Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Leukemia ; 30(3): 728-39, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26449661

ABSTRACT

The t(8;21) rearrangement, which creates the AML1-ETO fusion protein, represents the most common chromosomal translocation in acute myeloid leukemia (AML). Clinical data suggest that CBL mutations are a frequent event in t(8;21) AML, but the role of CBL in AML1-ETO-induced leukemia has not been investigated. In this study, we demonstrate that CBL mutations collaborate with AML1-ETO to expand human CD34+ cells both in vitro and in a xenograft model. CBL depletion by shRNA also promotes the growth of AML1-ETO cells, demonstrating the inhibitory function of endogenous CBL in t(8;21) AML. Mechanistically, loss of CBL function confers hyper-responsiveness to thrombopoietin and enhances STAT5/AKT/ERK/Src signaling in AML1-ETO cells. Interestingly, we found the protein tyrosine phosphatase UBASH3B/Sts-1, which is known to inhibit CBL function, is upregulated by AML1-ETO through transcriptional and miR-9-mediated regulation. UBASH3B/Sts-1 depletion induces an aberrant pattern of CBL phosphorylation and impairs proliferation in AML1-ETO cells. The growth inhibition caused by UBASH3B/Sts-1 depletion can be rescued by ectopic expression of CBL mutants, suggesting that UBASH3B/Sts-1 supports the growth of AML1-ETO cells partly through modulation of CBL function. Our study reveals a role of CBL in restricting myeloid proliferation of human AML1-ETO-induced leukemia, and identifies UBASH3B/Sts-1 as a potential target for pharmaceutical intervention.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , Preleukemia/genetics , Protein Tyrosine Phosphatases/genetics , Proto-Oncogene Proteins c-cbl/genetics , Animals , Cell Proliferation , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fetal Blood/cytology , Fetal Blood/drug effects , Fetal Blood/metabolism , Heterografts , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, SCID , MicroRNAs/genetics , MicroRNAs/metabolism , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Oncogene Proteins, Fusion/metabolism , Preleukemia/metabolism , Preleukemia/pathology , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/antagonists & inhibitors , Proto-Oncogene Proteins c-cbl/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RUNX1 Translocation Partner 1 Protein , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Thrombopoietin/pharmacology , Transgenes , Translocation, Genetic , src-Family Kinases/genetics , src-Family Kinases/metabolism
2.
Leukemia ; 29(12): 2382-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26172401

ABSTRACT

In acute myeloid leukemia (AML), about 25-30% of patients harbor a constitutively active receptor tyrosine kinase (RTK) FLT3 encoded by a FLT3 allele harboring internal tandem duplication (FLT3-ITD) mutation. The presence of FLT3-ITD correlates with poor prognosis in AML and it makes FLT3 an attractive therapeutic target in AML. Unfortunately, to date small-molecule inhibitors of FLT3 have resulted in only partial and transient clinical responses with residual leukemic blasts resistant to FLT3 inhibitors detected in blood or bone marrow. In this study, we investigated whether the RTK Axl is responsible for resistance of FLT3-ITD(+) AML cells to PKC412 and AC220, FLT3 inhibitors currently under clinical trials for FLT3-ITD(+) AML patients. Upon treatment with PKC412 or AC220, phosphorylation of Axl was significantly enhanced in the FLT3-ITD(+) MV4-11 AML cell line and in primary blasts from a FLT3-ITD(+) AML patient. Consistently, a PKC412-resistant AML cell line and PKC412-resistant primary blasts from FLT3-ITD(+) AML patients had significantly higher levels of constitutively phosphorylated Axl and total Axl when compared with a PKC412-sensitive AML cell line and PKC412-sensitive primary blasts from FLT3-ITD(+) AML patients. We also found that resistance of AML cells against the FLT3 inhibitor PKC412 and AC220 was substantially diminished by the inhibition of Axl via a small-molecule inhibitor TP-0903, a soluble receptor Axl fusion protein Axl-Fc or knockdown of Axl gene expression by shRNA. Collectively, our study suggests that Axl is required for resistance of FLT3-ITD(+) AML cells against the FLT3 inhibitor PKC412 and AC220, and that inhibition of Axl activation may overcome resistance to FLT3-targeted therapy in FLT3-ITD(+) AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Phosphorylation , Axl Receptor Tyrosine Kinase
3.
Leukemia ; 29(3): 567-75, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25204569

