Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Cell Mol Biol (Noisy-le-grand) ; 64(6): 23-30, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29808796

ABSTRACT

Chronic myeloid leukemia (CML) is a hematopoietic malignancy characterized by the t(9; 22) and the related oncogene, BCR-ABL. Tyrosine kinase activity of fusion protein BCR-ABL is the main cause of CML. Even if imatinib is used as a tyrosine kinase inhibitor (TKI) for CML therapy, drug resistance may occur in patients and the clinical failure of imatinib treatment in resistant patients had resulted with the use of another alternative TKIs. BCR-ABL dependent and independent molecular mechanisms have crucial roles in drug resistance. To reveal the underlying molecular mechanisms which play significant roles in imatinib resistance in CML, we established K562 imatinib-resistant cell line (K562r5) which was continuously exposed to (5µM) imatinib to investigate molecular mechanisms which play significant roles in drug resistance. First of all, we analyzed T315I, M351T, F315L and F359C/L/V mutations with DNA sequencing as a BCR-ABL dependent mechanism in our cell lines. Moreover, we investigated BCR-ABL independent mechanisms such as apoptosis, autophagy, drug transport and DNA repair which affect drug resistance in these cell lines. In vitro cell viability was determined by MTT assay. DNA sequencing analysis was performed to detect BCR-ABL mutations. The apoptotic effect of imatinib on CML cell lines was tested by flow cytometric Annexin V-PE staining and caspase activation assays. Apoptotic, autophagic, drug transporter and DNA repair genes expression levels were determined by RT-PCR. The conventional cytogenetic analysis was performed on K562s and K562r cells. Our results indicate that inhibition of apoptosis, induction of autophagy, overexpression of efflux gene MDR1 and down-regulation of influx gene OCT1 play crucial roles in the progression of imatinib resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Imatinib Mesylate/pharmacology , K562 Cells/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Biological Transport/drug effects , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Caspases/metabolism , DNA Mutational Analysis , DNA, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Activation/drug effects , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , K562 Cells/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mutation, Missense , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Point Mutation
2.
Br J Haematol ; 160(2): 177-87, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23157224

ABSTRACT

Aberrant JAK2 signalling plays an important role in the aetiology of myeloproliferative neoplasms (MPNs). JAK2 inhibitors, however, do not readily eliminate neoplastic MPN cells and thus do not induce patient remission. Further understanding JAK2 signalling in MPNs may uncover novel avenues for therapeutic intervention. Recent work has suggested a potential role for cellular cholesterol in the activation of JAK2 by the erythropoietin receptor and in the development of an MPN-like disorder in mice. Our study demonstrates for the first time that the MPN-associated JAK2-V617F kinase localizes to lipid rafts and that JAK2-V617F-dependent signalling is inhibited by lipid raft disrupting agents, which target membrane cholesterol, a critical component of rafts. We also show for the first time that statins, 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors, widely used to treat hypercholesterolaemia, induce apoptosis and inhibit JAK2-V617F-dependent cell growth. These cells are more sensitive to statin treatment than non-JAK2-V617F-dependent cells. Importantly, statin treatment inhibited erythropoietin-independent erythroid colony formation of primary cells from MPN patients, but had no effect on erythroid colony formation from healthy individuals. Our study is the first to demonstrate that JAK2-V617F signalling is dependent on lipid rafts and that statins may be effective in a potential therapeutic approach for MPNs.


Subject(s)
Janus Kinase 2/physiology , Membrane Microdomains/physiology , Mutation, Missense , Myeloproliferative Disorders/enzymology , Point Mutation , Signal Transduction/drug effects , Simvastatin/pharmacology , beta-Cyclodextrins/pharmacology , Apoptosis/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cholesterol/analysis , Cholesterol/physiology , Colony-Forming Units Assay , Drug Evaluation, Preclinical , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/enzymology , Humans , Janus Kinase 2/genetics , K562 Cells/drug effects , K562 Cells/enzymology , Leukemia, Erythroblastic, Acute/enzymology , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Megakaryoblastic, Acute/enzymology , Leukemia, Megakaryoblastic, Acute/pathology , Megakaryocyte Progenitor Cells/drug effects , Megakaryocyte Progenitor Cells/enzymology , Membrane Lipids/physiology , Membrane Microdomains/drug effects , Myeloproliferative Disorders/blood , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , STAT5 Transcription Factor/metabolism
3.
ChemMedChem ; 6(11): 2009-18, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-21990039

