The anthracycline daunorubicin is widely used in the treatment of acute nonlymphocytic leukemia. The drug has, of course, been the object of intense basic research, as well as preclinical and clinical study. As reviewed in this article, evidence stemming from this research clearly demonstrates that cell response to daunorubicin is highly regulated by multiple signaling events, including a sphingomyelinase-initiated sphingomyelin-ceramide pathway, mitogen-activated kinase and stress-activated protein/c-Jun N-terminal kinase activation, transcription factors such as nuclear factor κB, as well as the Fas/Fas-ligand system. These pathways are themselves influenced by a number of lipid products (diacylglycerol, sphingosine-1 phosphate, and glucosyl ceramide), reactive oxygen species, oncogenes (such as the tumor suppressor gene p53), protein kinases (protein kinase C and phosphoinositide-3 kinase), and external stimuli (hematopoietic growth factors and the extracellular matrix). In light of the complexity and diversity of these observations, a comprehensive review has been attempted toward the understanding of their individual implication (and regulation) in daunorubicin-induced signaling.

The anthracycline daunorubicin (DNR) is one of the major antitumor agents widely used in the treatment of acute myeloid leukemias (AMLs). Cytotoxicity mediated by DNR is generally thought to be the result of drug-induced damage to DNA. This damage is mediated by quinone-generated redox activity, intercalation-induced distortion of the double helix, or stabilization of the cleavable complex formed between DNA and topoisomerase II.1 However, how and why such events should bring about cell death remains unclear, especially when one considers that DNA interaction may not be a prerequisite for anthracycline cytotoxicity.2-5 Hence, the exploration and understanding of the process of apoptosis has forced a reconsideration of the mechanisms whereby myeloid leukemia cells respond to DNR. In this context, we have shown that, within a narrow concentration range (0.2-1 μM), DNR can trigger apoptosis in the monocytic U937 or the myelocytic HL-60 AML cells but not in the immature (CD 34+) KG1a, KG1, or HEL cells,6the latter being more resistant to DNR than the former.7These results suggested that DNR triggers apoptotic signals in drug-sensitive AML cells and that inhibition of these signals may contribute to drug resistance and, for example, to the inherent resistance of immature AML cells. For this reason, others and we have investigated the mechanism by which DNR activates apoptosis in DNR-sensitive AML cells.

DNR activates the sphingomyelin cycle in sensitive leukemic cells

In the early 1990s, a number of studies provided growing evidence that sphingomyelin breakdown products (ie, ceramide [CER] and phosphorylcholine) could play an important role in mediating the action of cytokines, including tumor necrosis factor α (TNFα) and interferon. For example, Obeid et al8 were the first to report that TNFα could stimulate a neutral sphingomyelinase (N-SMase) responsible for sphingomyelin hydrolysis and subsequent generation of CER in U937 cells and that C2-CER, a synthetic cell-permeable CER analog, induced apoptosis in these cells. These results strongly supported the implication of the sphingomyelin-CER pathway in apoptosis induced by TNFα and, perhaps, by other cytotoxic molecules. In further studies, Haimovitz-Friedmann et al9proposed the involvement of the sphingomyelin-CER pathway in apoptosis induced by γ-radiation in bovine endothelial cells.

On the basis of those studies, our group investigated the role of the sphingomyelin-CER pathway in DNR-induced apoptosis. DNR exposure at concentrations that induced apoptosis (0.5-1 μM) stimulated early (5-10 minutes) sphingomyelin cycle (hydrolysis and resynthesis) and subsequent CER generation in both U937 and HL-60 cells. The concentration of endogenous CER generated (∼10 μM, estimated by cell volume) is clearly sufficient to induce apoptosis.8Because these cells exhibit a mutated or deleted form of p53, it appears that CER operated through a p53-independant mechanism. Further studies have shown that DNR induced sphingomyelin hydrolysis associated with the stimulation of N-SMase, whereas acidic SMase did not appear to be involved.10 Moreover, we have recently reported that DNR induced sphingomyelin cycle, CER generation, and apoptosis in Epstein-Barr virus–transformed lymphoblastoid cell lines established from patients with Niemann-Pick disease, a genetic disorder characterized by the lack of acidic SMase activity.11These results suggest that acidic SMase is dispensable for DNR-induced sphingomyelin hydrolysis.

In another study, Bose et al12 have proposed that DNR may induce CER accumulation because of enhanced de novo synthesis through CER synthase stimulation, a mechanism that has also been suggested for TNFα-induced cell death in nonhematopoietic cell death.13,14 However, it should be noted that, in the study by Bose et al,12 cells were treated with very high DNR doses, whereas, at clinically relevant drug concentrations, we were unable to detect CER synthase stimulation.10 The fact that sphingomyelin hydrolysis occurred within 5 to 10 minutes after drug exposure and apoptosis was not detected before 4 to 6 hours raised some controversies about the implication of the sphingomyelin cycle in DNR apoptosis. Therefore, we have carefully examined the influence of DNR on sphingomyelin metabolism over a longer period of time (0-12 hours).

In this study, we have found that DNR exposure resulted in at least 4 sphingomyelin cycles (hydrolysis and resynthesis) with concomitant (4 cycles) CER production within the first 4 hours, after which poly(ADP-ribose) polymerase cleavage, DNA laddering, and apoptosis-associated morphologic changes occurred. Moreover, the successive waves of CER production were not influenced by fumonisin B1, a potent and specific CER synthase inhibitor.15 These results confirmed that de novo CER synthase plays little if any role in DNR-induced CER signaling. The fact that DNR induced several peaks of CER production suggested that sphingomyelin hydrolysis products (ie, CER or phosphorylcholine) might reactivate N-SMase. In fact, we have found that cell-permeant CER may indeed stimulate N-SMase, sphingomyelin hydrolysis, and endogenous CER production, suggesting that CER may enhance its own production through feedback control of N-SMase.15 Therefore, one can speculate that DNR activates a single sphingomyelin cycle that is sufficient for inducing CER autoproduction and that these repeated CER-mediated apoptotic signals are perhaps needed for maintaining an apoptotic (cell suicide) signaling pressure overcoming latent cell survival mechanisms. Finally, it is noteworthy that DNR-induced topoisomerase II cleavable complexes have not been described in the literature under these conditions (1 μM DNR treatment for 4 hours); of course this does not demonstrate unambiguously that DNR interaction with DNA, or more specifically with topoisomerase II, is not requisite for apoptosis signaling because it can be argued that present experimental procedures lack sensitivity in detecting minute amounts of DNR-topoisomerase-DNA complexes.

CER targets

The emergence of CER as an important mediator of cell death induced by cytotoxic molecules has stimulated an extensive search for downstream targets involved in CER-mediated apoptosis. In fact, CER was found to activate a large variety of signaling proteins, including Raf-1, extracellular-regulated kinases (ERKs), a CER-activated protein phosphatase, as well as stress-activated protein/c-Jun N-terminal kinase (SAPK/JNK). By using dominant- negative mutants, Verheij et al16 elegantly demonstrated that the MEKK1-SEK1-JNK cascade is one of the major components of CER-mediated apoptosis. This finding is consistent with another study which showed that c-Jun activation is required for CER-mediated apoptosis.17Further studies have supported the function of JNK in the initiation of programmed cell death. For example, it was shown that overexpression of MEKK1 or ASK1, 2 potent and specific in situ activators of SEK1 and JNKs, had lethal effect on fibroblasts.18-20 Because DNR triggers both CER production and apoptosis, it was expected that this drug may also activate JNK signaling. In fact, we and others have reported that both DNR and doxorubicin, in a dose-dependent manner, increased JNK1 tyrosine phosphorylation, stimulated JNK1 enzymatic activity, and enhanced DNA binding activity of the transcription factor activated protein-1 (AP-1).21-23 Anthracyclines are not the unique antitumor agents that activate the JNK/c-Jun pathway. Indeed, other genotoxic agents such as UV,24 ionizing radiation,25-27 alkylating agents,25etoposide,21 or cytosine arabinoside28 as well as nongenotoxic agents such as microtubule poisons29,30 also activate JNK. These observations suggest a general function for JNK in mediating cell death induced by antitumor agents (see Kyriakis and Avruch for review31). Therefore, it appears that, in leukemic-sensitive cells, DNR activates a complex signaling cascade that consists of repeated sphingomyelin cycles followed by several waves of CER production; this second messenger, in turn, activates a common downstream cell death effector-mediated pathway that involves JNK stimulation and c-Jun/AP-1 activation.

Regulation of CER production

CER production is limited by both N-SMase activity and the magnitude of sphingomyelin pool disposable for hydrolysis. Previous studies have shown that N-SMase activity is strongly influenced by protein kinase C (PKC) activity. For example, we have reported that phorbol ester– or phosphatidylserine-induced PKC stimulation resulted in the inhibition of N-SMase stimulation, sphingomyelin hydrolysis, CER production, and apoptosis induced by DNR in U937 cells.32Conversely, PKC inhibitors were found to stimulate N-SMase.33 The amount of hydrolyzable sphingomyelin is another candidate for regulating CER production. Indeed, it has been reported that, whereas sphingomyelin is preferentially distributed within the outer leaflet of the plasma membrane (sphingomyelin transverse asymmetry), the sphingomyelin pool disposable for hydrolysis consists primarily in the sphingomyelin component that is associated to the plasma membrane inner leaflet.34 Thus, it is conceivable that reduction of hydrolyzable sphingomyelin pool because of altered transverse asymmetry may result in reduced CER production. In 2 studies, we have reported that, in KG1a AML cells, which are naturally resistant to DNR,7 mitoxantrone,35and TNFα,36 the inner leaflet-associated sphingomyelin pool was greatly reduced, compared with the sensitive U937 cells, whereas KG1a and U937 cells exhibited similar total sphingomyelin amount. Interestingly, in the resistant cells, cytotoxic effectors such as TNFα,36 DNR, and mitoxantrone (A Bettaiëb et al, unpublished results, July 1997) failed to induce sphingomyelin hydrolysis, CER generation, and apoptosis. Thus, it is possible that, in some AML cells, the activation of yet undefined sphingomyelin translocases results not only in modification of sphingomyelin transverse asymmetry but also in reduced sphingomyelin hydrolyzable pool, decreased CER production, and inhibition of apoptosis.37 Confirming this hypothesis, it has recently been shown that the CER generated from the plasma membrane sphingomyelin gains access to a SMase because of phospholipid scrambling.38 

Regulation of CER metabolism

Intracellular CER concentration results from the equilibrium between CER production and CER metabolism. CER metabolism also plays an important role in regulating intracellular CER concentration and therefore DNR cytotoxicity. Theoretically, CER produced by DNR may enter into 3 distinct metabolic pathways that all result in decreasing intracellular CER levels.

First, CER can be transferred to a phosphorylcholine group to generate sphingomyelin (and diacylglycerol [DAG]) on sphingomyelin synthase stimulation. It is likely that this metabolic pathway is activated for sphingomyelin resynthesis after sphingomyelin hydrolysis during the sphingomyelin cycle. This enzyme therefore has the important ability to directly regulate, in opposite directions, CER and DAG levels within the cells.39 However, despite the great biological potential of sphingomyelin synthase, very little is known about location, distribution, and regulation of this enzyme.40Whether sphingomyelin synthase plays a role in DNR-induced cytotoxicity remains to be investigated.

