Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Asian Pac J Cancer Prev ; 22(12): 3959-3965, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34967577

ABSTRACT

OBJECTIVE: BCR ABL oncogene encodes the BCR-ABL chimeric protein, which is a constitutively activated non-receptor tyrosine kinase. The BCR-ABL oncoprotein is a key molecular basis for the pathogenesis of chronic myeloid leukemia (CML) via activation of several downstream signaling pathways including JAK/STAT pathway. Development of leukemia involves constitutive activation of signaling molecules including, JAK2, STAT3, STAT5A and STAT5B. Thymoquinone (TQ) is a bioactive constituent of Nigella sativa that has shown anticancer properties in various cancers. The present study aimed to evaluate the effect of TQ on the expression of BCR ABL, JAK2, STAT3, STAT5A and STAT5B genes and their consequences on the cell proliferation and apoptosis in K562 CML cells. METHODS: BCR-ABL positive K562 CML cells were treated with TQ. Cytotoxicity was determined by Trypan blue exclusion assay. Apoptosis assay was performed by annexin V-FITC/PI staining assay and analyzed by flow cytometry. Transcription levels of BCR ABL, JAK2, STAT3, STAT5A and STAT5B genes were evaluated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Protein levels of JAK2 and STAT5 were determined by Jess Assay analysis. RESULTS: TQ markedly decreased the cell proliferation and induced apoptosis in K562 cells (P < 0.001) in a concentration dependent manner. TQ caused a significant decrease in the transcriptional levels of BCR ABL, JAK2, STAT3, STAT5A and STAT5B genes (P < 0.001). TQ induced a significant decrease in JAK2 and STAT5 protein levels (P < 0.001). CONCLUSION: our results indicated that TQ inhibited cell growth of K562 cells via downregulation of BCR ABL/ JAK2/STAT3 and STAT5 signaling and reducing JAK2 and STAT5 protein levels.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Genes, abl/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Signal Transduction/drug effects , Apoptosis/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Janus Kinase 2/drug effects , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , STAT3 Transcription Factor/drug effects , STAT5 Transcription Factor/drug effects , Signal Transduction/genetics , Tumor Suppressor Proteins/drug effects
2.
Biol Pharm Bull ; 43(10): 1526-1533, 2020.
Article in English | MEDLINE | ID: mdl-32999163

ABSTRACT

Imatinib-resistance is a significant concern for Bcr-Abl-positive chronic myelogenous leukemia (CML) treatment. Emodin, the predominant compound of traditional medicine rhubarb, was reported to inhibit the multidrug resistance by downregulating P-glycoprotein of K562/ADM cells with overexpression of P-glycoprotein in our previous studies. In the present study, we found that emodin can be a potential inhibitor for the imatinib-resistance in K562/G01 cells which are the imatinib-resistant subcellular line of human chronic myelogenous leukemia cells with overexpression of breakpoint cluster region-abelson (Bcr-Abl) oncoprotein. Emodin greatly enhanced cell sensitivity to imatinib, suppressed resistant cell proliferation and increased potentiated apoptosis induced by imatinib in K562/G01 cells. After treatment of emodin and imatinib together, the levels of p-Bcr-Abl and Bcr-Abl were significantly downregulated. Moreover, Bcr-Abl important downstream target, STAT5 and its phosphorylation were affected. Furthermore, the expression of Bcr-Abl and signal transducers and activators of transcription 5 (STAT5) related molecules, including c-MYC, MCL-1, poly(ADP-ribose)polymerase (PARP), Bcl-2 and caspase-3, were changed. Emodin also decreased Src expression and its phosphorylation. More importantly, emodin simultaneously targeted both the ATP-binding and allosteric sites on Bcr-Abl by molecular docking, with higher affinity with the myristoyl-binding site for enhanced Bcr-Abl kinase inhibition. Overall, these data indicated emodin might be an effective therapeutic agent for inhibiting resistance to imatinib in CML treatment.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Emodin/pharmacology , Genes, abl/drug effects , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , STAT5 Transcription Factor/antagonists & inhibitors , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , Emodin/therapeutic use , Genes, abl/physiology , Humans , Imatinib Mesylate/therapeutic use , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Molecular Docking Simulation/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Secondary , STAT5 Transcription Factor/metabolism
3.
Nanomedicine ; 29: 102283, 2020 10.
Article in English | MEDLINE | ID: mdl-32777451

