The p210bcr-abl protein was shown to inhibit apoptosis induced by DNA damaging agents. Apoptotic DNA fragmentation is delayed in the bcr-abl+ K562 and KCL-22 compared with thebcr-abl U937 and HL-60 cell lines when treated with etoposide concentrations that induce similar DNA damage in the four cell lines. By the use of a cell-free system, we show that nuclei from untreated cells that express p210bcr-abl remain sensitive to apoptotic DNA fragmentation induced by triton-soluble extracts from p210bcr-abl− cells treated with etoposide. In the four tested cell lines, apoptotic DNA fragmentation is associated with a decreased expression of procaspase-3 (CPP32/Yama/apopain) and its cleavage into a p17 active fragment, whereas the long isoform of procaspase-2 (ICH-1L) remains unchanged and the poly(adenosine diphosphate–ribose)polymerase protein is cleaved. These events are delayed in bcr-abl+ compared with bcr-abl cell lines. The role of p210bcr-abl in this delay is confirmed by comparing the effect of etoposide on the granulocyte-macrophage colony-stimulating factor (GM-CSF)–dependent UT7 cells and thebcr-abl–transfected GM-CSF–independent UT7/9 clone. We conclude that the cytosolic pathway that leads to apoptotic DNA fragmentation in etoposide-treated leukemic cells is delayed upstream of procaspase-3–mediated events in bcr-abl+ cell lines.

THE PHILADELPHIA chromosome is the cytogenetic hallmark of chronic myelogenous leukemia (CML).1 Translocation of the c-abl gene, located on chromosome 9q, within the bcr gene, located on chromosome 22q, results in the formation of a chimeric protein, p210bcr-abl.2 This event is generally accepted to be the primary initiating event in the genesis of CML because mice transplanted with bone marrow cells infected with retroviral vectors encoding p210bcr-abl develop a CML-like disease.3 The c-ABL protein is a nuclear protein with low intrinsic tyrosine kinase activity, whereas the BCR-ABL protein is a cytoplasmic, membrane-associated protein that contains a constitutively high level of tyrosine kinase activity. It was initially assumed that CML was caused by uncontrolled cell proliferation resulting in the clonal expansion evident in this disease.4 Actually, CML progenitors have similar proliferation rates as their normal counterparts.5 Recent studies have shown that both the v-abl transforming oncogene product and the p210bcr-abl prolonged hematopoietic cell survival by inhibition of apoptosis.6-8 The BCR-ABL chimeric protein confers on hematopoietic cells the ability to survive treatments such as growth factor deprivation, cytotoxic drugs, protein tyrosine kinase inhibitors, and Fas-ligand.9-12 Antisense oligonucleotides that downregulate BCR-ABL protein expression in CML cell lines either induce apoptotic cell death13 or render the cells more susceptible to cell death induction by cytotoxic drugs and Fas-ligand.9,10 The mechanisms by which the deregulated v-ABL and BCR-ABL tyrosine kinases delay apoptotic cell death remain poorly understood. Translocation of the protein kinase C βII,14 increased glucose uptake,15 delayed G2/M transition after DNA damage,9 and activation of Ras functions16 were proposed to mediate this inhibitory effect, whereas the MAP kinase kinase/MAP kinase pathway was shown not to be required for BCR-ABL–mediated suppression of apoptosis.17 

Transduction of the apoptotic signal and execution of apoptosis require the coordinate action of several aspartate-specific cysteine proteases, the so-called caspases. The 10 human caspases identified so far can be divided into four subfamilies based on their structure and their homology to the human prototype interleukin-1β converting enzyme (ICE) and the nematode prototype CED-3.18 These subfamilies are the ICE-like caspases, the CED-3–like caspases, the caspases that contain prodomains highly related to the death effector domain of the Fas/APO-1 receptor, and the NEDD2/ICH-1 caspases. All these enzymes are initially synthesized as single-chain inactive proenzymes that require cleavage after aspartate residues to obtain the active protease. Under physiological conditions, this process probably involves heterotypic protein-protein interactions and a cascade of caspases.19Although molecular ordering of this cascade is only partly known, it seems that several apoptotic pathways probably exist. For example, deletion of the ICE (caspase-1) gene inhibits Fas-induced apoptosis without modifying several other apoptotic pathways.20 By contrast, activation of CED-3–like caspases could be common to most apoptotic pathways.21 Distinct from ICE, these caspases cleave the cell death substrate poly(adenosine diphosphate [ADP]–ribose) polymerase (PARP) into signature apoptotic fragments.22-24 Activation of CED-3–related caspases was shown recently to be controlled by the CED-9/Bcl-2 family of molecules. This control was suggested to take place at the mitochondrial membrane25 and to involve the so-far unidentified mammalian homolog of the nematode protein CED-4.26 

In the present study, we addressed the question of the relationship between BCR-ABL–mediated inhibition of apoptosis and the activation of CED-3–related caspases. As a model system, we used etoposide (VP-16)-induced apoptosis. This topoisomerase II-reactive agent produces double-strand breaks by stabilizing a transient intermediate of the topoisomerase reaction.27 We have shown previously that etoposide induced similar levels of these DNA damage inbcr-abl+ (K562, KCL-22) andbcr-abl (HL-60, U937) cell lines.28However, apoptosis was strongly delayed in bcr-abl+cells. Here, we show that this delay occurs upstream of procaspase-3 activation.

Drug and chemicals.

Etoposide was obtained from Sigma-Aldrich laboratories (St Quentin Fallavier, France). Stock solution (20 mmol/L) in dimethyl sulfoxide (DMSO) was conserved at −20°C for less than 1 month. Further dilutions were made in culture medium just before use. The final concentration of DMSO in culture did not exceed 1% (vol/vol), which was nontoxic to the cells. [2-14C]thymidine (50 mCi/mmol) was obtained from Amersham (Les Ulis, France). All other chemicals were purchased from local sources.