Second, on ceramidase stimulation, CER can be catabolized into sphingosine that, in turn, can be converted into sphingosine-1-phosphate through sphingosine-1-kinase. In a recent study, we have reported that DNR induced CER production and apoptosis in cells derived from Farber disease, which are genetically deficient for lysosomal ceramidase, the major component of cellular ceramidase activity.41 Although we cannot totally exclude the implication of extralysosomal ceramidases,42 this result suggests that sphingosine plays little if any role in DNR-induced apoptosis. However, from studies performed by Cuvillier et al,43,44 sphingosine-1-phosphate has emerged as one of the most potent regulators of apoptosis. Indeed, sphingosine-1-phosphate inhibits apoptosis induced by CER and other effectors.43,44 The mechanism by which sphingosine-1-phosphate interferes with CER-induced apoptotic signaling is not fully understood. However, it has been reported that sphingosine-1-phosphate inhibits CER-induced JNK stimulation and caspase activities.44,45 Therefore, sphingosine-1-phosphate and sphingosine-1-kinase might play an important role in regulating DNR-induced apoptosis. In fact, it has been demonstrated that sphingosine-1-phosphate inhibits doxorubicin-induced apoptosis.46 From these studies, one can speculate that sphingosine-1-kinase stimulation may contribute to DNR resistance. Because sphingosine-1-kinase is potently stimulated by PKC, it is possible that sphingosine-1-phosphate overproduction represents another mechanism by which PKC exerts its protective function in DNR-treated cells.

Third, CER can be transformed to glucosylceramide by a glucosylceramide synthase. Previous reports have indicated that glucosylceramide has no cytotoxic property or may even stimulate cell proliferation47 and that glucosylceramide synthase inhibitors have been found to display some antitumor activity.48,49 These results suggest that glucosylceramide synthase plays an important role in cellular protection. Indeed, it has been described that cells transduced by glucosylceramide synthase gene were highly resistant to anthracyclines50 and that enzyme activity was significantly boosted in multidrug-resistant (MDR) cells.51 Conversely, transfection of glucosylceramide synthase antisense reverses adriamycin resistance.52 DNR triggers the sphingomyelin cycle in MDR cells when used at high doses.53 Therefore, one can speculate that in DNR-treated MDR cells CER originated from sphingomyelin hydrolysis is rapidly converted to glucosylceramide because of glucosylceramide synthase overactivity.54,55 If this speculation is the case, glucosylceramide synthase appears as an attractive target for MDR reversal. In fact, inhibition of CER glycosylation pathway increases MDR cell sensitivity to cytotoxics.56 The same group has reported that most MDR modulators, including cyclosporin A, tamoxifen, and verapamil, are potent glucosylceramide synthase inhibitors, whereas the cyclosporin A analogue PSC 833 (Valspodar), a clinically used potent MDR reversal agent, increases intracellular CER concentration by stimulating CER synthase.51,57,58 These results suggest that those agents may exert their chemosensitizing effect not only through their P-glycoprotein binding capacity, as previously postulated, but also by facilitating CER accumulation. However, the mechanism by which CER modulates anthracycline-induced cytotoxicity in MDR cells remains to be determined. For example, there is no evidence that CER interferes with P-glycoprotein function or intracellular drug distribution in MDR cells.59 

Regulation of CER apoptotic signaling pathway

The sphingomyelin-CER pathway appears to be efficiently regulated downstream of CER generation. Bcl-2 inhibits apoptosis induced by DNR without interfering with DNR-induced sphingomyelin cycle activation.60 This result is consistent with previous studies which have shown that Bcl-2 inhibits apoptosis induced by cell-permeant CER.61 PKC is also a potent regulator of CER-induced apoptosis. Indeed, previous studies not only showed that PKC activators, including phorbol esters and DAG, could inhibit the ability of cell-permeant CER to induce apoptosis but also that PKC inhibitors enhanced CER-induced apoptosis.9,32,62,63Therefore, PKC appears as a critical regulator of the sphingomyelin-CER pathway because it operates both upstream and downstream of CER production. This finding may have important implications in our understanding of AML-cell drug resistance. Indeed, a large variety of hematopoietic growth factors (HGFs), including interleukin 3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), or basic fibroblast growth factor, were found to stimulate PKC activity through phosphatidylinositol or phosphatidylcholine hydrolysis and subsequent DAG production.64 Thus, it is conceivable that autocrine production of HGFs by leukemic cells may result in DNR-induced sphingomyelin-CER pathway inhibition and thereby contribute to reduce DNR cytotoxicity in AML. The fact that HGFs were found to inhibit DNR-induced apoptosis,65,66 as well as the chemosensitizing effect of PKC inhibitors on fresh AML cell progenitors cultured in the presence of HGFs,67 supports this hypothesis. Moreover, we have recently found that Kit signaling, activated either by its natural ligand (stem cell factor) or by modification of its intracellular domain, inhibits DNR-induced SMase stimulation, CER production, and apoptosis through a PKC-dependent mechanism.68 

During the treatment of the cells with anthracyclines, nicotinamide adenine dinucleotide phosphate (NADPH)–dependent flavin reductase reduces the drug to a semiquinone radical, which can donate its free electron to molecular oxygen and generate the superoxide radical (O2). At neutral pH, the main reaction of O2 is a relatively slow spontaneous dismutation to H2O2 and O2, but this reaction can be accelerated by superoxide dismutase. Superoxide anion and hydrogen peroxide may interact (with metal ions such as iron or copper as catalyst), by the Haber-Weiss reaction to generate hydroxyl radicals (OH). By employing electron spin resonance together with a spin-trap such as DMPO (5,5-dimethyl-1-pyrroline N-oxide), anthracycline-induced OH has been detected in several types of cancer cells (see Sinha and Mimmaugh for review69). Electron spin resonance radicals have also been detected in intact erythrocytes treated with DNR.70 This result suggests that anthracycline-induced free radical production may occur independently from the interaction of the drug with internal membrane or even DNA. In addition, it has been demonstrated that all forms of radical oxygen species (ROS) may also be generated through the direct interaction of anthracyclines with cell membranes and subsequent lipid peroxidation.71 Although there is strong evidence that ROS plays a role in the cytotoxicity of anthracyclines, the mechanism by which ROS influences cell viability remains unclear.

Previous studies have shown that DNR-induced apoptosis was inhibited by ROS scavengers such as pyrrolidine dithiocarbamate and N-acetylcystein, a thiol antioxidant and a glutathione precursor, and was enhanced by buthionine-sulfoximine, which depletes the glutathione store.6 These results encouraged us to investigate the role of ROS in the DNR-stimulated sphingomyelin-CER-JNK apoptotic pathway. Indeed, the level of endogenous H2O2generated is comparable to that of a cell treated with 12 μM H2O2. In fact, further studies showed that pretreatment with N-acetylcystein abrogated DNR-induced N-SMase stimulation, sphingomyelin hydrolysis, and CER generation induced by DNR, evoking a role for ROS in N-SMase regulation.23 This hypothesis is supported by 2 other studies which showed that H2O2 stimulates sphingomyelin hydrolysis and CER generation and that N-acetylcystein and pyrrolidine dithiocarbamate were potent inhibitors of TNFα-induced sphingomyelin degradation to CER.72,73 Thus, it is conceivable that ROS originated from the NADPH-dependent DNR reduction contributes to N-SMase stimulation and subsequent CER generation. However, the mechanism by which ROS regulates N-SMase activity is not known.

Other studies have demonstrated that ROS also plays a role in CER-induced apoptosis. Indeed, permeant CER induced early and transient (5-10 minutes) H2O2 production, followed by a second wave of H2O2 detected at 1 to 3 hours that originated from mitochondrial oxidative metabolism disturbances. Furthermore, N-acetylcystein inhibited H2O2production, JNK activation, AP-1 activation, and apoptosis induced by CER.23,74,75 These results suggest that ROS plays an important role in the sphingomyelin-CER apoptotic pathway triggered by DNR at 2 different levels: upstream CER generation by stimulating N-SMase activity and downstream CER activity by mediating CER-induced JNK activation. The results also suggest a novel function of cytosolic-originated ROS in the cytotoxicity mechanism of anthracyclines. Thus, antioxidants, including superoxide dismutase, catalase, glutathione peroxidase, or thioredoxin, may influence DNR-induced cytotoxicity.

However, the fact that antioxidants inhibit apoptosis induced by chemotherapeutic drugs that have not been documented to stimulate the sphingomyelin cycle (ie, actinomycin D, camptothecin, etoposide, and melphalan),76 raises the possibility that ROS interferes with other apoptotic pathways in DNR-treated cells. Furthermore, there is mounting evidence that ROS activates or mediates many other signaling pathways, which can contribute more generally to the cellular response to DNR. For example, ROS modulates both PKC77-79 and tyrosine kinase activities,80-85contributes to cell cycle block,86 stimulates Raf-1/ERK mitogen-activated protein (MAP) kinases,84,87 and triggers activation of critical transcription factors, including nuclear factor-κB (NF-κB) (see Baeuerle and Henkel for review88), a negative regulator of DNR-induced apoptosis,89 which we will discuss later. These results suggest a complex role for ROS, which may mediate both cell death and survival signaling pathways, and thereby could play a major role in orientating the cellular response to genotoxic insult.

Anthracyclines, as other genotoxic agents including alkylating agents and ionizing radiation, have been shown to increase cellular DAG levels and PKC activity.90 However, the source of DAG as well as its functional role in the cellular response to the drug were not determined. In another study, we have reported that, in U937 cells, DNR (and mitoxantrone) transiently stimulated concurrently with CER generation both DAG and phosphorylcholine production by phospholipase C hydrolysis of phosphatidylcholine. Moreover, pretreatment of cells with the xanthogenate compound D609, a potent inhibitor of phosphatidylcholine hydrolysis, led to a sustained increase in CER levels. This result suggests that DNR may trigger in parallel both cell death (CER) and survival (DAG and phosphorylcholine) mediators and that, in sensitive leukemic cells, CER overrides the protective function of DAG and phosphorylcholine.91 These results support the notion that reciprocal regulation through DAG and CER may be implicated in the regulation of apoptosis.92 The mechanism by which phosphatidylcholine-derived DAG influenced intracellular CER concentration in DNR-treated cells is not yet characterized; however, because phosphatidylcholine-derived DAG did not appear to influence N-SMase stimulation, it is conceivable that DAG enhanced CER metabolism by facilitating sphingomyelin synthesis. Furthermore, it has been documented that phosphatidylcholine-derived DAG binds and stimulates Raf-1 kinase activity93,94 and, through Raf-1, may activate the MEK1/ERK classical MAP kinase module, a negative regulator of apoptosis induced by various stresses.84,87,95-97 Phosphatidylcholine-derived DAG has been found to stimulate some PKC isoforms such as PKC ζ or ι “atypical” isoforms that have been involved in cell proliferation and/or survival.98-100 In fact, we have recently observed that DNR stimulates Raf-1, ERK1, and PKC ζ activities through a D609-inhibitable mechanism (V Mansat-De Mas, I Plo, C Bezombes, et al; unpublished results, November 2000). Altogether these findings suggest that, in DNR-treated cells, phosphatidylcholine-derived DAG potentially operates by modulating the activity of diverse serine kinases and thereby contributes to reduce drug cytotoxicity. Regardless of the mechanism by which phosphatidylcholine-derived DAG exerts its protective function, this result may have some clinical implications. Indeed, it has been demonstrated that p21Ras or src oncogenes, as well as cytokines, including IL-3, GM-CSF, and TNFα, are potent activators of phosphatidylcholine hydrolysis and thereby may induce sustained DAG production.101-104 Therefore, it is conceivable that, in some AML cells, expression of some oncogenic products, as well as stimulation by cytokines, may severely impair DNR-induced apoptosis through a phosphatidylcholine-derived DAG-dependent mechanism.