ABSTRACT

Nanotechnology has demonstrated great promise for the development of more effective and safer cancer therapies. We recently developed a highly selective inhibitor of BCR-ABL fusion tyrosine kinase for chronic myeloid leukemia (CML). However, the poor drug-like properties were hurdles to its further clinical development. Herein, we re-investigate it by conjugating an amphiphilic polymer and self-assembling into a nanoparticle (NP) with a high loading (~10.3%). The formulation greatly improved its solubility and drastically extended its circulation half-life from ~5.3 to ~117 h (>20-fold). In the 150 days long-term engraftment model experiment, long intravenous dosing intervals of the NPs (every 4 or 8 days) exhibited much better survival and negligible toxicities as compared to daily oral administration of the inhibitor. Moreover, the NPs showed excellent inhibition of tumor growth in the subcutaneous xenograft model. All results suggest that the ultra-long circulating pro-drug NP may provide an effective and safe therapeutic strategy for BCR-ABL-positive CML.


Subject(s)
Genes, abl/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Nanoparticles/chemistry , Protein Kinase Inhibitors/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Genes, abl/genetics , Humans , Imatinib Mesylate/chemistry , Imatinib Mesylate/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology
4.
Nucleic Acid Ther ; 27(3): 168-175, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28355131

ABSTRACT

Herein we described the synthesis of siRNA-NES (nuclear export signal) peptide conjugates by solid phase fragment coupling and the application of them to silencing of bcr/abl chimeric gene in human chronic myelogenous leukemia cell line K562. Two types of siRNA-NES conjugates were prepared, and both sense strands at 5' ends were covalently linked to a NES peptide derived from TFIIIA and HIV-1 REV, respectively. Significant enhancement of silencing efficiency was observed for both of them. siRNA-TFIIIA NES conjugate suppressed the expression of BCR/ABL gene to 8.3% at 200 nM and 11.6% at 50 nM, and siRNA-HIV-1REV NES conjugate suppressed to 4.0% at 200 nM and 6.3% at 50 nM, whereas native siRNA suppressed to 36.3% at 200 nM and 30.2% at 50 nM. We could also show complex of siRNA-NES conjugate and designed amphiphilic peptide peptideß7 could be taken up into cells with no cytotoxicity and showed excellent silencing efficiency. We believe that the complex siRNA-NES conjugate and peptideß7 is a promising candidate for in vivo use and therapeutic applications.


Subject(s)
Gene Silencing , Genes, abl/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Peptide Fragments/pharmacology , RNA, Small Interfering/pharmacology , Absorption, Physicochemical , Dose-Response Relationship, Drug , Drug Discovery/trends , Fusion Proteins, bcr-abl/genetics , Gene Expression Regulation, Leukemic , Genes, abl/genetics , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Nanomedicine/trends , Nuclear Export Signals/genetics , Nuclear Pore Complex Proteins/chemistry , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics , RNA-Binding Proteins/chemistry , Transcription Factor TFIIIA/chemistry , Transfection
5.
Int Immunopharmacol ; 36: 17-22, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27104314