ABSTRACT

N-[2-Methyl-5-(triazol-1-yl)phenyl]pyrimidin-2-amine derivatives were synthesized and evaluated in vitro for their potential use as inhibitors of Bcr-Abl. The design is based on the bioisosterism between the 1,2,3-triazole ring and the amide group. The synthesis involves a copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC) as the key step, with the exclusive production of anti-(1,4)-triazole derivatives. One of the compounds obtained shows general activity similar to that of imatinib; in particular, it was observed to be more effective in decreasing the fundamental function of cdc25A phosphatases in the K-562 cell line.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Apoptosis/drug effects , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , K562 Cells/enzymology , Models, Molecular , Mutation , Phosphorylation/drug effects , Piperazines/chemistry , Piperazines/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , STAT5 Transcription Factor/metabolism , Structure-Activity Relationship , Triazoles/chemistry , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics , cdc25 Phosphatases/antagonists & inhibitors , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism , src-Family Kinases/metabolism
4.
Leukemia ; 25(7): 1159-67, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21483442

ABSTRACT

The cytotoxic farnesyl transferase inhibitor BMS-214662 has been shown to potently induce mitochondrial apoptosis in primitive CD34+ chronic myeloid leukaemia (CML) stem/progenitor cells. Here, to enhance the BMS-214662 apoptotic effect, we further targeted the extracellular signal-regulated kinase (ERK) pathway, downstream of BCR-ABL, by treating CD34+ CML stem/progenitor cells with a highly selective adenosine triphosphate (ATP) non-competitive MEK inhibitor, PD184352. PD184352 increased the apoptotic effect of BMS-214662 in a CML blast crisis cell line, K562, and in primary chronic phase CD34+ CML cells. Compared with BMS-214662, after combination treatment we observed inhibition of ERK phosphorylation, increased Annexin-V levels, caspase-3, -8 and -9 activation and potentiated mitochondrial damage, associated with decreased levels of anti-apoptotic BCL-2 family protein MCL-1. Inhibition of K-RAS function by a dominant-negative mutant resulted in CML cell death and this process was further enhanced by the addition of BMS-214662 and PD184352. Together, these findings suggest that the addition of a MEK inhibitor improves the ability of BMS-214662 to selectively target CML stem/progenitor cells, notoriously insensitive to tyrosine kinase inhibitor treatment and presumed to be responsible for the persistence and relapse of the disease.


Subject(s)
Apoptosis/drug effects , Benzamides/pharmacology , Benzodiazepines/pharmacology , Blast Crisis/pathology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Chronic-Phase/pathology , MAP Kinase Kinase Kinases/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Antigens, CD34/analysis , Blast Crisis/enzymology , Drug Screening Assays, Antitumor , Drug Synergism , Genes, Dominant , Genes, ras , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/enzymology , Humans , K562 Cells/drug effects , K562 Cells/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myeloid, Chronic-Phase/enzymology , MAP Kinase Kinase 1/genetics , Neoplastic Stem Cells/enzymology , Oncogene Protein p21(ras)/genetics , Recombinant Fusion Proteins/genetics , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology
5.
Hematology ; 15(1): 33-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20132660

ABSTRACT

Chronic myeloid leukemia (CML) is a hematological malignancy characterized by high levels of immature white blood cells. CML is caused by the translocation between chromosomes 9 and 22 (which results in the formation of the Philadelphia chromosome) creating BCR-ABL fusion protein. Imatinib and nilotinib are chemotherapeutic drugs which specifically bind to the BCR-ABL and inhibit cancer cells. Nilotinib is more effective in this respect than imatinib. We have shown that nilotinib induces apoptosis in imatinib-resistant K562 CML cells which have the wild-type BCR-ABL fusion gene almost to the same extent as it does in the parental sensitive cells by the increase in caspase-3 enzyme activity and the decrease in mitochondrial membrane potential. This effect of nilotinib, even in low concentrations, may indicate the efficacy of the usage of nilotinib in imatinib-resistant CML with less risk of undesired cytotoxic effects in the remaining cells of the body.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , K562 Cells/drug effects , Neoplasm Proteins/antagonists & inhibitors , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Base Sequence , Benzamides , Caspase 3/metabolism , Cell Division/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , K562 Cells/cytology , K562 Cells/enzymology , Membrane Potential, Mitochondrial/drug effects , Molecular Sequence Data , Neoplasm Proteins/genetics , Sequence Analysis, DNA
6.
Leukemia ; 23(8): 1500-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19340007