Among other enzymes that are involved in signal transduction pathways, phosphoinositide 3-kinase (PI3K) plays an important role. PI3Ks are a family of enzymes that catalyze the phosphorylation of inositol lipids at the D3 position of the inositol ring, generating new intracellular second messengers (see Franke et al105 for review). The lipid products of PI3K are phosphatidylinositol-3-phosphate (PtdIns-3-P), phosphatidylinositol-3,4-biphosphate (PtdIns-3,4-P2), and phosphatidylinositol-3,4,5-triphosphate (PtdIns-3,4,5-P3). PtdIns-3,4-P2 and PtdIns-3,4,5-P3 have been demonstrated to interact with a large variety of downstream effectors, including serine-threonine kinase Akt,106 calcium-insensitive PKCδ, ε, and η isoforms,107 as well as atypical PKC ζ isoform108 or phospholipase C.109 The use of PI3K inhibitors, wortmannin and LY294002, and PI3K mutants has demonstrated a role for PI3K in cell survival after various stresses, including γ or UV irradiation.110,111 For this reason, we have investigated the implication of PI3K in the cellular response to DNR in U937 cells. In fact, DNR induced a 2-fold increase in PI3K activity with transient accumulation of PtdIns-3,4-P2 and PtdIns-3,4,5-P3, and Akt activation. Wortmannin and LY294002 also accelerated DNR-induced apoptosis, suggesting that PI3K stimulation may contribute to cell survival.112 However, as already described earlier for phospholipase C activation by DNR, the stimulation of PI3K integrates into a futile response of the cells to the drug and only slightly affect DNR cytotoxicity. However, according to a recent report, it appears that PI3K may play a much more prominent protective effect against doxorubicin-induced apoptosis.113 Moreover, these results suggest that potent and sustained PI3K activation induced by oncogenic products, such as Ras114 or BCR-ABL115-117, or by mutated forms of HGF receptors, such as Flt3118, may result in significant protection toward DNR and perhaps other antileukemic agents. The mechanism by which PI3K or PI3K products interfere with DNR-induced apoptosis is still under investigation. However, from other studies, one can speculate that the PI3K protective function involves Akt-mediated phosphorylation of Bcl-2 family,119inhibition of caspase activity,120,121 or stimulation of PKC isoform activities.107,108 Moreover, it has been reported that cell-permeant CER decreases Akt activity,122suggesting possible cross-talk between sphingomyelin-CER and PI3K-Akt pathways in DNR-treated cells.

NF-κB is a member of the mammalian Rel family of transcriptional activators that plays a central role in the regulation of immune responses, embryogenesis, and hematopoiesis. NF-κB is a heterodimer consisting of the 50-kD (p50) and 65-kD (p65) subunits that, in its inactive state, is located in the cytoplasm bound to an inhibitory protein (IκBα). On activation, NF-κB disassociates from IκBα, translocates to the nucleus, and binds to DNA to regulate the expression of many genes.88 By using electrophoretic mobility shift assays, it has been shown that NF-κB DNA binding activity is stimulated by cytotoxic effectors, including DNA-damaging agents, such as ionizing radiation, alkylating agents, anthracyclines, topoisomerase inhibitors, and cytosine arabinoside.89,123-130 However, the fact that nongenotoxic anticancer compounds such as vinca alkaloids and taxanes125 may also activate NF-κB suggests that DNA damage is not necessarily needed for drug-induced NF-κB activation. This finding could mean that these drugs share some NF-κB–activating signaling pathways that can be generated in the cytosol but not in the nucleus, similar to that observed in the UV response in which NF-κB activation was clearly demonstrated to be initiated not in the nucleus but at or near the plasma membrane through a Src- and Ras-dependent mechanism.80 DNR-induced NF-κB activation does not require de novo protein synthesis and appears to be specific.125 

Despite many efforts, the mechanism by which DNR activates NF-κB is not totally understood. Because, on one hand, NF-κB is considered to be an oxidative stress-responsive transcription factor,88and, on the other hand, DNR is known to generate ROS, one could speculate that ROS could be involved in DNR-induced NF-κB activation. However, the role of ROS was not firmly established. Indeed, whereas the antioxidant pyrrolidine dithiocarbamate was found to inhibit DNR-induced NF-κB activation in HL-60 and Jurkat T cells,124 opposite results have been reported in a human colon carcinoma cellular model.126 Moreover, mitoxantrone, which is unable to undergo redox cycling, was also found to activate NF-κB in a pyrrolidine dithiocarbamate–insensitive manner, suggesting that the inhibitory effect of pyrrolidine dithiocarbamate could not be related to its antioxidant property.129 CER was also a plausible candidate. Indeed, previous studies have shown that the addition of CER induced NF-κB activation.130 In fact, we found that the inhibition of DNR-induced SMase stimulation and subsequent CER production by serine-protease inhibitors reduces the capacity of the drug to activate NF-κB.131However, because of the broad specificity of these agents, the role of CER was not totally established, and one could not exclude a CER-independent mechanism as it has been proposed for TNFα-induced NF-κB activation.132,133 On the basis of previous studies which have shown that phosphorylation events are critical for NF-κB activation,88 the role of PKC has been investigated. Also, PKC inhibitors were found to block DNR-induced NF-κB activation,125 suggesting that DNR-triggered PKC stimulation plays an important role in NF-κB activation. In fact, previous studies have already suggested that PKC may act indirectly on phosphorylation and subsequent degradation of IκB.88More recent studies have indicated that IκB phosphorylation is under control of 2 IκB kinases (IKKα and ΙΚΚβ) which phosphorylate residues 32 and 36 of IκBα and that atypical PKC isoforms are critical regulators of the IKK activity.134Altogether these results render those PKC isoforms plausible candidates for mediating anthracycline-induced NF-κB activation. Finally, one cannot rule out the role of PI3K/Akt pathway. Indeed, it has been shown that activated Akt directly interacts and phosphorylates IκB-kinase (IKK).135,136 Whether or not DNR-induced PI3K/Akt pathway is involved in DNR-induced NF-κB activation should be examined.

Previous studies have shown that NF-κB inhibits apoptosis and/or enhances survival in a large variety of hematopoietic cells, including B lymphocytes,137 Hodgkin disease cells,138and CD34+ bone marrow cells.139 Therefore, it was tempting to speculate that this transcription factor provides significant protection against DNR-induced cytotoxicity. Such a hypothesis was supported by the fact that NF-κB was found to play an essential role in preventing TNFα-induced cell death.89,140,141 To address this question more directly for DNR, Wang et al89 have used a gene construct coding for a super-repressor form of the NF-κB inhibitor IκBα. This IκB repressor contains mutations, which inhibit signal-induced phosphorylation and subsequent proteasome-mediated degradation of IκB. This mutant protein binds to NF-κB and prevents nuclear translocation as well as DNA binding.142,143 Using this methodology, those investigators have shown that NF-κB inhibition resulted in increased cytotoxic effect of DNR (and ionizing radiation) in vitro.89 Although other studies suggest that the role of NF-κB in drug resistance may be a function of the cellular model,127,128 these results may have important clinical implications. Indeed, some pharmacologic agents are known to inhibit cytokine-induced NF-κB activation, including glucocorticoids, which act through induction of IκB synthesis.144,145Glucocorticoids are largely used in association with anthracyclines as part of front-line therapy of myeloma, acute lymphoid leukemia, and lymphoma. Whether or not glucocorticoids act in synergy with anthracyclines in lymphoid cells through a NF-κB–dependent mechanism has not been evaluated. Altogether these findings provide new insights in the understanding of synergistic drug combinations currently used in the clinical setting. These findings also provide a rationale for novel chemosensitization strategies based on NF-κB pathway inhibition. For example, it is possible that liposomal or viral delivery of an IκB super-repressor could result in sensitizing tumor cells in vivo to DNR, as it has been shown for the topoisomerase I inhibitor CPT-11.146 It is also conceivable to interfere with proximal upstream regulators of NF-κB activation (ie, Akt or PKCζ by inhibiting IKK activation147).

The mechanism by which NF-κB contributes to apoptosis inhibition remains uncertain. It has been suggested that NF-κB is a negative regulator of p53 gene expression, and it has also been reported that p53-activating signal is partially blocked by inhibition of NF-κB activation in DNR-treated cells.126 

Doxorubicin, as other antitumor compounds such as methotrexate, cisplatin, fludarabine, or bleomycin as well as ionizing radiation, was shown to enhance the expression of Fas and Fas-L on the surface of certain leukemic or epithelial malignant cells. For this reason, it has been proposed that doxorubicin-induced apoptosis occurs through autocrine or paracrine induction of the Fas-dependent pathway.148-151 In these studies, this hypothesis was supported by 2 lines of evidence: (1) cell lines resistant to Fas were found insensitive to anticancer drug-induced apoptosis, and (2) doxorubicin-induced apoptosis was prevented by CD95-neutralizing antibodies. However, this issue remains highly controversial mainly because antagonistic anti-Fas antibodies do not always inhibit drug-induced cell death.152-154 Moreover, many cells either naturally resistant to Fas (ie, HL-60 cells) or selected for Fas resistance152-154 remain sensitive to doxorubicin-induced apoptosis. Therefore, convincing evidence now exists showing that Fas/Fas-L pathway is not a principal and necessary mechanism of anthracycline-induced apoptosis.154 

Other investigators have reported that in carcinoma cells doxorubicin induced the clustering of Fas receptor and its interaction with Fas-associated death domain-containing protein (FADD) in a Fas-L–independent fashion.155 This has also been described for UV and ionizing radiation.156,157 FADD is an adapter molecule, which is recruited to Fas cell death domain, and then binds to and activates procaspase-8; active caspase-8, in turn, triggers activation of a proteolytic cascade that leads through caspase-7, caspase-3, and caspase-6 to apoptosis. Moreover, these investigators showed that FADD overexpression facilitated drug-induced apoptosis, whereas down-regulation of FADD by transient transfection of an antisense construct decreased tumor cell sensitivity to the drug.155 For this reason, it has been proposed that doxorubicin-induced cell death involves the Fas/FADD pathway without interfering with Fas-L production and that FADD expression may significantly influence the cellular response to this drug. Fas clustering by cytotoxic agents was also described for VP-16, cisplatinum, and vinblastin155 as well as for UV radiation.157 However, the role of FADD in doxorubicin-induced caspase activation and apoptosis may be a function of the cellular model. Indeed, in doxorubicin-treated Jurkat leukemic T cells, Wesselborg et al154 have shown a correlation between doxorubicin-apoptosis and cleavage of procaspase-8 to its active p18 subunit; however, the expression of a dominant-negative FADD construct selectively abrogated Fas but not drug-induced effects. This result suggests that caspase-8 can be activated by doxorubicin in the absence of Fas receptor signaling.154 Fas- and FADD-independent caspase-8 activation have been observed in lymphoid cells treated with ionizing radiation.158 Altogether these results suggest that, whatever the role of FADD, caspase-8 activation represents a critical step in the cascade resulting in terminal proteolytic events, including caspase-3 and caspase-6 activation responsible for the cleavage of poly(ADP-ribose) polymerase and nuclear lamins, respectively. However, other investigators have questioned the role of caspase-8. For example, Villunger et al153 have reported that expression of cowpox virus cytokine response modifier A, a potent inhibitor of distinct members of the caspase-protease family, including caspase-8,159 did not influence doxorubicin-induced apoptosis in T-acute lymphatic leukemia CEM cells, whereas it prevented Fas-mediated apoptosis. Moreover, expression of FLIP (FLICE-inhibitory protein), which binds to caspase-8 and interferes with its function, was found to have no influence on apoptosis induced by doxorubicin or by other antileukemic compounds or by γ irradiation, whereas it abrogated Fas-mediated apoptosis in Jurkat T cells.160 Although the implication of Fas system and/or Fas signaling proximal adapter molecules including FADD or caspase-8 remains a controversial issue, the role of caspase-3 in doxorubicin-induced apoptosis was firmly established through different approaches, including peptide inhibitor and antisens.161However, the fact that these inhibitors were effective, even if added several hours after drug treatment, indicates that this caspase is involved in the execution and not in the triggering phase of drug action.161 Finally, it should be pointed out that the link, if any, between caspase signaling and CER pathway activated by anthracyclines is not yet determined. Indeed, previous studies have established that cytokine response modifier A–induced caspase inhibition resulted in the inhibition of CER generation and cell death induced by cytotoxic molecules such as TNFα162 or Fas agonist,163 suggesting a role for caspase in controlling SMase stimulation. Whether or not caspase-mediated proteolytic events take place upstream, CER generation in anthracycline-treated cells has not been yet investigated.