Cell lines and culture.

The four human leukemic cell lines, HL60 (promyelocytic), U937 (monocytic), K562 (CML in erythroblastic blast crisis), and KCL22 (CML in lymphoid blast crisis) were grown in suspension in RPMI 1640 medium (BioWhittaker, Fontenay-sous-bois, France) supplemented with 10% (vol/vol) heat-inactivated fetal bovine serum and 2 mmol/L L-glutamine in an atmosphere of 95% air and 5% CO2. The human pluripotent UT-7 line, established from a patient with megakaryocytic leukemia, was cultured in the presence of 10 ng/mL granulocyte-macrophage colony-stimulating factor (GM-CSF). Transfer ofbcr-abl gene into UT-7 cells was accomplished using a defective amphotropic p210 retrovirus. The GM-CSF–independent UT7/9 clone was selected for its high expression of BCR-ABL mRNA as previously reported.29 Cell viability was determined using the trypan blue exclusion test. Cells were resuspended at a density of 1.5 × 106/mL in fresh medium before treatment.

Reconstituted cell-free system.

Triton-soluble extracts and nuclear fractions were prepared as previously described.30 Briefly, cells were washed twice in 10 mL ice-cold phosphate-buffered saline (PBS) without Ca2+and Mg2+, and once in a buffer containing 150 mmol/L NaCl, 1 mmol/L KH2PO4, 1 mmol/L EGTA, 1 mmol/L Na3VO4, 5 mmol/L MgCl2, and 10% glycerol (pH 7.2). Then, the cells were incubated for 10 minutes on ice in the same buffer containing 0.3% Triton X-100 (Sigma, Aldrich, France) before centrifugation (2,000g for 10 minutes at 4°C) and the supernatants collected were considered as triton-soluble extracts. Protein concentration in the supernatants was determined using the bicinchoninic acid method.31 Pellets (nuclei) were washed twice in the lysis buffer without Triton X-100. Triton-soluble extracts (500 μL) from untreated or treated cells were incubated for 30 minutes at 37°C in the presence of nuclei from 1.0 × 106 untreated cells and DNA fragmentation was measured as described below. Controls were performed by incubating nuclei from untreated cells with triton-soluble extracts from untreated cells, in the absence or in the presence of 100 μmol/L etoposide.

Quantification of DNA fragmentation.

DNA fragmentation was measured using a previously reported filter elution assay.32 Exponentially growing cells were prelabeled by adding 0.02 μCi/mL of [2-14C]thymidine in the culture medium for 2 days. Approximately 1.0 × 106 [14C]-labeled cells or [14C]-labeled nuclei were loaded onto a protein absorbing filter (Polyvinylidene fluorure filters, 0.65 μM pore size, 25 mm diameter; Durapore membrane, Millipore, St Quentin, France). After washing, lysis was performed with 5 mL of LS10 buffer (0.2% sodium sarkosyl, 2 mol/L NaCl, 0.04 mol/L EDTA, pH 10.0). Filters were washed with 7 mL of 0.02 mol/L EDTA, pH 10. DNA was depurinated by adding 0.4 mL of 1N HCl at 65°C for 45 minutes, then released from the filters by adding 2.5 mL of 0.4 N 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 in the lysis fraction plus EDTA wash relative to the total intracellular dpm.

Analysis of DNA fragmentation by agarose gel electrophoresis.

Cellular DNA was extracted by a salting-out procedure as described previously.33 Electrophoresis was performed in a 1.8% agarose gel in Tris-borate-EDTA buffer (pH 8) at 20 V for 15 hours. After electrophoresis, DNA was visualised by ethidium bromide staining.

Western blot analysis.

After treatment, cells were washed twice in PBS, lysed in lysis buffer (150 mmol/L NaCl, 1 mmol/L KH2PO4, 1 mmol/L EGTA, 1 mmol/L Na3VO4, and 5 mmol/L MgCl2; and 10% glycerol containing PmsF 0.1 mmol/L, Aprotinin 0.15 U/mL, and Pepstatin 1 μg/mL), and then centrifugated (15 minutes, 15000g). Fifty micrograms of proteins of supernatants were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) by using a 12% polyacrylamide gel and electroblotted to PVDF membrane (BioRad, Ivry sur Seine, France). After blocking nonspecific binding sites, the membrane was incubated for 2 hours at room temperature with anti-human CPP32 or Ich-1L monoclonal antibody (MoAb) (Transduction Laboratories, Lexington, KY), washed, and further incubated with horseradish peroxidase-conjugated goat anti-mouse antibody (Jackson Immunoresearch Laboratories, West Grove, PA) for 30 minutes at room temperature. Immunoblot was revealed by using enhanced chemiluminescence detection kit (Amersham) by autoradiography.

For the PARP and CPP32 cleavage analyses, cells were lysed in SDS-PAGE sample buffer (125 mmol/L Tris-HCl, pH 6.8; 10% β-mercaptoethanol; 2% SDS; 20% glycerol; and 0.003% bromophenol blue) and cell lysates were subjected to SDS-PAGE on a 8% polyacrylamide gel. The 116-kD native PARP protein and its 85-kD cleavage product were detected by immunoblotting with anti-human PARP polyclonal antibody (Vic. 5; kindly given by Dr G. De Murcia, Ecole supérieure de biotechnologies, Strasbourg, France) and horseradish-peroxidase conjugated anti-rabbit antibody (Amersham) as described above. A rabbit polyclonal anti-apopain/CPP32-p17 antibody (kindly provided by Dr D. Nicholson, Merck Center for Therapeutic Research, Pointe Claire, Daval, Quebec, Canada) that recognizes both CPP32 proenzyme and its p19 and p17 subunits34 was used to detect CPP32 activation in apoptotic U937 cells.