ABSTRACT

DNMT3B encodes a DNA methyltransferase implicated in aberrant epigenetic changes contributing to leukemogenesis. We tested whether DNMT3B expression, measured by NanoString nCounter assay, associates with outcome, gene and microRNA expression and DNA methylation profiles in 210 older (⩾60 years) adults with primary, cytogenetically normal acute myeloid leukemia (CN-AML). Patients were dichotomized into high versus low expressers using median cut. Outcomes were assessed in the context of known CN-AML prognosticators. Gene and microRNA expression, and DNA methylation profiles were analyzed using microarrays and MethylCap-sequencing, respectively. High DNMT3B expressers had fewer complete remissions (CR; P=0.002) and shorter disease-free (DFS; P=0.02) and overall (OS; P<0.001) survival. In multivariable analyses, high DNMT3B expression remained an independent predictor of lower CR rates (P=0.04) and shorter DFS (P=0.04) and OS (P=0.001). High DNMT3B expression associated with a gene expression profile comprising 363 genes involved in differentiation, proliferation and survival pathways, but with only four differentially expressed microRNAs (miR-133b, miR-148a, miR-122, miR-409-3p) and no differential DNA methylation regions. We conclude that high DNMT3B expression independently associates with adverse outcome in older CN-AML patients. Gene expression analyses suggest that DNMT3B is involved in the modulation of several genes, although the regulatory mechanisms remain to be investigated to devise therapeutic approaches specific for these patients.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Age Factors , Aged , Aged, 80 and over , Cytarabine/therapeutic use , DNA Methylation , Daunorubicin/therapeutic use , Female , Gene Expression Profiling , Humans , Induction Chemotherapy , Karyotyping , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Microarray Analysis , Middle Aged , Prognosis , Survival Analysis , DNA Methyltransferase 3B
4.
Leukemia ; 27(10): 2023-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23765227

ABSTRACT

Acute myeloid leukemia (AML) is hypothesized to be sustained by self-renewing leukemia stem cells (LSCs). Recently, gene expression signatures (GES) from functionally defined AML LSC populations were reported, and expression of a 'core enriched' (CE) GES, representing 44 genes activated in LCSs, conferred shorter survival in cytogenetically normal (CN) AML. The prognostic impact of the CE GES in the context of other molecular markers, including gene mutations and microRNA (miR) expression alterations, is unknown and its clinical utility is unclear. We studied associations of the CE GES with known molecular prognosticators, miR expression profiles, and outcomes in 364 well-characterized CN-AML patients. A high CE score (CE(high)) associated with FLT3-internal tandem duplication, WT1 and RUNX1 mutations, wild-type CEBPA and TET2, and high ERG, BAALC and miR-155 expression. CE(high) patients had a lower complete remission (CR) rate (P=0.003) and shorter disease-free (DFS, P<0.001) and overall survival (OS, P<0.001) than CE(low) patients. These associations persisted in multivariable analyses adjusting for other prognosticators (CR, P=0.02; DFS, P<0.001; and OS, P<0.001). CE(high) status was accompanied by a characteristic miR expression signature. Fifteen miRs were upregulated in both younger and older CE(high) patients, including miRs relevant for stem cell function. Our results support the clinical relevance of LSCs and improve risk stratification in AML.


Subject(s)
Biomarkers, Tumor/genetics , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Stem Cells/metabolism , Transcriptome , Adolescent , Adult , Aged , Aged, 80 and over , Cytogenetic Analysis , Female , Humans , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Prognosis , Remission Induction , Stem Cells/pathology , Survival Rate , Young Adult
7.
Leukemia ; 17(2): 350-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12592335