ABSTRACT

Polyphyllin D, a compound derived from Paris polyphylla rhizoma, demonstrated strong anticancer activities in a previous study. Our results demonstrated that polyphyllin D exerts a growth inhibitory effect by inducing apoptosis and differentiation in the human erythroleukemia cell line K562. Polyphyllin D induced apoptosis via the mitochondrial apoptotic pathway, as evidenced by the decreased Bcl-2 and Bcr/Abl expression levels, the disruption of MMP and increased Bax, cytochrome c and cleaved-caspase-3 levels. At a low dose, polyphyllin D increased CD14 expression on the surface of K562 cells and induced cells to differentiate into monocytes or mature macrophages. These data suggest that polyphyllin D has the potential to be a potent therapeutic agent for treating human chronic myelogenous leukemia.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Diosgenin/analogs & derivatives , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Liliaceae/immunology , Macrophages/drug effects , Medicine, Chinese Traditional , Mitochondria/drug effects , Saponins/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cell Differentiation/drug effects , Diosgenin/pharmacology , Genes, abl/drug effects , Humans , K562 Cells , Macrophages/physiology , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcr/genetics
6.
J Pharmacol Sci ; 129(4): 210-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26644081

ABSTRACT

Imatinib, a multitargeted tyrosine kinase inhibitor, exhibits potent anticancer activity against leukemia harboring the Bcr-Abl oncogene and some solid tumors overexpressing c-kit and PDGFR. However, its clinical efficacy is severely compromised by the emergence of resistance primarily due to acquired mutations in the Bcr-Abl kinase domain. In this study, we showed that combination of imatinib with platinum (Pt)-based anticancer agents, including cisplatin and oxaliplatin, exhibited synergistic cytotoxic effect specifically in Bcr-Abl+ human chronic myeloid leukemia cell line K562 but not in Bcr-Abl- RPMI8226 cells. Importantly, the synergistic effect was also found to circumvent imatinib resistance in an imatinib-selected resistant subline K562 ima1.0. The combination treatment increased apoptosis and DNA damage. Mechanistic study revealed that increased inhibition of Bcr-Abl and downstream ERK phosphorylation by the drug combination may contribute to the synergistic effect.


Subject(s)
Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Cisplatin/toxicity , Enzyme Inhibitors/toxicity , Genes, abl , Imatinib Mesylate/toxicity , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Organoplatinum Compounds/toxicity , DNA Damage/drug effects , DNA, Neoplasm , Drug Synergism , Genes, abl/drug effects , Humans , K562 Cells , MAP Kinase Signaling System/drug effects , Oxaliplatin , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors
7.
Biochem Pharmacol ; 92(2): 235-50, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25264277

ABSTRACT

Imatinib mesylate is a tyrosine kinase inhibitor with selectivity for abelson tyrosine-protein kinase 1 (c-Abl), breakpoint cluster region (Bcr)-Abl fusion protein (Bcr-Abl), mast/stem cell growth factor receptor Kit (c-Kit), and platelet-derived growth factor receptor (PDGFR). Previous studies demonstrated that imatinib in the low micromolar range exerted antiproliferative effects on neuroblastoma cell lines. However, although neuroblastoma cells express c-Kit and PDGFR, the imatinib concentrations required to achieve significant growth inhibitory effects (≥ 10 µM) are substantially higher than those required for inhibition of ligand-induced phosphorylation of wild type c-Kit and PDGFR (≤ 1 µM), suggesting that additional mechanisms are responsible for the antitumor activity of imatinib on these cells. In this study, we show that treatment of neuroblastoma cell lines with 1-15 µM imatinib resulted in a dose dependent inhibition of 5-bromo-2'-deoxyuridine (BrdU) incorporation into newly synthesized DNA. The antiproliferative effect of imatinib was dependent on the upregulation of the cyclin-dependent kinase (CDK) inhibitor p27(KIP1) in the nuclear compartment as a result of increased p27(KIP1) protein stability. We demonstrate that the mechanism of p27(KIP1) stabilization relied on inhibition of p27(KIP1) phosphorylation on tyrosine residues by c-Abl. We provide evidence that in neuroblastoma cell lines a significant fraction of cellular c-Abl is phosphorylated on Tyr-245, consistent with an open and active conformation. Notably, exposure to imatinib did not affect Tyr-245 phosphorylation. Given the low affinity of active c-Abl for imatinib, these data provide a molecular explanation for the relatively high imatinib concentrations required to inhibit neuroblastoma cell proliferation.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Genes, abl/physiology , Neuroblastoma/metabolism , Piperazines/pharmacology , Pyrimidines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Genes, abl/drug effects , Humans , Imatinib Mesylate , Up-Regulation/drug effects , Up-Regulation/physiology
8.
Mol Neurobiol ; 49(2): 1102-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24510275