Etoposide-induced apoptotic DNA fragmentation is delayed inbcr-abl+ cell lines.

We used a filter elution assay32 to measure apoptotic DNA fragmentation induced by continuous exposure to etoposide in the four human leukemic cell lines. In HL-60 and U937 cells, apoptotic DNA fragmentation appeared rapidly, beginning after 3 hours of exposure to 100 μmol/L etoposide, and increased dramatically because 80% to 100% of DNA was cleaved after 24 hours of drug exposure (Fig1A). Conversely, in K562 and KCL-22 cells, apoptotic DNA fragmentation was barely detected 24 hours after the beginning of cells exposure to 100 μmol/L etoposide (Fig 1A). DNA fragmentation was confirmed to be internucleosomal by agarose gel electrophoresis (Fig 1B). We had previously shown that the levels of DNA double-strand breaks induced by this concentration of etoposide were similar in the four cell lines.28 This suggested that the apoptotic pathway was delayed downstream of DNA double-strand breaks induction in the two bcr-abl+ resistant cell lines.

Fig. 1.

Etoposide-induced apoptotic DNA fragmentation and expression of Bcl-2/Bax proteins in four human leukemic cell lines. (A) DNA fragmentation was measured by a filter elution assay in HL60 (•), U937 (▪), K562 (○), and KCL-22 (▵) cell lines treated for 24 hours with indicated concentrations of etoposide (VP-16). (B) DNA fragmentation was identified as internucleosomal by agarose gel electrophoresis. Cells were treated for indicated times with 100 μmol/L etoposide.

Fig. 1.

Etoposide-induced apoptotic DNA fragmentation and expression of Bcl-2/Bax proteins in four human leukemic cell lines. (A) DNA fragmentation was measured by a filter elution assay in HL60 (•), U937 (▪), K562 (○), and KCL-22 (▵) cell lines treated for 24 hours with indicated concentrations of etoposide (VP-16). (B) DNA fragmentation was identified as internucleosomal by agarose gel electrophoresis. Cells were treated for indicated times with 100 μmol/L etoposide.

Close modal
Delayed apoptosis in bcr-abl+ cell lines is not related to Bcl-2 and Bax expression.

Bcl-2 overexpression was shown to delay drug-induced apoptosis in a variety of cell systems. Moreover, the Bcl-2 to Bax ratio was shown to determine the fate of tumor cell treatment with various cytotoxic drugs.35,36 Therefore, we analyzed the expression of Bcl-2 and Bax proteins in the tested cell lines (Fig 2). By using Western blot analysis, Bcl-2 protein was not detected in K562 cells, whereas its level was rather similar in the three other cell lines. Bax protein level was similar in the four cell lines (Fig 2). We also analyzed the expression of the Bcl-2–related protein Mcl-1, whose expression is prominent in hematopoietic cells. After 3 hours of treatment with etoposide, Bcl-2 and Mcl-1 expressions were slightly decreased in HL-60 and KCL-22 cells, whereas it was unchanged in U937 cells and Bax remained stable in the four cell lines (Fig 2B). These results indicated that the delay in etoposide-induced apoptosis in bcr-abl+ compared with bcr-abl cell lines could not be related to the Bcl-2 to Bax ratio, nor to Mcl-1 expression.

Fig. 2.

Expression of Bcl-2, Bax, and Mcl-1 in untreated and etoposide-treated leukemic cell lines. (A) Western blot analysis of Bcl-2, Bax, and Mcl-1 basal expression in the four studied cell lines. (B) Western blot analysis of Bcl-2, Bax, and Mcl-1 expression in the four studied cell lines treated for indicated times with 100 μmol/L etoposide.

Fig. 2.

Expression of Bcl-2, Bax, and Mcl-1 in untreated and etoposide-treated leukemic cell lines. (A) Western blot analysis of Bcl-2, Bax, and Mcl-1 basal expression in the four studied cell lines. (B) Western blot analysis of Bcl-2, Bax, and Mcl-1 expression in the four studied cell lines treated for indicated times with 100 μmol/L etoposide.

Close modal
The nuclei of bcr-abl+ cell lines are sensitive to apoptotic DNA fragmentation.

To determine whether differences between bcr-abl+and bcr-abl cell lines were related to differences in the ability of cell nuclei to undergo apoptotic DNA fragmentation, we used a cell-free system in which triton-soluble extracts from etoposide-treated cell lines were incubated with14C-labeled nuclei from untreated cell lines.30,32 We observed that triton-soluble extracts from HL-60 and U937 cells treated for 3 hours with 100 μmol/L etoposide induced apoptotic DNA fragmentation in nuclei frombcr-abl+ and bcr-abl cell lines (Fig 3A through D). These results suggested that the delayed induction of apoptotic DNA fragmentation could not be related to nuclei changes in bcr-abl+cell lines. Apoptotic DNA fragmentation was lower in K562 nuclei compared with the three other cell lines. Agarose gel electrophoresis confirmed that DNA fragmentation measured in nuclei from the four studied cell lines treated with triton-soluble extracts from etoposide-treated HL-60 and U937 cells was internucleosomal DNA degradation (Fig 3E). Conversely, extracts frombcr-abl+ cells treated with etoposide in the same conditions did not induce any DNA fragmentation in nuclei from eitherbcr-abl+ or bcr-abl cells (Fig 3). Thus, the apoptotic pathway that leads to apoptotic DNA fragmentation is blocked at the cytosolic level inbcr-abl+ cells.