ABSTRACT

In t(8;21) acute myeloid leukemia (AML), the AML1/ETO fusion protein promotes leukemogenesis by recruiting histone deacetylase (HDAC) and silencing AML1target genes important for hematopoietic differentiation. We hypothesized that depsipeptide (FR901228), a novel HDAC inhibitor evaluated in ongoing clinical trials, restores gene transcription and cell differentiation in AML1/ETO-positive cells. A dose-dependent increase in H3 and H4 histone acetylation was noted in depsipeptide-treated AML1/ETO-positive Kasumi-1 cells and blasts from a patient with t(8;21) AML. Consistent with this biological effect, we also showed a dose-dependent increase in cytotoxicity, expression of IL-3, here used as read-out for silenced AML1-target genes, upregulation of CD11b with other morphologic changes suggestive of partial cell differentiation in Kasumi-1 cells. Some of these biologic effects were also attained in other myeloid leukemia cell lines, suggesting that depsipeptide has differentiation and cytotoxic activity in AML cells, regardless of the underlying genomic abnormality. Notably, the activity of depsipeptide was enhanced by 5-aza-2'-deoxycytidine, a DNA methyltransferase inhibitor (DNMT). These two agents in combination resulted in enhanced histone acetylation, IL-3 expression, and cytotoxicity, suggesting HDAC and DNMT activities as a potential dual target in future therapeutic strategies for AML1/ETO and other molecular subgroups of AML.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibiotics, Antineoplastic/pharmacology , DNA Modification Methylases/antagonists & inhibitors , DNA-Binding Proteins/genetics , Depsipeptides , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , Interleukin-3/genetics , Neoplasm Proteins/genetics , Peptides, Cyclic , Transcription Factors/genetics , Transcription, Genetic/drug effects , Acetylation , Analysis of Variance , Cell Differentiation , Cell Survival , Core Binding Factor Alpha 2 Subunit , DNA Methylation , DNA Primers , Histones/drug effects , Humans , Proto-Oncogene Proteins/genetics , RUNX1 Translocation Partner 1 Protein , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
8.
Cancer Res ; 61(19): 7233-9, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585760

ABSTRACT

The FLT3 gene is mutated by an internal tandem duplication (ITD) in 20-25% of adults with acute myeloid leukemia (AML). We studied 82 adults <60 years of age with primary AML and normal cytogenetics, who received uniform high-dose therapy and found FLT3 ITD in 23 (28%) patients. When the 23 FLT3 ITD+ cases were compared with the 59 cases with wild-type (WT) FLT3, disease-free survival (DFS) was inferior (P = 0.03), yet overall survival (OS) was not different (P = 0.14). However, 8 (35%) of 23 FLT3 ITD/+ cases also lacked a FLT3 WT allele (FLT3(ITD-R)) as determined by PCR and loss of heterozygosity. Thus, three genotypic groups were identified: normal FLT3(WT/WT), heterozygous FLT3(ITD/WT), and hemizygous FLT3(ITD/-). DFS and OS were significantly inferior for patients with FLT3(ITD/-) (P = 0.0017 and P = 0.0014, respectively). Although DFS and OS for FLT3(WT/WT) and FLT3(ITD/WT) groups did not differ (P = 0.32 and P = 0.98, respectively), OS of the FLT3(ITD/-) group was worse than the FLT3(WT/WT) (P = 0.0005) and FLT3(ITD/WT) (P = 0.008) groups. We propose a model in which FLT3(ITD/-) represents a dominant positive, gain-of-function mutation providing AML cells with a greater growth advantage compared with cells having the FLT3(WT/WT) or FLT3(ITD/WT) genotypes. In conclusion, we have identified the FLT3(ITD/-) genotype as an adverse prognostic factor in de novo AML with normal cytogenetics. A poor prognosis of the relatively young FLT3(ITD/-) adults (median age, 37 years), despite treatment with current dose-intensive regimens, suggests that new treatment modalities, such as therapy with a FLT3 tyrosine kinase inhibitor, are clearly needed for this group of patients.


Subject(s)
Gene Duplication , Leukemia, Myeloid/genetics , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Acute Disease , Adult , Alleles , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Disease-Free Survival , Female , Humans , Karyotyping , Leukemia, Myeloid/drug therapy , Male , Middle Aged , Mutation , Tandem Repeat Sequences , Treatment Outcome , fms-Like Tyrosine Kinase 3
9.
Cancer Res ; 61(1): 59-63, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11196198

ABSTRACT

A partial nontandem duplication (PNTD) of mixed lineage leukemia (MLL) gene is described in B-cell acute lymphoid leukemia without structural cytogenetic abnormalities at 11q23 and 9p22. A duplicated portion of MLL is interrupted by the insertion of a region of 9p22 that includes the 3'-end of the AF9 gene. The PNTD encodes: (a) a PNTD transcript; (b) a partial tandem duplication of MLL; and (c) a chimeric transcript fusing MLL to the 3'-end of AF9, mimicking the t(9;11)(p22;q23) and expressed 1024-fold higher than the other two. The MLL PNTD, therefore, contributes toward leukemogenesis through simultaneous production of fusion transcripts that are otherwise encoded by three distinct genetic defects.