Whatever the intimate mechanism by which Fas and caspases are involved in the cellular response to anthracyclines, all these results have important clinical implications. First, on the basis of the stimulatory effect of anthracyclines on Fas-L production in some tumor cells and with the demonstration that Fas-L produced by tumor cells can kill the specific effector CTL and other activated T cells that express Fas,164 it can be speculated that chemotherapy may decrease cellular cytotoxicity of natural effectors and, therefore, may facilitate immune escape.165 Conversely, it has been proposed that drug-induced Fas expression may result in sensitization toward Fas-dependent cytotoxicity of cellular immune effectors.166-169 Second, previous studies have shown that, whereas short-term culture with doxorubicin enhanced Fas expression level, prolonged exposure to the drug may result in Fas reduction or even lack of expression, establishing an intriguing link between MDR phenotype and Fas resistance.170 Similar results have been obtained with mitoxantrone.171 Third, if Fas signaling molecules (or Fas itself) play an important role in anthracycline-induced apoptosis, it is conceivable that diminution of their expression and/or altered capacity to form the multimolecular death-initiating signaling complex constituted by Fas death domain, FADD, and procaspase-8 may impair drug cytotoxicity. In this perspective, it is important to note that AML cells (as well as immature progenitor cells) are generally insensitive to Fas commitment although most of them do express Fas, suggesting a disruption in Fas-mediated death signaling.172-174 Therefore, it is tempting to speculate that negative control of Fas signaling contributes not only to immune escape but also to decreased drug cytotoxicity in leukemia cells.

Finally, one should not disregard the emerging evidence that genotoxic stress, such as anthracycline treatment, has been described to increase death receptors such as DR5 for TRAIL/Apo-2 ligand in leukemia cells lines such as U937 and HL-60.175,176 These observations strongly suggest that one may optimize the antileukemic activity of anthracyclines by combining them with Apo-2 ligand treatment.

Previous studies have shown that doxorubicin, as many other DNA-damaging agents, activates p53-DNA binding.177 On the basis of the crucial role of p53 in the execution of some forms of apoptosis, it has been therefore speculated that p53 could play an important function in anthracycline cytotoxicity. In fact, the requirement for wild-type p53 for apoptosis after doxorubicin exposure has been demonstrated in rodent normal or minimally transformed fibroblasts and lymphocytes.178,179 However, the role of p53 mutations in drug-induced apoptosis and cytotoxicity in human tumor cells is much less clear (see Brown and Wouters for review180). As far as leukemic cells are concerned, it should be noted that most AML cell lines displayed mutated or deleted forms of p53, including U937 or HL-60 cells, which exhibit high sensitivity to DNR. This observation suggests that p53 plays a minor role in anthracycline-induced cytotoxicity in AML cells. However, some clinical studies have shown that p53 mutations predict poor clinical outcome in patients treated with anthracycline-containing regimens for hematologic neoplasias such as AML or large cell non-Hodgkin malignant lymphomas.181,182 It is therefore possible that loss of p53 function correlates with other drug resistance mechanisms or interferes with other apoptotic pathways. In this perspective, it is important to note that wild-type p53 is necessary for c-Myc–mediated facilitation of doxorubicin-induced apoptosis.183 

Besides its role in apoptosis, p53 plays an important function in regulating cell cycle transition in doxorubicin-treated cells. Indeed, it has been shown that doxorubicin-induced p53 activation contributes to the induction of the WAF1/CIP1 p21 gene product, which is a strong inhibitor of cyclin-dependent kinases involved in G1 to S transition.184 Although p53-independent doxorubicin-induced WAF1/CIP1 has been described,185,186this mechanism may account for G1 block after anthracycline exposure in p53 proficient cells. It has been suggested that WAF1 expression protects cells from doxorubicin-induced cytotoxicity because G1 block facilitates complete repair of DNA damage before the cells undergo DNA replication. In fact, it has been shown that high levels of constitutive WAF1/CIP1 protein are associated with chemoresistance in AML.187 

Other cell cycle alterations have been described in doxorubicin-treated cells. Indeed, depending on the dose and cellular model, anthracyclines as many other genotoxic agents may also induce G2-M block because of p34cdc2 kinase inhibition. Indeed, previous studies have shown that doxorubicin prevented p34cdc2 dephosphorylation through a cdc25-independent mechanism, resulting in alteration of p34cdc2/cyclin B1-mediated complex.188 The mechanism by which anthracyclines interfere with p34cdc2 function has not been carefully examined. However, Kharbanda and coworkers189,190 have shown that most DNA damaging agents, including cytosine arabinoside, mitomycin C, and ionizing radiation, induce activation of p56/p53lyn, a tyrosine kinase of the Src family, which, in turn, phosphorylates and inactivates p34Cdc2. Whether or not Lyn is involved in anthracycline-induced p34cdc2 kinase inhibition and subsequent G2-M block should be confirmed.

It is now admitted that the extracellular matrix (ECM) plays an important role in cell proliferation and survival through a complex network of signals regulating cytoplasmic kinases, growth factor receptors, and ion channels.191 In another study, it has been demonstrated that β1-integrin–mediated adhesion to ECM proteins protects tumor epithelial cells from doxorubicin-induced apoptosis.192 This study also provides strong evidence that ECM-activated outside-in signals involve caspase inhibition through a protein tyrosine kinase mechanism, whereas adhesion to ECM does not influence DNA damage.192 In another study, it has been reported that myeloma cells adhered to fibronectin, a predominant component of ECM, have a survival advantage over nonadhered cells after doxorubicin exposure.193 The fact that ECM adhesion offers protection against anthracyclines may have important implications in other clinical settings. Indeed, it has been documented that both normal and leukemic myeloid progenitors strongly interact with ECM in the bone marrow and that β1-integrins play a predominant role in this process.194 Therefore, it is possible that contact with ECM contributes significantly to the natural chemoresistance of immature hematopoietic cells. Moreover, it cannot be excluded that anthracyclines activate inside-out signals, which modulate stem cell/ECM interactions, and, for example, facilitate the mobilization of progenitor subsets from the bone marrow to the peripheral circulation. This possibility could explain the efficacy of anthracycline-contained regimens used for CD34+ cell mobilization before peripheral stem cell transplantation. Of course, the role of ECM relative to intracellular antiapoptotic pathways is at present unknown.

It is evident from these investigations that apoptosis induced by DNR brings into play a complex network of coordinated and highly controlled events. Although the characterizations of these different signaling pathways are still incomplete, we can at present attempt to concisely recapitulate (with a little speculation) as to the individual implication of these signaling pathways and mediators in DNR-induced cell signaling (Figure 1).

Fig. 1.

Daunorubicin triggers both apoptotic and survival pathways.

DNR indicates daunorubicin; ROS, reactive oxygen species; SM, sphingomyelin; PC, phosphatidylcholine; SMase, sphingomyelinase; PC-PLC, phosphatidylcholine-specific phospholipase C; DAG, diacylglycerol; PI3K, phosphoinositide 3-kinase; PKC, protein kinase C; JNK, c-Jun N-terminal kinase; AP-1, activated protein-1; MEK, mitogen-activated protein kinase kinase 1; NF-κB, nuclear factor-κB.

Fig. 1.

Daunorubicin triggers both apoptotic and survival pathways.

DNR indicates daunorubicin; ROS, reactive oxygen species; SM, sphingomyelin; PC, phosphatidylcholine; SMase, sphingomyelinase; PC-PLC, phosphatidylcholine-specific phospholipase C; DAG, diacylglycerol; PI3K, phosphoinositide 3-kinase; PKC, protein kinase C; JNK, c-Jun N-terminal kinase; AP-1, activated protein-1; MEK, mitogen-activated protein kinase kinase 1; NF-κB, nuclear factor-κB.

Close modal

Present knowledge suggests that in drug-sensitive cells clinically relevant concentrations of DNR trigger within minutes the generation of ROS that leads to a first wave of N-SMase activation, sphingomyelin hydrolysis, and consequently CER generation. This process makes the initial ROS burst the rate-limiting step in DNR-induced apoptosis signaling, and one can consider these early events (within the first 5 minutes) as the primary apoptotic initiation step. Then follows a cyclical cascade of ROS-dependent N-SMase activation and CER generation that perpetuates for about 4 hours until the apoptotic execution steps (caspase activation, mitochondrial depolarization, etc) come into play. It remains to be clearly determined, however, whether these cyclical events (which are p53-independent) are critical in the apoptotic signaling process. For example, does the inhibition of continual CER or ROS production significantly affect DNR-induced apoptosis (as is suggested by studies in which the overexpression of Bcl2 [thereby inhibiting DNR-induced apoptosis] failed to block initial CER generation)? The ROS-dependent sphingomyelin-CER pathway has been shown to be responsible for rapid activation of the MEKK1-SEK1-JNK cascade leading to enhanced DNA binding activity of the transcription factor AP-1. This latter pathway appears essential in DNR-triggered apoptosis. Of course, all of these events are absent in drug-resistant cells, and the literature proposes several potential mechanisms such as rapid CER metabolism into other lipid products (such as sphingosine-1-phosphate or glucosylceramide) that possess survival/proliferative properties. However, it remains to be determined to what extent (eg, which is dominant?) these metabolic events play a role in DNR resistance of myeloid leukemic cells.

Remarkably, in drug-sensitive cells, DNR also triggers pathways that should negatively regulate apoptosis. In parallel, a phospholipase C–dependent DAG/raf-1/MEK cascade leading to activation of the transcription factor NF-κB and a DAG-independent PI3K/PKCζ cascade play a significant role in cellular protection. Inhibition (pharmacologically or genetically) of one or more of these mediators significantly increases the apoptotic response to DNR. One can, of course, speculate that these “survival” pathways are overridden by the CER pathway in sensitive cells but are predominant in resistant cells because the CER pathway is effectively down-regulated. But it appears obvious that more research is needed to better understand the relationship between both proapoptotic and antiapoptotic pathways triggered by DNR (interdependency, cross-talk, degree to which they occur in dose-dependent studies, and, of course, their relevance in normal myeloid cells). Finally, the emerging data on gene induction by anthracyclines, based on cDNA microarray technology, raise the question of not only the role of these genes (such as DNA-damage-response genes, eg, GADD) in cell resistance but also the link between such gene induction and the above-mentioned drug-triggered signaling pathways.195,196 

In conclusion, we now know that DNR triggers both positive and negative regulatory pathways of apoptosis; therefore, it is now essential to discriminate among resistance mechanisms of AML cells: those that lead to decreased drug-target interactions and those that interfere with cell death commitment. It will also most certainly be of great importance to delineate the role of effector-induced cell damage in programmed cell death signaling, and it will also be necessary to determine if the discrimination between apoptosis and necrosis is an essential phenomenon for in vivo therapeutics. Finally, this review incites for a reinterpretation of the actions of well-established therapeutic agents in the light of recent advances in the basic sciences that should allow cellular pharmacology of antineoplastic agents to continue gathering momentum in the perspective of overcoming drug resistance by defining pharmacologic strategies capable of sensitizing resistant tumor cells and/or protecting normal physiologic cells to DNR.197 

We apologize to colleagues whose works were not cited because of the size restrictions of the review.