Fig. 3.

Apoptotic DNA fragmentation in a cell-free system. (A through D) Nuclei from the four cell lines indicated on axis in which DNA was labeled with [14C]-thymidine were incubated for 30 minutes at 37°C with either etoposide alone (100 μmol/L, □) or triton-soluble extracts from untreated (▨) or etoposide-treated (100 μmol/L for 3 hours, ▩) cell lines (A = HL60, B = U937, C = K562, D = KCL22). Apoptotic DNA fragmentation was measured by filter elution assay. Results shown are the mean ± SD of two different experiments performed in triplicate. (E) Agarose gel electrophoresis of nuclear DNA from indicated cell lines before (lane 1, 3, 5, 7, and 9) and after (lane 2, 4, 6, 8, and 10) a 30-minute incubation in the presence of triton-soluble extracts from etoposide-treated U937 cells (100 μmol/L for 3 hours).

Fig. 3.

Apoptotic DNA fragmentation in a cell-free system. (A through D) Nuclei from the four cell lines indicated on axis in which DNA was labeled with [14C]-thymidine were incubated for 30 minutes at 37°C with either etoposide alone (100 μmol/L, □) or triton-soluble extracts from untreated (▨) or etoposide-treated (100 μmol/L for 3 hours, ▩) cell lines (A = HL60, B = U937, C = K562, D = KCL22). Apoptotic DNA fragmentation was measured by filter elution assay. Results shown are the mean ± SD of two different experiments performed in triplicate. (E) Agarose gel electrophoresis of nuclear DNA from indicated cell lines before (lane 1, 3, 5, 7, and 9) and after (lane 2, 4, 6, 8, and 10) a 30-minute incubation in the presence of triton-soluble extracts from etoposide-treated U937 cells (100 μmol/L for 3 hours).

Close modal
Cleavage of procaspase-3 is delayed in bcr-abl+cell lines.

Using an MoAb that recognizes the 32-kD precursor form of the CED-3–related caspase-3 and immunoblot analysis, we investigated the effects of etoposide treatment on procaspase-3 expression in the four studied cell lines. A rabbit polyclonal anti–caspase-3–p17 antibody was used to identify the p19 intermediate cleavage form and the p17 active subunit of caspase-3. Expression of the long isoform of procaspase-2 (ICH-1L), a protease distinct from CED-3–related caspase, was studied simultaneously (Fig 4).Procaspase-3 basal expression was observed to be lower in HL-60 cells than in other cell lines. When HL-60 and U937 cells were treated for 3 hours with 50 μmol/L etoposide, procaspase-3 expression decreased, the p19 and p17 subunits appeared, and the expression of the long isoform of procaspase-2 remained unchanged. Conversely, whenbcr-abl+ K562 and KCL-22 cells were treated with 100 μmol/L etoposide for 3 hours, the procaspase-3 was cleaved only 48 hours after the beginning of drug treatment (Fig 4). These data suggested that the apoptotic pathway was delayed upstream of procaspase-3 activation in the bcr-abl+ cell lines.

Fig. 4.

Activation of procaspase-3 in etoposide-treated leukemic cells. Western blot analysis of procaspase-2L (the 45-kD long isoform of ICH-1), procaspase-3 (the 32 kD proenzyme CPP32/Yama/Apopain) and active caspase-3 fragments (p19 and p17) in cell lines treated for indicated times with 100 μmol/L etoposide.

Fig. 4.

Activation of procaspase-3 in etoposide-treated leukemic cells. Western blot analysis of procaspase-2L (the 45-kD long isoform of ICH-1), procaspase-3 (the 32 kD proenzyme CPP32/Yama/Apopain) and active caspase-3 fragments (p19 and p17) in cell lines treated for indicated times with 100 μmol/L etoposide.

Close modal
bcr-abl Overexpression in UT7 cells confirms that p210bcr-abl delays procaspase-3 activation and target protein cleavage.

Although the resistance of K562 cells to drug-induced apoptosis was previously shown to be related to BCR-ABL expression, we checked some of the previously related observations in the GM-CSF–dependent UT7 cells and a bcr-abl–transfected clone, UT7/9 (Fig5). The sensitivity of UT7 cells to etoposide-induced apoptosis was lower than that of U937 and HL60 cells. Nevertheless, drug treatment clearly induced apoptotic DNA fragmentation that was delayed in bcr-abl–transfected UT7/9 cells. The proteolytic cleavage of the nuclear enzyme PARP is a common apoptosis-associated event that was related to the proteolytic activation of caspases.37 Cleavage of both procaspase-3 and PARP was delayed in UT7/9 cells as compared with UT7 cells, whereas the level of the long isoform of procaspase-2 remained stable (Fig 5). These results confirmed that p210bcr-abl-mediated inhibition of the apoptotic pathway occured upstream of procaspase-3 activation. By use of the cell-free system, we showed that triton-soluble extracts from U937 cells treated for 3 hours with 100 μmol/L etoposide induced apoptotic DNA fragmentation in nuclei from UT7/9 cells. This observation confirmed that bcr-abl expression did not inhibit the ability of nuclear DNA to undergo apoptotic fragmentation (Fig 6).

Fig. 5.

p210bcr-abl expression delays apoptosis upstream of procaspase-3 activation and PARP cleavage. (A) Agarose gel electrophoresis of DNA from UT7 and UT7/9 cells treated for indicated times with 100 μmol/L etoposide (UT7, left panel; UT7/9: whole panel) or deprived from GM-CSF for indicated times (UT7, right panel). (B) Western blot analysis of procaspase-2L (the 45-kD long isoform of ICH-1), procaspase-3 (the 32-kD proenzyme CPP32/Yama/Apopain), active caspase-3 fragments and PARP in cell lines treated for indicated times with 100 μmol/L etoposide (UT7, left panel; UT7/9: whole panel) or deprived from GM-CSF for indicated times (UT7, right panel). (Co = control untreated cells = time 0.)