Subject(s)
Burkitt Lymphoma/genetics , DNA-Binding Proteins/genetics , Gene Rearrangement , Proto-Oncogenes , RNA, Messenger/genetics , Transcription Factors , Alternative Splicing/genetics , Blotting, Southern , Chromosome Breakage , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 9/genetics , Exons , Histone-Lysine N-Methyltransferase , Humans , Myeloid-Lymphoid Leukemia Protein , Nuclear Proteins/genetics , Recombinant Fusion Proteins/genetics , Repetitive Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic , Tumor Cells, Cultured
10.
Mol Cell Biol ; 20(16): 5818-27, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10913165

ABSTRACT

The global increase in transcription of cytoprotective genes induced in response to oxidative challenge has been termed the antioxidant response. Ferritin serves as the major iron-binding protein in nonhematopoietic tissues, limiting the catalytic availability of iron for participation in oxygen radical generation. Here we demonstrate that ferritin is a participant in the antioxidant response through a genetically defined electrophile response element (EpRE). The EpRE of ferritin H identified in this report exhibits sequence similarity to EpRE motifs found in antioxidant response genes such as those encoding NAD(P)H:quinone reductase, glutathione S-transferase, and heme oxygenase. However, the EpRE of ferritin H is unusual in structure, comprising two bidirectional motifs arranged in opposing directions on complementary DNA strands. In addition to EpRE-mediated transcriptional activation, we demonstrate that ferritin is subject to time-dependent translational control through regulation of iron-regulatory proteins (IRP). Although IRP-1 is initially activated to its RNA binding (ferritin-repressing) state by oxidants, it rapidly returns to its basal state. This permits the translation of newly synthesized ferritin transcripts and ultimately leads to increased levels of ferritin protein synthesis following oxidant exposure. Taken together, these results clarify the complex transcriptional and translational regulatory mechanisms that contribute to ferritin regulation in response to prooxidant stress and establish a role for ferritin in the antioxidant response.


Subject(s)
Ferritins/genetics , Oxidative Stress/genetics , Protein Biosynthesis , Transcription, Genetic , Animals , Base Sequence , Cell Line , Ferritins/metabolism , Mice , Molecular Sequence Data
11.
J Immunol ; 162(8): 4511-20, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10201989

ABSTRACT

NK cells constitutively express monocyte-derived cytokine (monokine) receptors and secrete cytokines and chemokines following monokine stimulation, and are therefore a critical component of the innate immune response to infection. Here we compared the effects of three monokines (IL-18, IL-15, and IL-12) on human NK cell cytokine and chemokine production. IL-18, IL-15, or IL-12 alone did not stimulate significant cytokine or chemokine production in resting NK cells. The combination of IL-18 and IL-12 induced extremely high amounts of IFN-gamma protein (225 +/- 52 ng/ml) and a 1393 +/- 643-fold increase in IFN-gamma gene expression over those in resting NK cells. IL-15 and IL-12 induced less IFN-gamma protein (24 +/- 10 ng/ml; p < 0.007) and only a 45 +/- 19-fold increase in IFN-gamma gene expression over those in resting NK cells. The CD56bright NK cell subset produced significantly more IFN-gamma following IL-18 and IL-12 compared with CD56dim NK cells (p < 0.008). However, the combination of IL-15 and IL-12 was significantly more potent than that of IL-18 and IL-12 for NK cell production of IL-10, macrophage inflammatory protein-1alpha, macrophage inflammatory protein-1beta, and TNF-alpha at the protein and transcript levels. Granulocyte-macrophage CSF was optimally induced by IL-15 and IL-18. Resting CD56+ NK cells expressed IL-18R transcript that was up-regulated by IL-12 or IL-15. Our results show that distinct cytokine and chemokine patterns are induced in NK cells in response to different costimulatory signals from these three monokines. This suggests that NK cell cytokine production may be governed in part by the monokine milieu induced during the early proinflammatory response to infection and by the subset of NK cells present at the site of inflammation.


Subject(s)
Chemokines/genetics , Cytokines/genetics , Cytokines/pharmacology , Gene Expression Regulation/immunology , Immunity, Cellular , Killer Cells, Natural/metabolism , Lymphocyte Activation/genetics , CD56 Antigen/blood , Chemokine CCL4 , Chemokines/biosynthesis , Cytokines/biosynthesis , Dose-Response Relationship, Immunologic , Drug Combinations , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Immunophenotyping , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interleukin-10/biosynthesis , Interleukin-12/pharmacology , Interleukin-15/pharmacology , Interleukin-18/metabolism , Interleukin-18/pharmacology , Interleukin-18 Receptor alpha Subunit , Interphase/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Macrophage Inflammatory Proteins/biosynthesis , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-18 , Recombinant Proteins/pharmacology , Transcription, Genetic/immunology , Tumor Necrosis Factor-alpha/biosynthesis
12.
Blood ; 92(4): 1384-9, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9694727