ABSTRACT

Once cleaved by caspases, the Lyn tyrosine kinase (LynDeltaN) is relocalized from the plasma membrane to the cytoplasm of apoptotic cells, but the function of such a cleavage is incompletely understood. We evaluated the effect of LynDeltaN overexpression on imatinib sensitivity of the chronic myelogenous leukemia (CML) cell line K562. Therefore, we generated stable cells that express plasmids encoding LynDeltaN or its catalytically inactive counterpart LynDeltaNKD. We established that Lyn is cleaved in imatinib-treated parental K562 cells in a caspase-dependent manner. Lyn cleavage also occurred following BCR-ABL silencing by specific short hairpin RNA (sh-RNA). Imatinib-induced apoptosis was abrogated in LynDeltaN-overexpressing cells, but not in cells overexpressing its inactive counterpart. Conversely, the overexpression of LynDeltaN failed to affect the differentiation of K562 cells. Importantly, the protective effect of LynDeltaN was suppressed by two inhibitors of Lyn activity. LynDeltaN also inhibits imatinib-mediated caspase-3 activation in the small proportion of nilotinib-resistant K562 cells overexpressing Lyn that can engage an apoptotic program upon imatinib stimulation. Finally, Lyn knockdown by sh-RNA altered neither imatinib-mediated apoptosis nor differentiation. Taken together, our data show that the caspase-cleaved form of Lyn exerts a negative feedback on imatinib-mediated CML cell apoptosis that is entirely dependent on its kinase activity and likely on the BCR-ABL pathway.


Subject(s)
Antineoplastic Agents/antagonists & inhibitors , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Neoplasm Proteins/physiology , Piperazines/antagonists & inhibitors , Protein Kinase Inhibitors/antagonists & inhibitors , Pyrimidines/antagonists & inhibitors , src-Family Kinases/physiology , Antineoplastic Agents/pharmacology , Benzamides , Caspase 9/genetics , Caspase Inhibitors , Enzyme Activation , Erythropoiesis/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/physiology , Humans , Imatinib Mesylate , Indoles/pharmacology , K562 Cells/drug effects , K562 Cells/enzymology , K562 Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/physiology , Signal Transduction/drug effects , Structure-Activity Relationship , Sulfonamides/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/chemistry , src-Family Kinases/genetics
7.
Ann Hematol ; 88(11): 1047-58, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19301004

ABSTRACT

During the last years remission rates of more than 72% for arsenic(III)-oxide (As(2)O(3)) treatment in relapsed or refractory acute promyelocytic leukemia have been published. As(2)O(3) is under clinical investigation for therapy of leukemia and solid tumors. Due to the chemical affinity of arsenic and antimony, we analyzed the potency of antimony(III)-oxide (Sb(2)O(3)) to exert As(2)O(3)-like effects. Based on the same molar concentrations, lower efficacy in apoptosis induction and caspase-independent decrease of mitochondrial membrane potential was observed for Sb(2)O(3). No difference in sensitivity to As(2)O(3) or Sb(2)O(3) was detected in CEM cells when compared to their multiple drug resistant derivatives. Apoptosis was induced by combining sub-apoptotic concentrations of Sb(2)O(3) or As(2)O(3) with sub-apoptotic concentrations of DL: -buthionine-[S,R]-sulfoximine (BSO). Other modulators of the cellular redox system showed this effect to a lower extent and enhancement was not consistent for the different cell lines tested. Caspase inhibitors protected cell lines from Sb(2)O(3)- and As(2)O(3)-induced apoptosis. When BSO was added, the inhibitors lost their protective ability. The ability of modulators of the cellular redox system in clinically applicable concentrations to enhance the apoptotic effects of the two oxides in a synergistic way may be helpful to reduce their toxicity by optimizing their dose.