Fig. 5.

p210bcr-abl expression delays apoptosis upstream of procaspase-3 activation and PARP cleavage. (A) Agarose gel electrophoresis of DNA from UT7 and UT7/9 cells treated for indicated times with 100 μmol/L etoposide (UT7, left panel; UT7/9: whole panel) or deprived from GM-CSF for indicated times (UT7, right panel). (B) Western blot analysis of procaspase-2L (the 45-kD long isoform of ICH-1), procaspase-3 (the 32-kD proenzyme CPP32/Yama/Apopain), active caspase-3 fragments and PARP in cell lines treated for indicated times with 100 μmol/L etoposide (UT7, left panel; UT7/9: whole panel) or deprived from GM-CSF for indicated times (UT7, right panel). (Co = control untreated cells = time 0.)

Close modal
Fig. 6.

Nuclear DNA fragmentation in the p210bcr-abl-expressing UT7/9 cells. Nuclei from UT7/9 cells in which DNA was labeled with [14C]-thymidine were incubated for 30 minutes at 37°C with buffer alone (Buffer) or 100 μmol/L etoposide alone (VP-16) or triton-soluble extracts from either untreated (U937) or etoposide-treated (100 μmol/L for 3 hours; U937/VP) U937 cells. Apoptotic DNA fragmentation was then measured by filter elution assay. Results are the mean ± SD of two different experiments performed in duplicate.

Fig. 6.

Nuclear DNA fragmentation in the p210bcr-abl-expressing UT7/9 cells. Nuclei from UT7/9 cells in which DNA was labeled with [14C]-thymidine were incubated for 30 minutes at 37°C with buffer alone (Buffer) or 100 μmol/L etoposide alone (VP-16) or triton-soluble extracts from either untreated (U937) or etoposide-treated (100 μmol/L for 3 hours; U937/VP) U937 cells. Apoptotic DNA fragmentation was then measured by filter elution assay. Results are the mean ± SD of two different experiments performed in duplicate.

Close modal
Untreated BCR-ABL–expressing cells do not contain an inhibitor of the apoptotic pathway triggered by etoposide.

To further explore the mechanisms by which BCR-ABL delays etoposide-induced apoptosis in leukemic cells, we analyzed the influence of triton-soluble extracts from untreatedbcr-abl+ cells (K562, KCL22, and UT7/9) on the ability of extracts from etoposide-treated U937 cells to trigger apoptotic DNA fragmentation in the nuclei from untreated U937 cells (Fig 7). Extracts from untreatedbcr-abl cell lines (U937, HL60, and UT7) were used as control. A mixture of an equal volume of extracts from untreated cells and etoposide-treated U937 cells (100 μmol/L for 3 hours) was incubated for 30 minutes at 37°C with [14C]-thymidine–labeled nuclei from untreated U937 cells. DNA fragmentation was measured by the filter elution assay. This experiment indicated that untreated bcr-abl+ cells do not contain an inhibitor of the apoptotic pathway triggered by etoposide, further supporting the observation that the p210bcr-abl protein inhibits the caspase cascade at or before the cleavage of procaspase-3.

Fig. 7.

Lack of apoptosis inhibitory effect of triton-soluble extracts from untreated bcr-abl+ cell lines. Nuclei from U937 cells in which DNA was labeled with [14C]-thymidine were incubated for 30 minutes at 37°C with either triton-soluble extracts from untreated (U937) or etoposide-treated (100 μmol/L for 3 hours; U-VP) U937 cells or a mixture or an equal volume of triton-soluble extracts from etoposide-treated (100 μmol/L for 3 hours) U937 cells and triton-soluble extracts from untreated cell lines (U-VP + name of the untreated cell line). Results of a representative experiment are expressed as the mean ± SD of triplicate measurements standardized to the U-VP sample (100%).

Fig. 7.

Lack of apoptosis inhibitory effect of triton-soluble extracts from untreated bcr-abl+ cell lines. Nuclei from U937 cells in which DNA was labeled with [14C]-thymidine were incubated for 30 minutes at 37°C with either triton-soluble extracts from untreated (U937) or etoposide-treated (100 μmol/L for 3 hours; U-VP) U937 cells or a mixture or an equal volume of triton-soluble extracts from etoposide-treated (100 μmol/L for 3 hours) U937 cells and triton-soluble extracts from untreated cell lines (U-VP + name of the untreated cell line). Results of a representative experiment are expressed as the mean ± SD of triplicate measurements standardized to the U-VP sample (100%).

Close modal

Decreased ability to undergo apoptosis in response to drug-induced cell damage is one of the mechanisms by which tumor cells can escape their cytotoxic activity.27 Cells which overexpressed v-abl or p210bcr-abl receive all the measurable damage induced by cytotoxic drugs but are unable to couple this damage to the apoptotic pathway.7,28 Antisense oligonucleotides that downregulate p210bcr-abl expression render K562 cells more susceptible to cell death induction by a variety of cytotoxic drugs.8,10 These oligonucleotide antisenses also render Fas-transfected K562 cells more sensitive to Fas-induced apoptosis.12 Thus, v-abl and p210bcr-abl act downstream of the drug-target interaction to prevent the coupling of drug-induced DNA damage to the apoptotic pathways. This decreased sensitivity to apoptosis induction could contribute to the progression of CML if resistant cells that are genetically damaged subsequently proliferate. Therefore, it is essential to understand the molecular mechanisms by which p210bcr-abl delays the apoptotic pathway triggered by DNA damaging agents.