ABSTRACT

We have synthesized a novel six-coordinate metal chelator from the triamine cis-1,3,5-triaminocyclohexane by the addition of a 2-pyridylmethyl pendant arm on each nitrogen, which we term tachpyr. The experiments described here were designed to explore whether this compound exhibits potential antitumor activity. When added to MBT2 or T24 cultured bladder cancer cells, tachpyr was profoundly cytotoxic, with an IC50 of approximately 4.6 micromol/L compared with 70 micromol/L for desferioxamine. To explore the mode of action of tachpyr, several metal complexes were prepared, including Fe(II), Ca(II), Mn(II), Mg(II), Cu(II), and Zn(II) tachpyr complexes. Of these, the Zn(II), Cu(II), and Fe(II) complexes were without toxic effect, whereas the Ca(II), Mn(II), and Mg(II) complexes remained cytotoxic. To further probe the role of Zn(II) and Cu(II) chelation in the cytotoxicity of tachpyr, sterically hindered tachpyr derivatives were prepared through N-alkylation of tachpyr. These derivatives were unable to strongly bind Fe(III) or Fe(II) but were able to bind Zn(II) and Cu(II). When added to cells, these sterically hindered tachpyr derivatives were nontoxic, consistent with a role of iron depletion in the cytotoxic mechanism of tachpyr. Further, the addition of tachpyr to proliferating cultures resulted in an early and selective inhibition of ferritin synthesis, an iron storage protein whose translation is critically dependent on intracellular iron pools. Taken together, these experiments suggest that tachpyr is a cytotoxic metal chelator that targets intracellular iron, and that the use of tachpyr in cancer therapy deserves further exploration.


Subject(s)
Antineoplastic Agents/pharmacology , Chelating Agents/pharmacology , Cyclohexylamines/pharmacology , Iron , Pyridines/pharmacology , Alkylation , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cations, Divalent , Cell Division , Cells, Cultured , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Cyclohexylamines/chemical synthesis , Cyclohexylamines/chemistry , Deferoxamine/pharmacology , Ferritins/biosynthesis , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Iron/metabolism , Molecular Conformation , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Urinary Bladder Neoplasms/pathology
13.
J Biol Chem ; 272(38): 23481-4, 1997 Sep 19.
Article in English | MEDLINE | ID: mdl-9295281

ABSTRACT

1-beta-D-Arabinofuranosylcytosine (ara-C) stimulates the formation of both diglyceride and ceramide in the acute myelogenous leukemia cell line HL-60 (Strum, J. C., Small, G. W., Pauig, S. B., and Daniel, L. W. (1994) J. Biol. Chem 269, 15493-15497). ara-C also causes apoptosis in HL-60 cells which can be mimicked by exogenous ceramide. However, the signaling role for ara-C-induced diacylglycerol (DAG) is not defined. We found that Bcl-2 levels were increased by treatment of HL-60 cells with exogenous DAG or 12-O-tetradecanoylphorbol-13-acetate (TPA). In contrast, exogenous ceramide treatment caused a decrease in cellular Bcl-2 levels. Thus, ara-C stimulates the synthesis of two second messengers with opposing effects on Bcl-2. Since the effects of ara-C-induced DAG could be due to protein kinase C (PKC) activation, we determined the effects of ara-C on PKC isozymes. ara-C caused an increase in membrane-bound PKCbetaII (but not PKCalpha or PKCdelta). ara-C or TPA-induced translocation of PKCbetaII was inhibited by 1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine (ET-18-OCH3), and ara-C-induced apoptosis was stimulated by pretreatment of the cells with ET-18-OCH3. ET-18-OCH3 also inhibited stimulation of Bcl-2 by TPA and enhanced the decrease in Bcl-2 observed in ara-C-treated cells. These data indicate that ara-C-induced apoptosis is limited by ara-C-stimulated PKCbetaII through effects on Bcl-2. To further determine the role of PKC, we used antisense oligonucleotides directed toward PKCbetaII. The antisense, but not the sense, oligonucleotide inhibited PKCbetaII activation and enhanced ara-C-induced apoptosis. These data demonstrate that the stimulation of apoptosis by ara-C is self-limiting and can be enhanced by inhibition of PKC.


Subject(s)
Apoptosis , Cytarabine/pharmacology , Down-Regulation , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Biological Transport , DNA Fragmentation , Enzyme Activation , HL-60 Cells , Humans , Oligonucleotides, Antisense/pharmacology , Phospholipid Ethers/pharmacology , Protein Kinase C beta , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...