Subject(s)
Antimony/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenicals/pharmacology , Glutathione/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, T-Cell/pathology , Oxides/pharmacology , Arsenic Trioxide , Buthionine Sulfoximine/pharmacology , Caspase Inhibitors , Caspases/metabolism , Cell Line, Tumor/cytology , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , HL-60 Cells/cytology , HL-60 Cells/drug effects , HL-60 Cells/enzymology , Humans , K562 Cells/cytology , K562 Cells/drug effects , K562 Cells/enzymology , Membrane Potential, Mitochondrial/drug effects , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oxidation-Reduction
8.
Clin Cancer Res ; 15(3): 914-23, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19188162

ABSTRACT

PURPOSE: Telomerase is considered currently as a hallmark of cancer, and its inhibition is expected to become an important anticancer modality. In contrast to abundant data concerning the effect of cytotoxic drugs on telomerase activity (TA), there is scant information on the effect of radiation on telomerase. The mechanism of telomerase regulation by irradiation has never been evaluated in detail. In the present study, we investigated the effect of radiation on TA and its regulation in cancer cells. EXPERIMENTAL DESIGN: The effect of various radiation doses on TA in several malignant and nonmalignant cell lines was evaluated. All malignant cells exhibited similar telomerase response to radiation and its regulation was assessed at transcriptional and post-translational levels in K562 cells. Next step was the evaluation of the upstream signaling pathways leading to changes in TA using kinetics and specific inhibitors. RESULTS: Radiation up-regulated TA in dose-dependent manner only in cancer cells. Telomerase was activated by phosphorylation by Akt and by cytoplasmic-nuclear shift. Transcriptional processes were not involved in TA. This telomerase regulation is mediated by Ras/phosphatidylinositol 3-kinase/Akt pathway. The canonical membrane effectors of irradiation (epidermal growth factor receptor, insulin-like growth factor-I receptor, and Ca2+ influx) were not involved in this process. CONCLUSIONS: Radiation up-regulates telomerase activity specifically in cancer cells. This study adds to accumulating evidence pointing to post-translational level as important mode of telomerase regulation. Telomerase activation due to radiation may be detrimental in treatment of cancer. Data described in this study may add to future interventions aiming at inhibition of telomerase activation during irradiation.


Subject(s)
Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Processing, Post-Transcriptional , Telomerase/radiation effects , ras Proteins/metabolism , Cell Compartmentation/radiation effects , Cell Line, Tumor , Cell Proliferation , Cell Survival , Dose-Response Relationship, Radiation , Humans , K562 Cells/enzymology , Kinetics , Signal Transduction , Telomerase/metabolism , Up-Regulation
9.
Leukemia ; 23(3): 585-90, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18754030
10.
Leukemia ; 23(3): 477-85, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19039322

ABSTRACT

The detailed molecular mechanism of action of second-generation BCR-ABL tyrosine kinase inhibitors, including perturbed targets and pathways, should contribute to rationalized therapy in chronic myeloid leukemia (CML) or in other affected diseases. Here, we characterized the target profile of the dual SRC/ABL inhibitor bosutinib employing a two-tiered approach using chemical proteomics to identify natural binders in whole cell lysates of primary CML and K562 cells in parallel to in vitro kinase assays against a large recombinant kinase panel. The combined strategy resulted in a global survey of bosutinib targets comprised of over 45 novel tyrosine and serine/threonine kinases. We have found clear differences in the target patterns of bosutinib in primary CML cells versus the K562 cell line. A comparison of bosutinib with dasatinib across the whole kinase panel revealed overlapping, but distinct, inhibition profiles. Common among those were the SRC, ABL and TEC family kinases. Bosutinib did not inhibit KIT or platelet-derived growth factor receptor, but prominently targeted the apoptosis-linked STE20 kinases. Although in vivo bosutinib is inactive against ABL T315I, we found this clinically important mutant to be enzymatically inhibited in the mid-nanomolar range. Finally, bosutinib is the first kinase inhibitor shown to target CAMK2G, recently implicated in myeloid leukemia cell proliferation.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , K562 Cells/drug effects , Leukemia, Myeloid, Accelerated Phase/enzymology , Neoplasm Proteins/antagonists & inhibitors , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinolines/pharmacology , Aniline Compounds/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Dasatinib , Drug Delivery Systems , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/antagonists & inhibitors , Gene Expression Profiling , Humans , K562 Cells/enzymology , Leukemia, Myeloid, Accelerated Phase/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Nitriles/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrimidines/pharmacology , Quinolines/chemistry , Signal Transduction/drug effects , Substrate Specificity , Thiazoles/pharmacology , src-Family Kinases/antagonists & inhibitors
11.
Leukemia ; 22(3): 572-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18079735