Supported by la Ligue Nationale Contre le Cancer and les Comités Départementaux du Gers, de l'Aveyron, du Lot et de la Haute-Garonne (J.P.J.), in part by l'Association pour la Recherche sur le Cancer grant 5526 (G.L.), by La Faculté de Médecine Toulouse-Rangueil (G.L.), and by le Centre National d'Etudes Spaciales (J.P.J.).

1
Myers
CE
Chabner
BA
Antracyclines.
Cancer Chemotherapy. Principles and Practice.
Chabner
BA
Collins
JM
1990
356
381
JB Lippincott
Philadelphia, PA
2
Tritton
TR
Cell surface actions of adriamycin.
Pharmac Ther.
49
1991
293
309
3
Potmesil
M
Kirschenbaum
J
Israel
M
Levin
M
Khetarpal
VK
Silber
R
Relationship of adriamycin concentrations to the DNA lesions induced in hypoxic and euoxic L1210 cells.
Cancer Res.
43
1983
3528
3533
4
Ross
WE
Glaubiger
K
Kohn
KW
Qualitative and quantitative aspects of intercalator-induced DNA strand breaks.
Biochim Biophys Acta.
5629
1979
41
50
5
Zwelling
LA
Kerrigan
D
Michaels
S
Cytotoxicity and DNA strand breaks by 5-iminodaunorubicin in mouse leukemia L1210 cells: comparison with adriamycin and 4′-(9-acridinylamino)- methanesulfon-m-aniside.
Cancer Res.
42
1982
2687
2691
6
Quillet-Mary
A
Mansat
V
Duchayne
E
et al
Daunorubicin-induced internucleosomal DNA fragmentation in acute myeloid cell lines.
Leukemia.
10
1996
417
425
7
Bailly
JD
Muller
C
Jaffrézou
JP
et al
Lack of correlation between expression and function of P-glycoprotein in acute myeloid leukemic cells.
Leukemia.
9
1995
799
807
8
Obeid
LM
Linardic
CM
Karolak
LA
Hannun
YA
Programmed cell death induced by ceramide.
Science.
259
1993
1769
1771
9
Haimovitz-Friedmann
A
Kan
CC
Ehleiter
D
et al
Ionizing radiation acts on cellular membranes to generate ceramide and initiate apoptosis.
J Exp Med.
180
1994
525
535
10
Jaffrézou
JP
Levade
T
Bettaieb
A
et al
Daunorubicin induced apoptosis: triggering of ceramide generation through sphingomyelin hydrolysis.
EMBO J.
15
1996
2417
2424
11
Bezombes
C
Ségui
B
Cuvillier
O
et al
Lysosomal sphingomyelinase is not solicited for apoptosis signaling.
FASEB J.
15
2001
297
299
12
Bose
R
Verheij
M
Haimovitz-Friedman
A
Scotto
K
Fuks
Z
Kolesnick
R
Ceramide synthase mediates daunorubicin-induced apoptosis: an alternative mechanism for generating death signals.
Cell.
82
1995
405
414
13
Xu
J
Chen-Hsiung
Y
Chen
S
et al
Involvement of de novo ceramide biosynthesis in tumor necrosis factor-α/cycloheximide-induced cerebral endothelial cell death.
J Biol Chem.
273
1998
16521
16526
14
Bourteele
S
Hauber
A
Döppler
H
et al
Tumor necrosis factor induces ceramide oscillations and negatively controls sphingolipid synthases by caspases in apoptotic Kym-1 cells.
J Biol Chem.
273
1998
31245
31251
15
Jaffrézou
JP
Maestre
N
Mansat
V
Bezombes
C
Levade
T
Laurent
G
Positive feedback control sphingomyelinase activity by ceramide.
FASEB J.
12
1998
999
1006
16
Verheij
M
Bose
R
Lin
XH
et al
Requirement for ceramide-initiated SAPK/JNK signalling in stress-induced apoptosis.
Nature.
380
1996
75
79
17
Sawai
H
Okazaki
T
Yamamoto
H
et al
Requirement of AP-1 for ceramide-induced apoptosis in human leukemia HL-60 cells.
J Biol Chem.
270
1995
27326
27331
18
Lange-Carter
CA
Johnson
GL
Ras-dependent growth factor regulation of MEK kinase in PC12 cells.
Science.
265
1994
1458
1461
19
Johnson
NL
Gardner
AM
Diener
KM
et al
Signal transduction pathways regulated by mitogen-activated/extracellular response kinase kinase kinase induce cell death.
J Biol Chem.
271
1996
3229
3237
20
Ichijo
H
Nishida
E
Irie
K
et al
Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signalling pathways.
Science.
275
1997
90
94
21
Osborn
MT
Chambers
TC
Role of the stress-activated/c-Jun NH2-terminal protein kinase pathway in the cellular response to adriamycin and other chemotherapeutic drugs.
J Biol Chem.
271
1996
30950
30955
22
Yu
R
Shtil
AA
Tan
TH
Roninson
IB
Kong
AN
Adriamycin activates c-jun N-terminal kinase in human leukemia cells: a relevance to apoptosis.
Cancer Lett.
107
1996
73
81
23
Mansat-De Mas
V
Bezombes
C
Quillet-Mary
A
et al
Implication of radical oxygen species in ceramide generation, c-Jun N-terminal kinase activation and apoptosis induced by daunorubicin.
Mol Pharmacol.
56
1999
867
874
24
Devary
Y
Gottlieb
RA
Smeal
T
Karin
M
The mammalian ultraviolet response is triggered by activation of Src tyrosine kinases.
Cell.
71
1992
1081
1091
25
Kharbanda
S
Ren
R
Pandley
P
et al
Activation of the c-Abl tyrosine kinase in the stress response to DNA-damaging agents.
Nature.
376
1995
785
788
26
Shafman
TD
Saleem
A
Kyriakis
J
Weichselbaum
R
Kharbanda
S
Kufe
DW
Defective induction of stress-activated protein kinase activity in ataxia-telangiectasia cells exposed to ionizing radiation.
Cancer Res.
55
1995
3242
3245
27
Chen
Y-R
Meyer
CF
Tan
T-H
Persistant activation of c-Jun N-terminal kinase 1 (JNK1) in γ radiation-induced apoptosis.
J Biol Chem.
27
1996
631
634
28
Kharbanda
S
Pandey
P
Ren
R
Mayer
BJ
Zon
L
Kufe
D
c-Abl activation regulates induction of the SEK1/stress-activated protein kinase pathway in the cellular response to 1-β-D-arabinofuranosylcytosine.
J Biol Chem.
270
1995
30278
30281
29
Amato
SF
Swart
JM
Berg
M
Wanebo
HJ
Mehta
SR
Chiles
TC
Transient stimulation of the c-Jun-NH2-terminal kinase/activator protein 1 pathway and inhibition of extracellular signal-regulated kinase are early effects in paclitaxel-mediated apoptosis in human B lymphoblasts.
Cancer Res.
58
1998
241
247
30
Lee
L-F
Li
G
Templeton
DJ
Ting
JP
Paclitaxel (Taxol)-induced gene expression and cell death are both mediated by the activation of c-Jun NH2-terminal kinase (JNK/SAPK).
J Biol Chem.
273
1998
28253
28260
31
Kyriakis
JM
Avruch
J
Sounding the alarm: protein kinase cascades activated by stress and inflammation.
J Biol Chem.
271
1996
24313
24316
32
Mansat
V
Laurent
G
Levade
T
Bettaı̈eb
A
Jaffrézou
JP
The protein kinase C activators phorbol esters and phosphatidylserine inhibit neutral sphingomyelinase activation, ceramide generation, and apoptosis triggered by daunorubicin.
Cancer Res.
57
1997
5300
5304
33
Chmura
SJ
Nodzenski
E
Weichselbaum
RR
Quintans
J
Protein kinase C inhibition induces apoptosis and ceramide production through activation of a neutral sphingomyelinase.
Cancer Res.
56
1996
2711
2714
34
Linardic
CM
Hannun
YA
Identification of a distinct pool of sphingomyelin involved in the sphingomyelin cycle.
J Biol Chem.
269
1994
23530
23537
35
Bailly
JD
Skladanowski
A
Bettaieb
A
Mansat
V
Larsen
AK
Laurent
G
Natural resistance of acute myeloid leukemia cell lines to mitoxantrone is associated with lack of apoptosis.
Leukemia.
11
1997
1523
1532
36
Bettaieb
A
Record
M
Côme
MG
et al
Opposite effects of tumor necrosis factor α on the sphingomyelin-ceramide pathway in two myeloid leukemia cell lines: role of transverse sphingomyelin distribution in the plasma membrane.
Blood.
88
1996
1465
1472
37
Bezombes
C
Maestre
N
Laurent
G
Levade
T
Bettaieb
A
Jaffrézou
JP
Restoration of TNF-α-induced ceramide generation in resistant human leukemia KG1a cells by the P-glycoprotein blocker PSC833.
FASEB J.
12
1998
101
109
38
Tepper
AD
Ruurs
P
Wiedmer
T
Sims
PJ
Borst
J
van Blitterswijk
WJ
Sphingomyelin hydrolysis to ceramide during the execution phase of apoptosis results from phospholipid scrambling and alters cell-surface morphology.
J Cell Biol.
150
2000
155
164
39
Luberto
C
Hannun
YA
Sphingomyelin synthase, a potential regulator of intracellular levels of ceramide and diacylglycerol during SV40 transformation: does sphingomyelin synthase account for the putative phosphatidylcholine-specific phospholipase C?
J Biol Chem.
273
1998
14550
14559
40
Hampton
RY
Morand
OH
Sphingomyelin synthase and PKC activation.
Science.
246
1989
1050
41
Ségui
B
Bezombes
C
Uro-Costes
E
et al
Stress-induced apoptosis is not mediated by endolysosomal ceramide.
FASEB J.
14
2000
36
47
42
Coroneos
E
Martinez
M
McKenna
S
Kester
M
Differential regulation of sphingomyelinase and ceramidase activities by growth factors and cytokines: implications for cellular proliferation and differentiation.
J Biol Chem.
270
1995
23305
23309
43
Cuvillier
O
Pirianov
G
Kleuser
B
et al
Suppression of ceramide-mediated programmed cell death by sphingosine-1-phosphate.
Nature.
381
1996
800
803
44
Cuvillier
O
Rosenthal
DS
Smulson
ME
Spiegel
S
Sphingosine 1-phosphate inhibits activation of caspases that cleave poly(ADP-ribose) polymerase and lamins during Fas- and ceramide-mediated apoptosis in Jurkat T lymphocytes.
J Biol Chem.
273
1998
2910
2916
45
Kleuser
B
Cuvillier
O
Spiegel
S
1α,25-dihydroxyvitamin D3 inhibits programmed cell death in HL-60 cells by activation of sphingosine kinase.
Cancer Res.
58
1998
1817
1824
46
Perez
GI
Knudson
CM
Leykin
L
Korsmeyer
SJ
Tilly
JL
Apoptosis-associated signaling pathways are required for chemotherapy-mediated female germ cell destruction.
Nat Med.
3
1997
1228
1232
47
Shayman
JA
Deshmukh
GD
Mahdiyoun
S
et al
Modulation of renal epithelial cell growth by glucosylceramide: association with protein kinase C, sphingosine, and diacylglycerol.
J Biol Chem.
266
1991
22968
22974
48
Abe
A
Radin
NS
Shayman
JA
et al
Structural and stereochemical studies of potent inhibitors of glucosylceramide synthase and tumor cell growth.