The bcl-2 oncogene is a well-characterized inhibitor of drug-induced cell death. Apoptosis induction by DNA damaging agents is suppressed by transfection of cells with the oncogene bcl-2despite similar levels of drug-induced DNA damage in both parent and transfected cells.27 The clinical syndromes produced by BCR-ABL expression (CML) and Bcl-2 deregulation (follicular lymphoma) both exhibit an initial indolent phase with uninterrupted differentiation and are both incurable with conventional doses of chemotherapy.38 Therefore, p210bcr-abl-mediated inhibition of cell death could have been mediated by overexpression of Bcl-2. Actually, we did not observe any relationship between Bcl-2 and Bax protein levels in both untreated (Fig 1) and treated (not shown) cell lines and their sensitivity to etoposide-induced cell death. Accordingly, overexpression of v-abl in a clone of HL-60 cells in which no Bcl-2 protein could be detected remains capable of inducing an antiapoptotic state.39,40 Bcl-2 is now known to belong to a growing family of apoptosis-regulatory gene products which may either be death antagonists or death agonists and we cannot rule out a role for other Bcl-2 family members as mediators of p210bcr-abl-induced inhibition of cell death.

We have shown previously that triton-soluble extracts from etoposide-treated leukemic cell lines such as HL-60 and U937 activated a nuclear endonuclease that triggered apoptotic DNA fragmentation at neutral pH in the presence of Mg2+.21 In the present study, experiments performed in this cell-free system show that the nuclei from untreated cells that express p210bcr-ablremain sensitive to triton-soluble extracts from p210bcr-abl-negative cells treated with etoposide. These results indicate that the nuclease responsible for apoptotic DNA fragmentation in these cells is not inhibited by p210bcr-abl.

Growth factors influence the sensitivity of myeloid cell lines to apoptosis induced by chemotherapeutic drugs.41The phosphorylation patterns induced by p210bcr-abl and growth factors show some similarities. Calphostin C, an inhibitor of protein kinases such as protein kinase C, restores drug sensitivity in cells with active v-abl, suggesting a role for protein kinases in the suppression of drug-induced apoptosis by v-ABL and p210bcr-abl.7 However, drug treatment induces more rapid apoptosis in p210bcr-ablUT7 cells cultured in the presence of GM-CSF compared with UT7/9 cells in the absence of GM-CSF, suggesting that p210bcr-ablimposes a survival advantage over that imparted by GM-CSF and that cellular components other than p210bcr-abl are probably involved in its antiapoptotic effect.9,14-16 

Recent evidences indicate that activation of procaspases in apoptosis occurs via a proteolytic cascade.26 For example, caspase-4 can activate procaspase-1 that, in turn, can process procaspase-3.42 Under normal physiological conditions, procaspase-1 (pro-ICE) is activated in monocytes and cleaves pro–interleukin (IL)-1β to produce mature IL-1β cytokine. Other downstream procaspases such as procaspase-3 might also be activated by active caspase-1; nevertheless, the cells do not undergo apoptosis. This suggests that a threshold of tolerance of active caspases exists in different cells. The present study indicates that p210bcr-abl-mediated delay in apoptosis induction is associated with a delayed activation of procaspase-3 in response to DNA damage and growth factor–deprivation rather than to an increased threshold of active caspase-3 tolerance. Human PARP is one of the specific targets of apoptosis-associated proteolysis.36Although most caspases cleave this protein in vitro,22-24,43 kinetic properties of CED-3–related caspases suggest these proteases to be the most involved in PARP cleavage.21,23 Inhibition of PARP cleavage in p210bcr-abl+ UT7/9 cells treated with etoposide suggests that p210bcr-abl prevents the activation of all CED-3–related caspases. Interestingly, the long isoform of caspase-2 is not cleaved and activated in any tested cell line when exposed to VP-16, indicating that all the caspases are probably not implicated in the VP-16–induced cell death pathway. Experiments performed in the cell-free system showed that neither p210bcr-abl nor other cellular components involved in its antiapoptotic effect could prevent apoptotic DNA fragmentation triggered by extracts from VP-16–treated U937 cells, further supporting the conclusion that p210bcr-abl acts at or upstream the procaspase-3 activation level.

Inhibition of the etoposide-mediated apoptotic pathway upstream of the activation of this CED-3–related caspases is in accordance with the recent observation that p210bcr-abl-mediated resistance to apoptosis is overcome by cytotoxic T cells44 and NK and LAK cells.29 These lymphocytes use two systems to induce cell death in target cells, namely, the granzyme and the Fas systems. The substantial role played by granzyme B in the former has been confirmed by targeted gene disruption. Granzyme B is a serine protease with an unusual specificity for cleaving synthetic substrates after Asp residues. Granzyme B activates several CED-3–related procaspases including procaspase-3 and procaspase-7, whereas procaspase-1 is not a substrate for granzyme B.45 Several recent studies established that Bcl-2 and Bcl-XL function upstream of the CED-3–related caspases. The present study shows that p210bcr-abl similarly delays drug-induced apoptosis upstream of these proteases.

Several methods were proposed to overcome drug resistance of CML cells. Tyrosine kinase inhibitors inhibit the growth of the murine IL-3–dependent myeloid 32Dc13(G) cell line as well as a subclone transformed to IL-3–independent growth by retroviral transduction and expression of BCR-ABL. However, these compounds induced apoptosis in the parental cells and necrosis in bcr-abl–transformed cells, confirming that BCR-ABL could suppress apoptotic signal transduction.11 Combination of antisense oligonucleotides that downregulate BCR-ABL protein with a conventional chemotherapeutic agent such as mafosfamide was shown to be an efficient strategy to eliminate CML cells in mice.46 The present study indicates that targeting CED-3–related caspases could be another efficient way to overcome drug resistance of CML cells by acting downstream of the apoptotic pathway inhibition.