ABSTRACT

Small molecule tyrosine kinase inhibitors, such as imatinib, are effective therapies for BCR-ABL-mediated human leukemias. However, clinical drug resistance occurs, which warrants development of alternative and/or complementary therapeutic strategies to target critical downstream signaling molecules. We recently demonstrated that disrupting 14-3-3/ligand association by a peptide-based 14-3-3 competitive antagonist R18 induces significant apoptosis, partially through reactivation of AKT-inhibited proapoptotic FOXO3a, in FGFR1 fusion-transformed hematopoietic cells. Here, we report that targeting 14-3-3 by R18 effectively induced significant apoptosis in Ba/F3 and K562 cells expressing BCR-ABL, similarly through liberation and reactivation of FOXO3a. Moreover, R18 sensitized BCR-ABL-transformed cells to inhibition with MEK1 inhibitor U0126, Bcl-2 inhibitor GX15-070, or mTOR inhibitor rapamycin. Treatment with these reagents potentiated R18-induced reactivation of proapoptotic FOXO3a with enhanced expression of downstream transcription targets p27(kip1) and Bim1. Furthermore, R18-induced apoptotic cell death in cells expressing diverse imatinib-resistant BCR-ABL mutants, including T315I. This inhibition was enhanced by R18 in combination with U0126 and rapamycin. Thus, our findings suggest that targeting 14-3-3 may potentiate the effects of conventional therapy for BCR-ABL-associated hematopoietic malignancies, and overcome drug resistance.


Subject(s)
14-3-3 Proteins/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Butadienes/pharmacology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Nitriles/pharmacology , Peptides/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Sirolimus/pharmacology , Amino Acid Sequence , Apoptosis/drug effects , Benzamides , Drug Resistance, Neoplasm/genetics , Drug Screening Assays, Antitumor , Drug Synergism , Forkhead Box Protein O3 , Forkhead Transcription Factors/physiology , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , Indoles , Intracellular Signaling Peptides and Proteins , K562 Cells/drug effects , K562 Cells/enzymology , Molecular Sequence Data , Mutation, Missense , Neoplasm Proteins/physiology , Peptides/genetics , Piperazines/pharmacology , Point Mutation , Pyrimidines/pharmacology , Recombinant Fusion Proteins/antagonists & inhibitors , Signal Transduction/drug effects
12.
Altern Lab Anim ; 35(1): 79-85, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17411355

ABSTRACT

Methyl methanesulphonate (MMS) is a DNA damaging agent, which induces oxidative stress, ATP depletion, and consequently, cell death, in HL-60 and K562 cells. The cell death induced by MMS predominantly exhibited the morphological and biochemical hallmarks of necrosis. A minor population of dying cells exhibited apoptotic hallmarks, especially in K562 cell cultures. Cyclosporin A (CsA) was used to modulate the MMS-induced cell death. Our results indicated that CsA did not prevent cells from dying, but changed the mode of death from necrotic to apoptotic. Surprisingly, CsA enhanced oxidative stress and increased the overall number of dead cells. Based on these results, we conclude that the modulatory effect of CsA on MMS-induced cell death might arise from an interference by CsA with mitochondrial metabolism, rather than from inhibition of the MMS efflux mediated by P-glycoprotein.