J Lipid Res.
36
1995
611
621
49
Rani
CS
Abe
A
Chang
Y
et al
Cell cycle arrest induced by an inhibitor of glucosylceramide synthase: correlation with cyclin-dependent kinases.
J Biol Chem.
270
1995
2859
2867
50
Liu
YY
Han
TY
Giulano
AE
Cabot
MC
Expression of glucosylceramide synthase, converting ceramide to glucosylceramide, confers adriamycin resistance in human breast cancer cells.
J Biol Chem.
274
1999
1140
1146
51
Lavie
Y
Cao
H-T
Volner
A
et al
Agents that reverse multidrug resistance, tamoxifen, verapamil, and cyclosporin A, block glycosphingolipid metabolism by inhibiting ceramide glycosylation in human cancer cells.
J Biol Chem.
272
1997
1682
1687
52
Liu
Y-Y
Han
T-Y
Giulano
AE
Hansen
N
Cabot
MC
Uncoupling ceramide glycosylation by transfection of glucosylceramide synthase antisense reverses adriamycin resistance.
J Biol Chem.
275
2000
7138
7143
53
Come
MG
Bettaieb
A
Skladanowski
A
Larsen
AK
Laurent
G
Alteration of the daunorubicin-triggered sphingomyelin-ceramide pathway and apoptosis in MDR cells: influence of drug transport abnormalities.
Int J Cancer.
81
1999
580
587
54
Lavie
Y
Cao
H-t
Bursten
SL
Giulano
AE
Cabot
MC
Accumulation of glucosylceramide in multidrug-resistant cancer cells.
J Biol Chem.
271
1996
19530
19536
55
Lucci
A
Cho
WI
Han
TY
Giulano
AE
Cabot
MC
Glucosylceramide: a marker for multiple-drug resistant cancers.
Anticancer Res.
18
1998
475
480
56
Lucci
A
Han
TY
Liu
YY
Giuliano
AE
Cabot
MC
Modification of ceramide metabolism increases cancer cell sensitivity to cytotoxics.
Int J Oncol.
15
1999
541
546
57
Lucci
A
Han
TY
Liu
YY
Giuliano
AE
Cabot
MC
Multidrug resistance modulators and doxorubicin synergize to elevate ceramide levels and elicit apoptosis in drug-resistant cancer cells.
Cancer.
86
1999
299
310
58
Cabot
MC
Giulano
AE
Han
T-Y
Liu
Y-Y
SDZ PSC 833, the cyclosporin A analogue and multidrug resistance modulator, activates ceramide synthesis and increases vinblastine sensitivity in drug-sensitive and drug-resistant cancer cells.
Cancer Res.
59
1999
880
885
59
Veldman
RJ
Sietsma
H
Klappa
K
Hoeckstr
D
Kok
JW
Inhibition of P-glycoprotein activity and chemosensitization of multidrug-resistant ovarian carcinoma 2780 AD cells by dexanoyl glucosylceramide.
Biochem Res Commun.
266
1999
492
496
60
Allouche
M
Bettaı̈eb
A
Vindis
C
Rousse
A
Grignon
C
Laurent
G
Influence of Bcl-2 overexpression on the ceramide pathway in daunorubicin-induced apoptosis of leukemic cells.
Oncogene.
14
1997
1837
1845
61
Martin
SJ
Takayama
S
McGahon
A
et al
Inhibition of ceramide-induced apoptosis by Bcl-2.
Cell Death Differ.
2
1995
253
257
62
Jarvis
WDJ
Fornari
FA
Browning
JL
Gewirtz
DA
Kolesnick
RN
Grant
SG
Attenuation of ceramide-induced apoptosis by diglyceride in human myeloid leukemia cells.
J Biol Chem.
269
1994
31685
31692
63
Jarvis
WD
Fornari
FA
Traylor
RS
et al
Induction of apoptosis and potentiation of ceramide-mediated cytotoxicity by sphingoid bases in human myeloid leukemia cells.
J Biol Chem.
271
1996
8275
8284
64
Miyajima
A
Mui
AL-F
Ogorochi
T
Sakamaki
K
Receptors for granulo-macrophage colony-stimulating factor, interleukin-3, and interleukin-5.
Blood.
82
1993
1960
1974
65
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
1757
66
Kaplinski
C
Lotem
J
Sachs
L
Protection of human myeloid leukemic cells against doxorubicin-induced apoptosis by granulocyte-macrophage colony-stimulating factor and interleukin 3.
Leukemia.
10
1996
460
465
67
Laredo
J
Huyn
A
Muller
C
et al
Effect of the protein kinase C inhibitor staurosporine on chemosensitivity to daunorubicin of normal and leukemic fresh myeloid cells.
Blood.
84
1994
229
237
68
Plo
I
Lautier
D
Casteran
N
Dubreuil
P
Arock
M
Laurent
G
Kit activation inhibits daunorubicin-induced sphingomyelin cycle and apoptosis through a phospholipase Cγ and protein kinase C-dependent mechanism.
Hematolgie.
7
2001
25a
69
Sinha
BK
Mimmaugh
EG
Free radicals and anticancer drug resistance: oxygen free radicals in the mechanisms of drug cytotoxicity and resistance by certain tumors.
Free Radicals Biol Med.
8
1990
567
581
70
Pedersen
JZ
Marcocci
L
Rossi
L
Mavelli
I
Rotilio
G
Generation of daunomycin radicals on the outer side of the erythrocyte membrane.
Biochem Pharmacol.
168
1990
240
247
71
Gutteridge
JM
Lipid peroxidation and possible hydroxyl radical formation stimulated by the self-reduction of a doxorubicin-iron (III) complex.
Biochem Pharmacol.
33
1984
1725
1728
72
Liu
B
Andrieu-Abadie
N
Levade
T
Zhang
P
Obeid
LM
Hannun
YA
Glutathione regulation of neutral sphingomyelinase in tumor necrosis factor-α-induced cell death.
J Biol Chem.
273
1998
11313
11320
73
Singh
I
Pahan
K
Khan
M
Singh
A
Cytokine-mediated induction of ceramide production is redox-sensitive.
J Biol Chem.
273
1998
20354
20362
74
Garcia-Ruiz
C
Colell
A
Mari
M
Morales
A
Fernandez-Checa
JC
Direct effect of ceramide on the mitochondrial electron transport chain leads to generation of reactive species.
J Biol Chem.
272
1997
11369
11377
75
Quillet-Mary
A
Jaffrézou
JP
Mansat
V
Bordier
C
Naval
J
Laurent
G
Implication of mitochondrial hydrogen peroxide generation in ceramide-induced apoptosis.
J Biol Chem.
272
1997
21388
21395
76
Verhaegen
S
McGowan
AJ
Brophy
AR
Fernandes
RS
Cotter
TG
Inhibition of apoptosis by antioxidants in the human HL-60 leukaemia cell line.
Biochem Pharmacol.
50
1995
1021
1029
77
Fiorani
M
Cantoni
O
Tasinato
A
Boscoboinik
D
Azzi
A
Hydrogen peroxide- and fetal bovine serum-induced DNA synthesis in vascular smooth muscle cells: positive and negative regulation by protein kinase C isoforms.
Biochim Biophys Acta.
1269
1995
98
104
78
Ohmori
S
Shirai
Y
Sakai
N
et al
Three distinct mechanisms for translocation and activation of the delta subspecies of protein kinase C.
Mol Cell Biol.
18
1998
5263
5271
79
Chen
CC
Liau
CS
Lee
YT
Tumor necrosis factor-alpha, platelet-activating factor, and hydrogen peroxide activate protein kinase C subtypes alpha and epsilon in human saphenous vein endothelial cells.
J Cardiovasc Pharmacol.
28
1996
240
244
80
Devary
Y
Rosette
C
DiDonato
JA
Karin
M
NF-κB activation by ultraviolet light not dependent on a nuclear signal.
Science.
261
1993
1442
1445
81
Kasid
U
Suy
S
Dent
P
Ray
S
Whiteside
TL
Sturgill
TW
Activation of Raf by ionizing radiation.
Nature.
382
1996
813
816
82
Suy
S
Anderson
WB
Dent
P
Chang
E
Kasid
U
Association of Grb2 with Sos and Ras with Raf-1 upon gamma irradiation of breast cancer cells.
Oncogene.
15
1997
53
61
83
Konishi
H
Tanaka
M
Takemura
Y
et al
Activation of protein kinase C by tyrosine phosphorylation in response to H2O2.
Proc Natl Acad Sci U S A.
94
1997
11233
11237
84
Aikawa
AR
Komuro
I
Yamazaki
T
et al
Oxidative stress activates extracellular signal-regulated kinases through Src and Ras in cultured cardiac myocytes of neonatal rats.
J Clin Invest.
100
1997
1813
1821
85
Min
DS
Kim
E-G
Exton
JH
Involvement of tyrosine phosphorylation and protein kinase C in the activation of phospholipase D by H2O2 in swiss 3T3 fibroblasts.
J Biol Chem.
273
1998
29986
29994
86
Russo
T
Zambrano
N
Esposito
F
et al
A p53-independent pathway for activation of WAF1/CIP1 expression following oxidative stress.
J Biol Chem.
270
1995
29386
29391
87
Guyton
KZ
Liu
Y
Gorospe
M
Xu
Q
Holbrook
NJ
Activation of mitogen-activated protein kinase by H2O2: role in cell survival following oxidant injury.
J Biol Chem.
271
1996
4138
4142
88
Baeuerle
PA
Henkel
T
Function and activation of NF-κB in the immune system.
Annu Rev Immunol.
12
1994
141
179
89
Wang
C-Y
Mayo
MW
Baldwin
AS
TNF-α and cancer therapy-induced apoptosis: potentiation by inhibition of NF-κB.
Science.
274
1996
784
787
90
Posada
J
Vichi
P
Tritton
TR
Protein kinase C in adriamycin action and resistance in mouse sarcoma 180 cells.
Cancer Res.
49
1989
6634
6639
91
Bettaieb
A
Plo
I
Mansat-De Mas
V
et al
Daunorubicin- and mitoxantrone-triggered phosphatidylcholine hydrolysis: implication in drug-induced ceramide generation.
Mol Pharmacol.
55
1999
118
125
92
Kolesnick
R
Fuks
Z
Ceramide: a signal for apoptosis or mitogenesis?
J Exp Med.
181
1995
1949
1952
93
Bjorkoy
G
Overvathn
A
Diaz-Meco
MT
Moscat
J
Johansen
T
Evidence for a bifurcation of the mitogenic signaling pathway activated by Ras and phosphatidylcholine-hydrolyzing phospholipase C.
J Biol Chem.
270
1995
21299
21306
94
Van Dijk
MCM
Hilkmann
H
van Blitterswijk
WJ
Platelet-derived growth factor activation of mitogen-activated protein kinase depends on the sequential activation of phosphatidylcholine-specific phospholipase C, protein kinase C-δ and Raf-1.
Biochem J.
325
1997
303
307
95
Xia
Z
Dickens
M
Raingeaud
J
Davis
RJ
Greenberg
ME
Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis.
Science.
270
1995
1326
1331
96
Gokhale
PC
Soldatenkov
V
Wang
FH
Rahman
A
Dritschilo
A
Kasid
U
Antisense raf oligodeoxyribonucleotide is protected by liposomal encapsulation and inhibits Raf-1 protein expression in vitro and in vivo: implication for gene therapy of radioresistant cancer.
Gene Ther.
4
1997
1289
1299
97
Pirollo
KF
Hao
Z
Rait
A
Ho
CW
Chang
EH
