary
E
The role of cell cycle regulation and apoptosis triggering in determining the sensitivity of leukemic cells to topoisomerase I and II inhibitors.
Leukemia.
9
1995
1013
1024
32
Fulda
S
Strauss
G
Meyer
E
Debatin
KM
Functional CD95 ligand and CD95 death-inducing signaling complex in activation-induced cell death and doxorubicin-induced apoptosis in leukemic T cells.
Blood.
95
2000
301
308
33
Jaffrezou
JP
Levade
T
Bettaieb
A
et al
Daunorubicin-induced apoptosis: triggering of ceramide generation through sphingomyelin hydrolysis.
EMBO J.
5
1996
2417
2424
34
Kizaki
H
Tadakuma
T
Odaka
C
Muramatsu
J
Ishimura
Y
Activation of a suicide process of thymocytes through DNA fragmentation by calcium ionophores and phorbol esters.
J Immunol.
143
1989
1790
1794
35
Gubina
E
Rinaudo
MS
Szallasi
Z
Blumberg
PM
Mufson
RA
Overexpression of protein kinase C isoform ε but not δ in human interleukin-3–dependent cells suppresses apoptosis and induces bcl-2 expression.
Blood.
91
1998
823
829
36
Mayne
GC
Murray
AW
Evidence that protein kinase C epsilon mediates phorbol ester inhibition of calphostin C- and tumor necrosis factor-alpha–induced apoptosis in U937 histiocytic lymphoma cells.
J Biol Chem.
273
1998
24115
24121
37
Seimiya
H
Sawabe
T
Toho
M
Tsuruo
T
Phorbol ester-resistant monoblastoid leukemia cells with a functional mitogen-activated protein kinase cascade but without responsive protein tyrosine phosphatases.
Oncogene.
11
1995
2047
2054
38
Wang
X
Studzinski
GP
Antiapoptotic action of 1,25-dihydroxyvitamin D3 is associated with increased mitochondrial MCL-1 and RAF-1 proteins and reduced release of cytochrome c.
Exp Cell Res.
235
1997
210
217
39
Kikuchi
H
Lizuka
R
Sugiyama
S
et al
Monocytic differentiation modulates apoptotic response to cytotoxic anti-Fas antibody and tumor necrosis factor alpha in human monoblast U937 cells.
J Leukoc Biol.
60
1996
778
783
40
Wang
J
Walsh
K
Resistance to apoptosis conferred by Cdk inhibitors during myocyte differentiation.
Science.
273
1996
359
361
41
Kluck
RM
Bossy-Wetzel
E
Green
DR
Newmeyer
DD
The release of cytochrome c from mitochondria: a primary site for Bcl-2 regulation of apoptosis.
Science.
275
1997
1132
1136
42
Saleh
A
Srinivasula
SM
Balkir
L
Robbins
PD
Alnemri
ES
Negative regulation of the apaf-1 apoptosome by hsp70.
Nat Cell Biol.
2
2000
476
483
43
Moulding
DA
Giles
RV
Spiller
DG
White
MR
Tidd
DM
Edwards
SW
Apoptosis is rapidly triggered by antisense depletion of MCL-1 in differentiating U937 cells.
Blood.
96
2000
1756
1763
44
Gebbia
V
Borsellino
N
Testa
A
et al
Cisplatin and epirubicin plus oral lonidamine as first-line treatment for metastatic breast cancer: a phase II study of the Southern Italy Oncology Group (GOIM).
Anticancer Drugs.
8
1997
943
948
45
Dudak
SD
Lopez
A
Block
NL
Lokeshwar
BL
Enhancement of radiation response of prostatic carcinoma by lonidamine.
Anticancer Res.
16
1996
3665
3671
46
Amadori
D
Frassineti
GL
De Matteis
A
et al
Modulating effect of lonidamine on response to doxorubicin in metastatic breast cancer patients: results from a multicenter prospective randomized trial.
Breast Cancer Res Treat.
49
1998
209
217
47
Gardin
G
Barone
C
Nascimben
O
et al
Lonidamine plus epirubicin and cyclophosphamide in advanced breast cancer: a phase II study.
Eur J Cancer.
32A
1996
176
177
48
Silvestrini
R
Zaffaroni
N
Villa
R
Orlandi
L
Costa
A
Enhancement of cisplatin activity by lonidamine in human ovarian cancer cells.
Int J Cancer.
52
1992
813
817
49
Ben-Horin
J
Tassini
M
Vivi
A
Navon
G
Kaplan
O
Mechanisms of action of the antineoplastic drug lonidamine: 31P and 13C nuclear magnetic resonance studies.
Cancer Res.
55
1995
2814
2821
50
Floridi
A
Paggi
MG
D'Atri
S
et al
Effect of lonidamine on the energy metabolism of Ehrlich ascites tumor cells.