Subject(s)
Cyclosporine/toxicity , HL-60 Cells/drug effects , Immunosuppressive Agents/toxicity , K562 Cells/drug effects , Methyl Methanesulfonate/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Apoptosis/drug effects , Caspase 3/biosynthesis , Cell Survival/drug effects , DNA Damage , DNA, Neoplasm/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Glutathione/metabolism , Glutathione Disulfide/metabolism , HL-60 Cells/enzymology , HL-60 Cells/pathology , Humans , K562 Cells/enzymology , K562 Cells/pathology , Necrosis/chemically induced , Oxidative Stress/drug effects , RNA, Messenger/metabolism , RNA, Neoplasm/analysis
13.
J Biol Chem ; 282(19): 14364-72, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17374613

ABSTRACT

Chronic myeloid leukemia is a hematopoietic stem cell cancer, originated by the perpetually "switched on" activity of the tyrosine kinase Bcr-Abl, leading to uncontrolled proliferation and insensitivity to apoptotic stimuli. The genetic phenotype of myeloid leukemic K562 cells includes the suppression of cytosolic sialidase Neu2. Neu2 transfection in K562 cells induced a marked decrease (-30% and -80%) of the mRNA of the anti-apoptotic factors Bcl-XL and Bcl-2, respectively, and an almost total disappearance of Bcl-2 protein. In addition, gene expression and activity of Bcr-Abl underwent a 35% diminution, together with a marked decrease of Bcr-Abl-dependent Src and Lyn kinase activity. Thus, the antiapoptotic axis Bcr-Abl, Src, and Lyn, which stimulates the formation of Bcl-XL and Bcl-2, was remarkably weakened. The ultimate consequences of these modifications were an increased susceptibility to apoptosis of K562 cells and a marked reduction of their proliferation rate. The molecular link between Neu2 activity and Bcr-Abl signaling pathway may rely on the desialylation of some cytosolic glycoproteins. In fact, three cytosolic glycoproteins, in the range 45-66 kDa, showed a 50-70% decrease of their sialic acid content upon Neu2 expression, supporting their possible role as modulators of the Bcr-Abl complex.


Subject(s)
Apoptosis , Fusion Proteins, bcr-abl/metabolism , Neuraminidase/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , src-Family Kinases/metabolism , Blotting, Western , Cell Proliferation , Down-Regulation , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Glycoproteins , Humans , K562 Cells/enzymology , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sphingolipids , Tyrosine/metabolism , bcl-X Protein/metabolism
14.
Haematologica ; 91(5): 711-2, 2006 May.
Article in English | MEDLINE | ID: mdl-16670078

ABSTRACT

We evaluated the effect of the human immunodeficiency virus (HIV) protease inhibitor saquinavir on the imatinib-sensitive and imatinib-resistant chronic myelogenous leukemia cell lines. Saquinavir, which is also a proteasome blocker, showed dose- and time-related anti-proliferative activity, particularly on the imatinib-resistant lines and a pro-apoptotic effect. Association with imatinib caused a significant increase of activity.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Piperazines/pharmacology , Protease Inhibitors/pharmacology , Pyrimidines/pharmacology , Saquinavir/pharmacology , Adolescent , Apoptosis/drug effects , Benzamides , Blast Crisis/drug therapy , Blast Crisis/pathology , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Imatinib Mesylate , Interferons/therapeutic use , K562 Cells/drug effects , K562 Cells/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myeloid, Chronic-Phase/drug therapy , Leukemia, Myeloid, Chronic-Phase/pathology , Male , Middle Aged , Proteasome Inhibitors
15.
Biochem Pharmacol ; 72(1): 11-8, 2006 Jun 28.
Article in English | MEDLINE | ID: mdl-16678798