Evidence supporting a signal transduction pathway leading to the radiation-resistant phenotype in human tumor cells.
Biochem Biophys Res Commun.
230
1997
196
201
98
Berra
E
Diaz-Meco
MT
Dominguez
I
et al
Protein kinase C δ isoform is critical for mitogenic signal transduction.
Cell.
74
1993
555
563
99
Berra
E
Municio
MM
Sanz
L
Frutos
S
Diaz-Meco
MT
Moscat
J
Positioning atypical PKC isoforms in the UV-induced apoptotic signaling cascade.
Mol Cell Biol.
17
1997
4346
4354
100
Murray
NR
Fields
AP
Atypical protein kinase C ι protects human leukemia cells against drug-induced apoptosis.
J Biol Chem.
272
1997
27521
27524
101
Diaz-Laviada
I
Larrodera
P
Diaz-Meco
MT
et al
Evidence for a role of phosphatidylcholine-hydrolysing phospholipase C in the regulation of protein kinase C by ras and src oncogenes.
EMBO J.
9
1990
3907
3912
102
Rao
P
Kitamura
T
Miyajima
A
Mufson
RA
Human IL-3 receptor signaling: rapid induction of phosphatidylcholine hydrolysis is independent of protein kinase C but dependent on tyrosine phosphorylation in transfected NIH 3T3 cells.
J Immunol.
154
1995
1664
1674
103
Rao
P
Mufson
RA
Human interleukin-3 stimulates a phosphatidylcholine specific phospholipase C and protein kinase C translocation.
Cancer Res.
54
1994
777
781
104
Machleidt
T
Krämer
B
Adam
D
et al
Function of the p55 tumor necrosis factor receptor “death domain” mediated by phosphatidylcholine-specific phospholipase C.
J Exp Med.
184
1996
725
733
105
Franke
TF
Kaplan
DR
Cantley
LC
Toker
A
Direct regulation of the Akt proto-oncogene product by phosphatidylinositol-3,4-biphophate.
Science.
275
1997
665
668
106
Franke
TF
Kaplan
DR
Cantley
LC
PI3K: downstream AKTion blocks apoptosis.
Cell.
88
1997
435
437
107
Toker
A
Meyer
M
Reddy
KK
et al
Activation of protein kinase C family members by the novel polyphosphoinositides PtdIns-3,4-P2 and PtdIns-3,4,5-P3.
J Biol Chem.
269
1994
32358
32367
108
Nakanishi
H
Brewer
KA
Exton
JH
Activation of the ζ isozyme of protein kinase C by phosphatidylinositol 3,4,5-triphosphate.
J Biol Chem.
268
1993
13
16
109
Bae
YS
Cantley
LG
Chen
C-S
Kim
S-R
Kwon
K-S
Rhee
SG
Activation of phospholipase C-γ by phosphatidylinositol 3,4,5-triphosphate.
J Biol Chem.
273
1998
4465
4469
110
Price
BD
Youmell
MB
The phosphatidylinositol 3-kinase inhibitor wortmannin sensitizes murine fibroblasts and human tumor cells to radiation and blocks induction of p53 following DNA damage.
Cancer Res.
56
1996
246
250
111
Kulik
G
Klippel
A
Weber
MJ
Antiapoptotic signalling by the insulin-like growth factor I receptor, phosphatidylinositol 3-kinase, and Akt.
Mol Cell Biol.
17
1997
1595
1606
112
Plo
I
Bettaieb
A
Payrastre
B
et al
Phosphoinositide 3-kinase/Akt-mediated survival pathway is activated by daunorubicin in human acute myeloid leukemia cell lines.
FEBS Lett.
452
1999
150
154
113
O'Gorman
DM
Mc Kenna
SL
McGahon
AJ
Knox
KA
Cotter
TG
Sensitization of HL60 human apoptosis by inhibition of PI3-kinase survival signals.
Leukemia.
14
2000
602
611
114
Rodriguez-Viciana
P
Warne
PH
Khwaja
A
et al
Role of phosphoinositide 3-OH kinase in cell transformation and control of the actin cytoskeleton by Ras.
Cell.
89
1997
457
467
115
Varticovski
L
Daley
GQ
Jackson
P
Baltimore
D
Cantley
LC
Activation of phosphatidylinositol 3-kinase in cells expressing abl oncogene variants.
Mol Cell Biol.
11
1991
1107
1113
116
Skorski
T
Bellacosa
A
Nieborovska-Skorska
M
et al
Transformation of hematopoietic cells by BCR/ABL requires activation of a PI-3k/Akt-dependent pathway.
EMBO J.
16
1997
6151
6161
117
Skorski
T
Kanakaraj
P
Nieborovska-Skorska
M
et al
Phosphatidylinositol-3 kinase activity is regulated by BCR/ABL and is required for the growth of Philadelphia chromosome-positive cells.
Blood.
86
1995
726
736
118
Casteran
N
Rottapel
R
Beslu
N
Lecocq
E
Birnbaum
D
Dubreuil
P
Analysis of the mitogenic pathway of the FLT3 receptor and characterization in its C terminal region of a specific binding site for the PI3-kinase.
Cell Mol Biol.
40
1994
443
456
119
Del Peso
L
Gonzales-Garcia
M
Page
C
Herrera
R
Nunez
G
Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt.
Science.
278
1997
687
689
120
Kennedy
SG
Wagner
AJ
Conzen
SD
et al
The PI 3-kinase/Akt signaling pathway delivers an apoptotic signal.
Genes Dev.
11
1997
701
713
121
Cardone
MH
Roy
N
Stennicke
HR
et al
Regulation of cell death protease caspase-9 by phosphorylation.
Science.
282
1998
1318
1321
122
Zhou
H
Summers
SA
Birbaum
MJ
Pittman
RN
Inhibition of Akt kinase by cell-permeable ceramide and its implication for ceramide-induced apoptosis.
J Biol Chem.
273
1998
16568
16575
123
Piret
B
Pirette
J
Topoisimerase poisons activate the transcription factor NF-kappaB in ACH-2 and CEM cells.
Nucleic Acids Res.
24
1996
4242
4248
124
Boland
MP
Foster
SJ
O'Neill
LAJ
Daunorubicin activates NFκB and induces κB-dependent gene expression in HL-60 promyelocytic and Jurkat T lymphoma cells.
J Biol Chem.
272
1997
12952
12960
125
Das
KC
White
CW
Activation of NF-κB by antineoplastic agents: role of protein kinase C.
J Biol Chem.
272
1997
14914
14920
126
Hellin
A-C
Calmant
P
Gielin
J
Bours
V
Merville
M-P
Nuclear factor-κB-dependent regulation of p53 gene expression induced by daunomycin genotoxic drug.
Oncogene.
16
1998
1187
1195
127
Bentires-Alj
M
Hellin
A-C
Ameyar
M
Chouaib
S
Merville
M-P
Bours
V
Stable inhibition of nuclear factor kB in cancer cells does not increase sensitivity to cytotoxic drugs.
Cancer Res.
59
1999
811
815
128
Brach
MA
Kharbanda
SM
Herrmann
F
Kufe
DW
Activation of the transcription factor kB in human KG-1 myeloid leukemia cells treated with 1-β-D-Arabinofuranosylcytosine.
Mol Pharmacol.
41
1991
60
63
129
Slater
AF
Kimland
M
Jiang
SA
Orrenius
S
Constitutive nuclear NF kappa B/rel DNA-binding activity of rat thymocytes is increased by stimuli that promote apoptosis, but not inhibited by pyrrolidine dithiocarbamate.
Biochem J.
312
1995
833
838
130
Schutze
SK
Potthoff
T
Machleidt
D
Berkovic
K
Wiegmann
K
Kronke
M
TNF activates NF-κB by phosphatidylcholine-specific phospholipase C-induced “acidic” sphingomyelin breakdown.
Cell.
71
1992
765
771
131
Mansat
V
Bettaieb
A
Levade
T
Laurent
G
Jaffrézou
JP
Serine-protease inhibitors block neutral sphingomyelinase activation, ceramide generation and apoptosis triggered by daunorubicin.
FASEB J.
11
1997
695
702
132
Johns
LD
Sarr
T
Ranges
GE
Inhibition of ceramide pathway does not affect ability of TNF-α to activate nuclear factor-κB.
J Immunol.
152
1994
5877
5882
133
Betts
JC
Agranoff
AB
Nabel
GJ
Shayman
JA
Dissociation of endogenous cellular ceramide from NF-κB activation.
J Biol Chem.
269
1994
8455
8458
134
Lallena
MJ
Diaz-Meco
MT
Bren
G
Paya
CV
Moscat
J
Activation of IkappaB kinase beta by protein kinase C isoforms.
Mol Cell Biol.
19
1999
2180
2188
135
Nidai Ozes
O
Mayo
LD
Gustin
JA
Pfeffer
SR
Pfeffer
LM
Donner
DB
NF-κB activation by tumour necrosis factor requires the Akt serine-threonine kinase.
Nature.
401
1999
82
85
136
Romashkova
JA
Makarov
SS
NF-κB is a target of AKT in anti-apoptotic PDGF signalling.
Nature.
401
1999
86
89
137
Wu
M
Lee
H
Bellas
RE
et al
Inhibition of NF-κB/Rel induces apoptosis of murine B cells.
EMBO J.
15
1996
4682
4690
138
Bargou
RC
Emmerich
F
Krappmann
D
et al
Constitutive nuclear factor-κB-RelA activation is required for proliferation and survival of Hodgkin's disease tumor cells.
J Clin Invest.
100
1997
2961
2969
139
Pyatt
DW
Stillman
WS
Yang
Y
Gross
S
Zheng
J-h
Irons
RD
An essential role for NF-κB in human CD34+ bone marrow cell survival.
Blood.
93
1999
3302
3308
140
Beg
AA
Baltimore
D
An essential role for NF-κB in preventing TNF-α-induced cell death.
Science.
274
1996
782
784
141
Van Antwerp
DJ
Martin
SJ
Kafri
T
Green
DR
Verma
IM
Suppression of TNF-α-induced apoptosis by NF-κB.
Science.
274
1996
787
789
142
Brockman
JA
Scherer
DC
McKinsey
TA
et al
Coupling of a signal response domain in I kappa B alpha to multiple pathways for NF-kappa B activation.
Mol Cell Biol.
15
1995
2809
2818
143
Palombello
V
Rando
O
Goldberg
A
Maniatis
T
The ubiquitin-proteasome pathway is required for processing the NF-kappa B1 precursor protein and the activation of NF-kappa B.
Cell.
78
1994
773
785
144
Auphan
N
DiDonato
JA
Rosette
C
Helmberg
A
Karin
M
Immunosuppression by glucocorticoids: inhibition of NF-κB activity through induction of IκB synthesis.
Science.
270
1995
286
290
145
Scheinman
RI
Cogswell
PC
Lofquist
AK
Baldwin
AS
Role of transcriptional activation of IkBa in mediation of immunosuppression by glucocorticoids.
Science.
270
1995
283
286
146
Wang
CY
Cusack
JC
Jr
Liu
R
Baldwin
AS
Jr
Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-kappaB.
Nat Med.
5
1999
412
417
147
Diaz-Meco
MT
Lallena
M-J
Monjas
A
Frutos
S
Moscat
J
Inactivation of the inhibitory κB protein kinase/nuclear factor κB pathway by Par-4 expression potentiates tumor necrosis factor α-induced apoptosis.
J Biol Chem.
274
1999
19606
19612
148
Friesen
C
Herr
I
Krammer
PH
Debatin
KM
Involvement of the CD95 (APO-1/FAS) receptor/ligand system in drug-induced apoptosis in leukemia cells.
Nat Med.
2
1996
574
577
149
Müller
M
Strand
S
Hug
H
et al
Drug-induced apoptosis in hepatoma cells is mediated by the CD95 (APO-1/Fas) receptor/ligand system and involves activation of wild-type p53.