Supported by grants from the Burgundy, Saône et Loire and Yonne Comittees of the Ligue Nationale Contre le Cancer, the Association pour la Recherche contre le Cancer (ARC#4075) and the Conseil Régional de Bourgogne.

Address reprint requests to Eric Solary, MD, INSERM CJF 94-08, Faculté de Médecine, 7, Blvd Jeanne d'Arc, 21033 DIJON Cedex, France.

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

1
Nowell
PC
Hungerford
DA
A minute chromosome in human chronic granulocytic leukemia.
Science
132
1960
1497
2
Ben-Neriah
Y
Daley
QG
Mes-Mason
AM
Witte
ON
Baltimore
D
The chronic myelogenous leukemia-specific p210 protein is the product of the bcr-abl hybrid gene.
Science
233
1986
212
3
Daley
GQ
Van Etten
RA
Baltimore
D
Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome.
Science
247
1990
824
4
Eaves
AC
Cashman
JD
Gaboury
LA
Kalousek
DK
Eaves
CJ
Unregulated proliferation of primitive chronic myelogenous leukemia progenitors in the presence of normal marrow adherent cells.
Proc Natl Acad Sci USA
83
1986
5306
5
Stryckmans
P
Debussher
L
Socquet
M
Regulation of bone marrow myeloblast proliferation in chronic myeloid leukemia.
Cancer Res
36
1976
3034
6
Spooncer
E
Fairbairn
L
Cowling
GJ
Dexter
TM
Whetton
AD
Owen-Lynch
PJ
Biological consequences of p160v-abl protein tyrosine kinase activity in a primitive, multipotent hematopoietic cell line.
Leukemia
8
1994
620
7
Chapman
RS
Whetton
AD
Dive
C
The suppression of drug-induced apoptosis by activation of v-abl protein tyrosine kinase.
Cancer Res
54
1994
5131
8
Bedi
A
Zehnbauer
BA
Barber
JP
Sharkis
SJ
Jones
RJ
Inhibition of apoptosis by BCR-ABL in chronic myelogenous leukemia.
Blood
83
1994
2038
9
Bedi
A
Berber
JP
Bedi
GC
el-Deiry
WS
Sidransky
D
Vala
MS
Akhtar
AJ
Hilton
J
Jones
RJ
BCR-ABL-mediated inhibition of apoptosis with delay of G2/M transition after DNA damage: A mechanism of resistance to multiple anticancer agents.
Blood
86
1995
1148
10
McGahon
A
Bissonnette
R
Schmitt
M
Cotter
KM
Green
DR
Cotter
TG
BCR-ABL maintains resistance of chronic myelogenous leukemia cells to apoptotic cell death.
Blood
83
1994
1179
11
Laneuville
P
Timm
M
Hudson
AT
Bcr/abl expression in 32D c13(G) cells inhibits apoptosis induced by protein tyrosine kinase inhibitors.
Cancer Res
54
1994
1360
12
McGahon
AJ
Nishioka
WK
Martin
SJ
Mahboubi
A
Cotter
TG
Green
DR
Regulation of the Fas apoptotic cell death pathway by Abl.
J Biol Chem
270
1995
22625
13
Smetsers
TF
van de Locht
LTF
Pennings
AH
Wessels
HM
de Witte
T
Mensink
EJ
Phosphorothioate BCR-ABL antisense oligonucleotides induce cell death, but fail to reduce cellular bcr-abl protein levels.
Leukemia
8
1994
129
14
Evans
CA
Lord
JM
Owen-Lynch
PJ
Johnson
G
Dive
C
Whetton
AD
Suppression of apoptosis by v-ABL protein tyrosine kinase is associated with nuclear translocation and activation of protein kinase C in an interleukin-3-dependent haematopoietic cell line.
J Cell Sci
108
1995
2591
15
Kan
O
Baldwin
SA
Whetton
AD
Apoptosis is regulated by the rate of glucose transport in an interleukin 3 dependent cell line.
J Exp Med
180
1994
917
16
Cortez
D
Stoica
G
Pierce
JH
Pendergast
AM
The BCR-ABL tyrosine kinase inhibits apoptosis by activating a Ras-dependent signaling pathway.
Oncogene
13
1996
2589
17
Owen-Lynch
PJ
Wong
AK
Whetton
AD
V-Abl-mediated apoptotic suppression is associated with SHC phosphorylation without concomitant mitogen-activated protein kinase activation.
J Biol Chem
270
1995
5956
18
Alnemri
ES
Livingston
DJ
Nicholson
DW
Salvesen
G
Thornberry
NA
Wong
WW
Yuan
J
Human ICE/CED-3 protease nomenclature.
Cell
87
1996
171
19
Patel T, Gores GJ, Kaufmann SH: The role of proteases during apoptosis. FASEB J 10:587, 1996
20
Kuida
K
Lippke
JA
Ku
G
Harding
MW
Livingston
DJ
Su
MS
Flavell
RA
Altered cytokine export and apoptosis in mice deficient in interleukin-1β-converting enzyme.
Science
267
1995
2000
21
Dubrez
L
Savoy
I
Hammann
A
Solary
E
Pivotal role of a DEVD-sensitive step in etoposide-induced and Fas-mediated apoptotic pathways.
EMBO J
15
1996
5504
22
Fernandes-Alnemri
T
Litwack
G
Alnemri
ES
Mch2, a new member of apoptotic Ced-3/Ice cysteine protease gene family.