ABSTRACT

The role of topoisomerase (topo) II in DNA repair has yet to be fully elucidated. Current evidence suggesting a role for topo II in the repair of DNA damage has been obtained by using in vitro model systems or inferred from correlative data in drug resistant cell lines. In this study we directly examined the role of topo IIalpha and beta in mediating the repair of melphalan-induced crosslinks in cellular DNA. To accomplish this, we used siRNA technology to knock down either topo IIalpha or beta in human chronic myelogenous leukemia K562 and histiocytic lymphoma U937 cell line. Our data demonstrate that topo IIbeta levels, (but not alpha), are a determinant of melphalan-induced crosslinks and sensitivity to melphalan. Furthermore, we show that knocking down topo IIbeta inhibits the repair of melphalan-induced crosslinks in K562 cells. These studies represent the first direct evidence that topo IIbeta participates in the repair of DNA damage induced by an alkylating agent in cellular DNA. Finally, these results suggest non-redundant roles for these two isoforms in mediating repair of DNA crosslinks.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cross-Linking Reagents/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , DNA/drug effects , Melphalan/pharmacology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Apoptosis/drug effects , Cell Death/drug effects , DNA Damage , DNA Repair/drug effects , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Humans , K562 Cells/drug effects , K562 Cells/enzymology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Topoisomerase II Inhibitors , Transfection , U937 Cells/drug effects , U937 Cells/enzymology
16.
Haematologica ; 91(5): 659-62, 2006 May.
Article in English | MEDLINE | ID: mdl-16627254

ABSTRACT

To identify a fast and sensitive method for screening for mutations in patients with imatinib- resistant chronic myeloid leukemia (CML), we compared allele specific oligonucleotide- polymerase chain reaction (ASO-PCR) assay with conventional direct sequencing. Among the 68 imatinib resistant CML patients studied, 18 amino acid substitutions were detected in 44 patients by two assays. The sensitivity of ASO-PCR was superior to that of direct sequencing as it could detect one mutant allele in 100 approximately 100,000 wild type sequences. The fastness, simplicity, and sensitivity of ASO-PCR assays will be useful for routine monitoring of mutations, especially for frequently identified mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Genes, abl , Genetic Testing/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Piperazines/pharmacology , Polymerase Chain Reaction/methods , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Sequence Analysis, DNA , Alleles , Amino Acid Substitution , Antineoplastic Agents/therapeutic use , Benzamides , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Imatinib Mesylate , K562 Cells/drug effects , K562 Cells/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Mutation, Missense , Piperazines/therapeutic use , Point Mutation , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Sensitivity and Specificity , Time Factors
18.
Protein Sci ; 12(10): 2282-90, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14500886

ABSTRACT

In this study, the human multienzyme aminoacyl-tRNA synthetase "core" complex has been isolated from the nuclear and cytosolic compartments of human cells and purified to near homogeneity. It is clear from the polypeptide compositions, stoichiometries, and three-dimensional structures that the cytosolic and nuclear particles are very similar to each other and to the particle obtained from rabbit reticulocytes. The most significant difference observed via aminoacylation activity assays and densitometric analysis of electrophoretic band patterns is a lower amount of glutaminyl-tRNA synthetase in the human particles. However, this is not enough to cause major changes in the three-dimensional structures calculated from samples negatively stained with either uranyl acetate or methylamine vanadate. Indeed, the latter samples produce volumes that are highly similar to an initial structure previously calculated from a frozen hydrated sample of the rabbit multisynthetase complex. New structures in this study reveal that the three major structural domains have discrete subsections. This information is an important step toward determination of specific protein interactions and arrangements within the multisynthetase core complex and understanding of the particle's cellular function(s). Finally, gel filtration and immunoblot analysis demonstrate that a major biological role for the cytokine precursor p43 is as an integral part of the multisynthetase complex.


Subject(s)
Amino Acyl-tRNA Synthetases/chemistry , Cell Nucleus/chemistry , Cytokines/chemistry , Cytoplasm/chemistry , Neoplasm Proteins/chemistry , RNA-Binding Proteins/chemistry , Amino Acids/metabolism , Amino Acyl-tRNA Synthetases/metabolism , Amino Acyl-tRNA Synthetases/ultrastructure , Animals , Cell Fractionation , Chromatography, High Pressure Liquid , Cytokines/isolation & purification , Cytokines/metabolism , Electrophoresis, Polyacrylamide Gel , Glutamate-tRNA Ligase/chemistry , Glutamate-tRNA Ligase/metabolism , Humans , Image Processing, Computer-Assisted , Immunoblotting , K562 Cells/chemistry , K562 Cells/enzymology , Kinetics , Microscopy, Electron , Models, Molecular , Neoplasm Proteins/isolation & purification , Neoplasm Proteins/metabolism , RNA-Binding Proteins/isolation & purification , RNA-Binding Proteins/metabolism , Rabbits , Structural Homology, Protein
19.
Leuk Res ; 27(6): 529-37, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12648513