J Clin Invest.
99
1997
403
413
150
Fulda
S
Sieverts
H
Friesen
C
Herr
I
Debatin
KM
The CD95 (APO-1/Fas) system mediates drug-induced apoptosis in neuroblastoma cells.
Cancer Res.
57
1997
3823
3829
151
Herr
I
Wilhelm
D
Bohler
T
Angel
P
Debatin
KM
Activation of CD95 (APO-1/Fas) signaling by ceramide mediates cancer therapy-induced apoptosis.
EMBO J.
16
1997
6200
6208
152
Eischen
CM
Kottke
TJ
Martins
LM
et al
Comparison of apoptosis in wild-type and Fas-resistant cells: chemotherapy-induced apoptosis is not dependent on Fas/Fas ligand interactions.
Blood.
90
1997
935
943
153
Villunger
A
Egle
A
Kos
M
et al
Drug-induced apoptosis is associated with enhanced Fas (APO-1/CD95) ligand expression but occurs independently of Fas (APO-1/CD95) signaling in human T-acute lymphatic leukemia cells.
Cancer Res.
57
1997
3331
3334
154
Wesselborg
S
Engels
IH
Rossmann
E
Los
M
Schulze-Osthoff
K
Anticancer drugs induce caspase-8/FLICE activation and apoptosis in the absence of CD95 receptor/ligand interaction.
Blood.
93
1999
3053
3063
155
Micheau
O
Solary
E
Hammann
A
Dimanche-Boitrel
M-T
Fas ligand-independent, FADD-mediated activation of the Fas death pathway by anticancer drugs.
J Biol Chem.
274
1999
7987
7992
156
Rehemtulla
A
Hamilton
CA
Chinnaiyan
AM
Dixit
VM
Ultraviolet radiation-induced apoptosis is mediated by activation of CD-95 (Fas/APO-1).
J Biol Chem.
272
1997
25783
25786
157
Aragane
Y
Kulms
D
Metze
D
et al
Ultraviolet light induces apoptosis via direct activation of CD95 (Fas/APO-1) independently of its ligand CD95L.
J Cell Biol.
140
1998
171
182
158
Belka
C
Marini
P
Lepple-Wienhues
A
et al
The tyrosine kinase Lck is required for CD95-independent caspase-8 activation and apoptosis in response to ionizing radiation.
Oncogene.
18
1999
4983
4992
159
Muzio
M
Chinnaiyan
AM
Kischkel
FC
et al
FLICE, a novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95(Fas/Apo-1) death-inducing signaling complex.
Cell.
85
1996
817
827
160
Kataoka
T
Schroter
M
Hahne
M
et al
FLIP prevents apoptosis induced by death receptors but not by perforin/granzyme B, chemotherapeutic drugs, and gamma irradiation.
J Immunol.
161
1998
3936
3942
161
Los
M
Herr
I
Friesen
C
Fulda
S
Schulze-Osthoff
K
Debatin
KM
Cross-resistance of CD95- and drug-induced apoptosis as a consequence of deficient activation of caspases (ICE/Ced-3 proteases).
Blood.
90
1997
3118
3129
162
Dbaibo
GS
Perry
DK
Gamard
CJ
et al
Cytokine response modifier A (CrmA) inhibits ceramide formation in response to tumor necrosis factor (TNF)-α: CrmA and Bcl-2 target distinct components in the apoptotic pathway.
J Exp Med.
185
1997
481
490
163
Tepper
AD
de Vries
E
van Blitterswijk
WJ
Borst
J
Ordering of ceramide formation, caspase activation, and mitochondrial changes during CD95- and DNA damage-induced apoptosis.
J Clin Invest.
103
1999
971
978
164
Zeytun
A
Hassuneh
M
Nagarkatti
M
Nagarkatti
PS
Fas-Fas ligand-based interactions between tumor cells and tumor-specific cytotoxic T lymphocytes: a lethal two-way street.
Blood.
90
1997
1952
1959
165
Strand
S
Hofmann
WJ
Hug
H
et al
Lymphocyte apoptosis induced by CD95 (APO-1/Fas) ligand-expressing tumor cells—a mechanism of immune evasion?
Nat Med.
2
1996
1361
1366
166
Schmidt-Wolf
IG
Lefterova
P
Johnston
V
et al
Sensitivity of multidrug-resistant tumor cell lines to immunologic effector cells.
Cell Immunol.
169
1996
85
90
167
Frost
P
Ng
CP
Belldegrun
A
Bonavida
B
Immunosensitisation of prostate carcinoma cell lines for lymphocytes (CTL, TIL, LAK)-mediated apoptosis via the Fas-Fas-ligand pathway of cytotoxicity.
Cell Immunol.
180
1997
70
83
168
Micheau
O
Solary
E
Hammann
A
Martin
F
Dimanche-Boitrel
M-T
Sensitization of cancer cells treated with cytotoxic drugs to Fas-mediated cytotoxicity.
J Natl Cancer Inst.
89
1997
783
789
169
Classen
CF
Fulda
S
Friesen
C
Debatin
KM
Decreased sensitivity of drug-resistant cells towards T cell cytotoxicity.
Leukemia.
13
1999
410
418
170
Cai
Z
Stancou
R
Körner
M
Chouaib
S
Impairment of Fas-antigen expression in adriamycin-resistant but not TNF-resistant MCF-7 tumor cells.
Int J Cancer.
68
1996
535
546
171
Landowski
TH
Gleason-Guzman
MC
Dalton
W
Selection for drug resistance results in resistance to Fas-mediated apoptosis.
Blood.
89
1997
1854
1861
172
Barcena
A
Park
SW
Banapour
B
Muench
MO
Mechetner
E
Expression of Fas/CD95 and Bcl-2 by primitive hematopoietic progenitors freshly isolated from human fetal liver.
Blood.
88
1996
2013
2025
173
Dirks
W
Schöne
S
Uphoff
C
Quentmeier
H
Pradella
S
Drexler
HG
Expression and function of CD95 (FAS/apo-1) in leukemia-lymphoma tumour lines.
Br J Haematol.
96
1997
584
593
174
Iijima
N
Miyamura
K
Itou
T
Tanimoto
M
Sobue
R
Saito
H
Functional expression of Fas (CD95) in acute myeloid leukemia cells in the context of CD34 and CD38 expression: possible correlation with sensitivity to chemotherapy.
Blood.
90
1997
4901
4909
175
Wen
J
Ramadevi
N
Nguyen
D
Perkins
C
Worthington
E
Bhalla
K
Antileukemic drugs increase death receptor 5 levels and enhance Apo-2L-induced apoptosis of human acute leukemia cells.
Blood.
96
2000
3900
3906
176
Sheikh
MS
Burns
TF
Huang
Y
et al
p53-dependent and -independent regulation of the death receptor killer/DR5 gene expression in response to genotoxic stress and tumor necrosis factor alpha.
Cancer Res.
58
1998
1593
1598
177
Tishler
RB
Calderwood
SK
Coleman
N
Price
BD
Increase in sequence specific DNA binding by p53 following treatment with chemotherapeutic and DNA damaging agents.
Cancer Res.
53
1993
2212
2216
178
Lowe
SW
Bodis
S
McClatchey
A
et al
p53 status and the efficacy of cancer therapy in vivo.
Science.
266
1994
807
810
179
Lowe
SW
Ruley
HE
Jacks
T
Housman
DE
p53-dependent apoptosis modulates the cytotoxicity of anticancer agents.
Cell.
74
1993
957
967
180
Brown
JM
Wouters
BG
Apoptosis, p53, and tumor cell sensitivity to anticancer agents.
Cancer Res.
59
1999
1391
1399
181
Wattel
E
Preudhomme
C
Hecquet
B
et al
p53 mutations are associated with resistance to chemotherapy and short survival in hematologic malignancies.
Blood.
84
1994
3148
3157
182
Navaratnam
S
Williams
GJ
Rubinger
M
et al
Expression of p53 predicts treatment failure in aggressive non-Hodgkin's lymphomas.
Leuk Lymphoma.
29
1998
139
144
183
Han
J-W
Dionne
CA
Kedersha
NL
Goldmacher
VS
p53 status affects the rate of the onset but not the overall extent of doxorubicin-induced cell death in Rat-1 fibroblasts constitutively expressing c-Myc.
Cancer Res.
57
1997
176
182
184
El-Deiry
WS
Tokino
T
Velculescu
VE
et al
WAF1, a potential mediator of p53 tumor suppression.
Cell.
75
1993
817
825
185
Vikhanskaya
F
D'Incalci
M
Broggini
M
Decreased cytotoxic effects of doxorubicin in a human ovarian cancer-cell line expressing wild-type p53 and WAF1/CIP1 genes.
Int J Cancer.
61
1995
397
401
186
Gartenhaus
RB
Wang
P
Hoffmann
P
Induction of the WAF1/CIP1 protein and apoptosis in human T-cell leukemia virus type I-transformed lymphocytes after treatment with adriamycin by using a p53-independent pathway.
Proc Natl Acad Sci U S A.
93
1996
265
268
187
Zhang
W
Kornblau
SM
Kobayashi
T
Gambel
A
Claxton
D
Deisseroth
AB
High levels of constitutive WAF1/Cip1 protein are associated with chemoresistance in acute myelogenous leukemia.
Clin Cancer Res.
1
1995
1051
1057
188
Ling
Y-H
El-Naggar
AK
Priebe
W
Perez-Soler
R
Cell cycle-dependent cytotoxicity, G2/M phase arrest, and disruption of p34cdc2/cyclin B1 activity induced by doxorubicin in synchronized P388 cells.
Mol Pharmacol.
49
1996
832
841
189
Kharbanda
S
Yuan
ZM
Rubin
E
Weichselbaum
R
Kufe
D
Activation of Src-like p56/p53lyn tyrosine kinase by ionizing radiation.
J Biol Chem.
269
1994
20739
20743
190
Kharbanda
S
Yuan
ZM
Taneja
N
Weichselbaum
R
Kufe
D
p56/p53lyn tyrosine kinase activation in mammalian cells treated with mitomycin C.
Oncogene.
9
1994
3005
3011
191
Giancotti
FG
Ruoslahti
E
Integrin signaling.
Science.
285
1999
1028
1032
192
Sethi
T
Rintoul
RC
Moore
SM
et al
Extracellular matrix proteins protect small cell lung cancer cells against apoptosis: a mechanism for small cell lung cancer growth and drug resistance in vivo.
Nat Med.
5
1999
662
668
193
Damiano
JS
Cress
AE
Hazlehurst
LA
Shtil
AA
Dalton
WS
Cell adhesion mediated drug resistance (CAM-DR): role of integrins and resistance to apoptosis in human myeloma cell lines.
Blood.
93
1999
1658
1667
194
Verfaillie
CM
Adhesion receptors as regulators of the hematopoietic process.
Blood.
92
1998
2609
2612
195
Chen
JJW
Wu
R
Yang
PC
et al
Profiling expression patterns and isolating differentially expressed genes by cDNA microarray system with colorimetry detection.
Genomics.
51
1998
313
324
196
Kudoh
K
Ramana
M
Ravata
R
et al
Monitoring the expression profiles of doxorubicin-induced and doxorubicin-resistant cancer cells by cDNA microarray.
Cancer Res.
60
2000
4161
4166
197
Hannun
YA
Apoptosis and the dilemma of cancer chemotherapy.
Blood.
89
1997
1845
1853

Author notes

Guy Laurent, INSERM E9910, Institut Claudius Régaud, 20 rue du Pont St Pierre, 31052 Toulouse, France; e-mail:laurent@icr.fnclcc.fr.

Sign in via your Institution