Cancer Res
55
1995
273
23
Fernandes-Alnemri
T
Takahashi
A
Armstrong
R
Krebs
J
Fritz
L
Tomaselli
KJ
Wang
L
Yu
Z
Croce
CM
Salveson
G
Earnshaw
WC
Litwack
G
Alnemri
ES
Mch3, a novel human apoptotic cysteine protease highly related to CPP32.
Cancer Res
55
1995
6045
24
Nicholson
DW
Ali
A
Thornberry
NA
Vaillancourt
JP
Ding
CK
Gallant
M
Gareau
Y
Griffin
PR
Labelle
M
Lazebnik
YA
Munday
NA
Raju
SM
Smulson
ME
Yamin
T
Yu
VL
Miller
DK
Identification and inhibition of the ICE/CED-3 protease necessary for mammalian apoptosis.
Nature
376
1995
37
25
Susin
SA
Zamzami
N
Castedo
M
Hirsch
T
Marchetti
P
Macho
A
Daugas
E
Geuskens
M
Kroemer
G
Bcl-2 inhibits the mitochondrial release of an apoptogenic protease.
J Exp Med
184
1996
1331
26
Goldstein
P
Controlling cell death.
Science
275
1997
1081
27
Pommier
Y
Leteurtre
F
Fesen
M
Fujimori
A
Bertrand
R
Solary
E
Kohlhagen
G
Kohn
KW
Cellular determinants of sensitivity and resistance to DNA topoisomerase inhibitors.
Cancer Invest
12
1994
530
28
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
29
Roger
R
Issaad
C
Pallardy
M
Leglise
MC
Turhan
AG
Bertoglio
J
Breard
J
BCR-ABL does not prevent apoptotic cell death induced by human natural killer or lymphokine-activated killer cells.
Blood
87
1996
1113
30
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
31
Smith
PK
Krohn
RI
Hermanson
GT
Mallia
AK
Gartner
FH
Provenzano
MD
Fujimoto
EK
Goeke
NM
Olson
BJ
Klenk
DC
Measurement of protein using bicinchoninic acid.
Anal Biochem
150
1985
76
32
Bertrand
R
Kohn
KW
Solary
E
Pommier
Y
Detection of apoptosis-associated DNA fragmentation using a rapid and quantitative filter binding assay.
Drug Develop Res
34
1995
138
33
Miller
SA
Dykes
DD
Polesky
HF
A simple salting out procedure for extracting DNA from human nucleated cells.
Nucleic Acids Res
16
1988
1215
34
Schlegel
J
Peters
I
Orrenius
S
Miller
DK
Thornberry
NA
Yamin
T
Nicholson
DW
CPP32/Apopain is a key interleukin 1β converting enzyme-like protease involved in Fas-mediated apoptosis.
J Biol Chem
271
1996
1841
35
Korsmeyer
S
Bcl-2 initiates a new category of oncogenes: Regulators of cell death.
Blood
80
1992
879
36
Fisher
TC
Milner
AE
Gregory
CD
Jackman
AL
Aherne
GW
Hartley
JA
Dive
C
Hickman
JA
Bcl-2 modulation of apoptosis induced by anticancer drugs: Resistance to thymidilate stress is independent of classical resistance pathways.
Cancer Res
53
1993
3321
37
Kaufmann
SH
Desnoyers
S
Ottaviano
Y
Davidson
NE
Poirier
GG
Specific proteolytic cleavage of poly(ADP-ribose) polymerase: An early marker of chemotherapy-induced apoptosis.
Cancer Res
53
1993
3976
38
Solary
E
Dubrez
L
Eymin
B
The role of apoptosis in the pathogenesis and treatment of diseases.
Eur Respir J
9
1996
1293
39
Green
DR
McGahon
A
Martin
SJ
Regulation of apoptosis by oncogenes.
J Cell Biochem
60
1996
33
40
Chapman
RS
Whetton
AD
Chresta
CM
Dive
C
Characterization of drug resistance mediated via the suppression of apoptosis by Abelson protein tyrosine kinase.
Mol Pharmacol
48
1995
334
41
Lotem
J
Sachs
L
Hematopoietic cytokines inhibit apoptosis induced by transforming growth factor β1 and cancer chemotherapy compounds in myeloid leukemic cells.
Blood
80
1992
1750
42
Enari
M
Talanian
RV
Wong
WW
Nagata
S
Sequential activation of ICE-like and CPP32-like proteases during Fas-mediated apoptosis.
Nature
380
1996
723
43
Gu
Y
Sarnecki
C
Aldape
RA
Livingston
DJ
Su
MSS
Cleavage of Poly(ADP-ribose)polymerase by interleukin-1β converting enzyme and its homologs Tx and Nedd-2.
J Biol Chem
32
1995
18715
44
Fuchs
EJ
Bedi
A
Jones
RJ
Hess
AD
Cytotoxic T cells overcome BCR-ABL-mediated resistance to apoptosis.
Cancer Res
55
1995
463
45
Darmon
AJ
Nicholson
DW
Bleackley
RC
Activation of the apoptotic protease CPP32 by cytotoxic T-cell-derived granzyme B.
Nature
377
1995
446
46
Skorski
T
Nieborowska-Skorska
M
Barletta
C
Malaguarnera
L
Szczylik
C
Chen
TS
Lange
B
Calabretta
B
Highly efficient elimination of Philadelphia leukemic cells by exposure to bcr/abl antisense oligonucleotides combined with mafosfamide.
J Clin Invest
92
1993
194
Sign in via your Institution