ABSTRACT

A novel method for simultaneously detecting clonality by FISH, and presence of telomerase activity (telo+ cells) or histone H3 mRNA (H3+) in single cells from a mixed leukemic population is reported. The methods were validated using K562 cells mixed with peripheral blood granulocytes and bone marrow aspirate cells from newly diagnosed AML patients. Fifty patients with AML were analyzed for telo+ cells, while eight AML patients were analyzed for FISH-Telomerase and FISH-H3+ during remission induction therapy. Our results demonstrate that: (1). changes in the leukemic populations during therapy could be followed; (2). a favorable response to chemotherapy occurred when there was a reduction in both the cytogenetically abnormal cells along with reduction in telo+ cells within this abnormal population; (3). reduction of either telo+ cells or FISH+ cells alone did not correlate with good response. H3+ could be detected in only 4% of the leukemic population, most of which were cytogenetically abnormal. These newly established methods allow sub-populations of cells to be followed during disease progression and treatment and to elucidate factors that give a specific clone proliferative advantage.


Subject(s)
Histones/genetics , K562 Cells/enzymology , Telomerase/analysis , Acute Disease , Bone Marrow/enzymology , Bone Marrow/metabolism , DNA Primers/chemistry , Granulocytes/enzymology , Granulocytes/metabolism , Histones/metabolism , Humans , In Situ Hybridization, Fluorescence/methods , Karyotyping , Polymerase Chain Reaction , Prognosis , RNA, Messenger/analysis , Remission Induction
20.
Oncogene ; 22(7): 1012-23, 2003 Feb 20.
Article in English | MEDLINE | ID: mdl-12592388

ABSTRACT

beta-Hydroxyisovalerylshikonin (beta-HIVS), which was isolated from the plant, Lithospermum radix, induces apoptosis in various lines of human tumor cells. To identify genes involved in beta-HIVS-induced apoptotic process, we performed cDNA array analysis and found that beta-HIVS suppresses the expression of the gene for a polo-like kinase 1 (PLK1) that is involved in control of the cell cycle. When U937 and HL60 cells were treated with 10(-6) M beta-HIVS for 0.5 h, both the amount of PLK1 itself and the kinase activity of this enzyme were decreased. By contrast, Bcr-Abl-positive K562 cells were resistant to the induction of apoptosis by beta-HIVS and this compound did not suppress the kinase activity of PLK1 in these cells. However, simultaneous treatment of K562 cells with both beta-HIVS and STI571, which selectively inhibits the protein tyrosine kinase (PTK) activity of Bcr-Abl, strongly induced apoptosis. Moreover, beta-HIVS increased the inhibitory effect of STI571 on PTK activity. Treatment of K562 cells with antisense oligodeoxynucleotides (ODNs) specific for PLK1 sensitized these cells to the beta-HIVS-induced fragmentation of DNA. These results suggest that suppression of the activity of PLK1 via inhibition of tyrosine kinase activity by beta-HIVS might play a critical role in the induction of apoptosis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid/pathology , Naphthoquinones/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors , Protein Processing, Post-Translational/drug effects , Benzamides , Cell Cycle Proteins , Cell Line/drug effects , Cysteine Proteinase Inhibitors/pharmacology , DNA, Complementary/genetics , Fusion Proteins, bcr-abl/antagonists & inhibitors , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Genistein/pharmacology , HL-60 Cells/drug effects , HL-60 Cells/enzymology , Humans , Imatinib Mesylate , K562 Cells/drug effects , K562 Cells/enzymology , Kidney , Leukemia, Myeloid/enzymology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Oligodeoxyribonucleotides, Antisense/pharmacology , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Kinases/genetics , Protein Kinases/physiology , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-bcl-2/physiology , Pyrimidines/pharmacology , U937 Cells/drug effects , U937 Cells/enzymology , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...