Cancer Res.
41
1981
4661
4666
51
Cai
X
Shen
YL
Zhu
Q
et al
Arsenic trioxide-induced apoptosis and differentiation are associated respectively with mitochondrial transmembrane potential collapse and retinoic acid signaling pathways in acute promyelocytic leukemia.
Leukemia.
14
2000
262
270
52
Chen
GQ
Zhu
J
Shi
XG
et al
In vitro studies on cellular and molecular mechanisms of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia: As2O3 induces NB4 cell apoptosis with down-regulation of Bcl-2 expression and modulation of PML-RAR alpha/PML proteins.
Blood.
88
1996
1052
1061
53
Wang
ZG
Rivi
R
Delva
L
et al
Arsenic trioxide and melarsoprol induce programmed cell death in myeloid leukemia cell lines and function in a PML and PML-RAR alpha independent manner.
Blood.
92
1998
1497
1504
54
Huang
XJ
Wiernik
PH
Klein
RS
Gallagher
RE
Arsenic trioxide induces apoptosis of myeloid leukemia cells by activation of caspases.
Med Oncol.
16
1999
58
64
55
Bazarbachi
A
El-Sabban
ME
Nasr
R
et al
Arsenic trioxide and interferon-alpha synergize to induce cell cycle arrest and apoptosis in human T-cell lymphotropic virus type I-transformed cells.
Blood.
93
1999
278
283
56
Akao
Y
Mizoguchi
H
Kojima
S
Naoe
T
Ohishi
N
Yagi
K
Arsenic induces apoptosis in B-cell leukaemic cell lines in vitro: activation of caspases and down-regulation of Bcl-2 protein.
Br J Haematol.
102
1998
1055
1060
57
Soignet
SL
Maslak
P
Wang
ZG
et al
Complete remission after treatment of acute promyelocytic leukemia with arsenic trioxide.
N Engl J Med.
339
1998
1341
1348
58
Konig
A
Wrazel
L
Warrell
RP
Jr
et al
Comparative activity of melarsoprol and arsenic trioxide in chronic B-cell leukemia lines.
Blood.
90
1997
562
570
59
Kinjo
K
Kizaki
M
Muto
A
et al
Arsenic trioxide (As2O3)-induced apoptosis and differentiation in retinoic acid-resistant acute promyelocytic leukemia model in hGM-CSF–producing transgenic SCID mice.
Leukemia.
14
2000
431
438
60
Doza
YN
Hall-Jackson
CA
Cohen
P
Arsenite blocks growth factor induced activation of the MAP kinase cascade, upstream of Ras and downstream of Grb2-Sos.
Oncogene.
17
1998
19
24
61
Ludwig
S
Hoffmeyer
A
Goebeler
M
et al
The stress inducer arsenite activates mitogen-activated protein kinases extracellular signal-regulated kinases 1 and 2 via a MAPK kinase 6/p38-dependent pathway.
J Biol Chem.
273
1998
1917
1922
62
Quignon
F
De Bels
F
Koken
M
Feunteun
J
Ameisen
JC
de The
H
PML induces a novel caspase-independent death process.
Nat Genet.
20
1998
259
265
63
Gianni
M
Koken
MH
Chelbi-Alix
MK
et al
Combined arsenic and retinoic acid treatment enhances differentiation and apoptosis in arsenic-resistant NB4 cells.
Blood.
91
1998
4300
4310
64
Jing
Y
Wang
L
Xia
L
et al
Combined effect of all-trans retinoic acid and arsenic trioxide in acute promyelocytic leukemia cells in vitro and in vivo.
Blood.
97
2001
264
269
65
Watson
RW
Redmond
HP
Wang
JH
Bouchier-Hayes
D
Mechanisms involved in sodium arsenite-induced apoptosis of human neutrophils.
J Leukoc Biol.
60
1996
625
632
66
Scott
N
Hatlelid
KM
MacKenzie
NE
Carter
DE
Reactions of arsenic(III) and arsenic(V) species with glutathione.
Chem Res Toxicol.
6
1993
102
106
67
Jing
Y
Dai
J
Chalmers-Redman
RM
Tatton
WG
Waxman
S
Arsenic trioxide selectively induces acute promyelocytic leukemia cell apoptosis via a hydrogen peroxide-dependent pathway.
Blood.
94
1999
2102
2111
68
Shen
ZX
Chen
GQ
Ni
JH
et al
Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL), II: clinical efficacy and pharmacokinetics in relapsed patients.
Blood.
89
1997
3354
3360

Author notes

Eric Solary, Faculty of Medicine and Pharmacy, INSERM U517, 7 boulevard Jeanne d'Arc, 21033 Dijon, France; e-mail: esolary@u-bourgogne.fr.

Sign in via your Institution