ABSTRACT

Prion diseases are neurodegenerative disorders characterized by the accumulation of a disease-associated prion protein and apoptotic neuronal death. Previous studies indicated that the ubiquitous expression of c-Abl tyrosine kinase transduces a variety of extrinsic and intrinsic cellular signals. In this study, we demonstrated that a synthetic neurotoxic prion fragment (PrP106-126) activated c-Abl tyrosine kinase, which in turn triggered the upregulation of MST1 and BIM, suggesting the activation of the c-Abl-BIM signaling pathway. The peptide fragment was found to result in cell death via mitochondrial dysfunction in neuron cultures. Knockdown of c-Abl using small interfering RNA protected neuronal cells from PrP106-126-induced mitochondrial dysfunction, production of reactive oxygen species, and apoptotic events inducing translocation of Bax to the mitochondria, cytochrome c release into the cytosol, and activation of caspase-9 and caspase-3. Blocking the c-Abl tyrosine kinase also prevented neuronal cells from PrP106-126-induced apoptotic morphological changes. This is the first study reporting that c-Abl tyrosine kinase as a novel upstream activator of MST1 and BIM plays an important role in prion-induced neuron apoptosis via mitochondrial dysfunction. Our findings suggest that c-Abl tyrosine kinase is a potential therapeutic target for prion disease.


Subject(s)
Apoptosis/physiology , Genes, abl/physiology , Homeostasis/physiology , Mitochondria/enzymology , Neurons/enzymology , Peptide Fragments/toxicity , Prions/toxicity , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Genes, abl/drug effects , Homeostasis/drug effects , Mice , Mitochondria/drug effects , Mitochondria/ultrastructure , Molecular Sequence Data , Neurons/drug effects , Neurons/ultrastructure , Rats
9.
Biochem Pharmacol ; 87(2): 229-42, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24161787

ABSTRACT

Andrographolide is a diterpenoid compound isolated from Andrographis paniculata that exhibits anticancer activity. We previously reported that andrographolide suppressed v-Src-mediated cellular transformation by promoting the degradation of Src. In the present study, we demonstrated the involvement of Hsp90 in the andrographolide-mediated inhibition of Src oncogenic activity. Using a proteomics approach, a cleavage fragment of Hsp90α was identified in andrographolide-treated cells. The concentration- and time-dependent induction of Hsp90 cleavage that accompanied the reduction in Src was validated in RK3E cells transformed with either v-Src or a human truncated c-Src variant and treated with andrographolide. In cancer cells, the induction of Hsp90 cleavage by andrographolide and its structural derivatives correlated well with decreased Src levels, the suppression of transformation, and the induction of apoptosis. Moreover, the andrographolide-induced Hsp90 cleavage, Src degradation, inhibition of transformation, and induction of apoptosis were abolished by a ROS inhibitor, N-acetyl-cysteine. Notably, Hsp90 cleavage, decreased levels of Bcr-Abl (another known Hsp90 client protein), and the induction of apoptosis were also observed in human K562 leukemia cells treated with andrographolide or its active derivatives. Together, we demonstrated a novel mechanism by which andrographolide suppressed cancer malignancy that involved inhibiting Hsp90 function and reducing the levels of Hsp90 client proteins. Our results broaden the molecular basis of andrographolide-mediated anticancer activity.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Diterpenes/pharmacology , Genes, abl/physiology , Genes, src/physiology , HSP90 Heat-Shock Proteins/metabolism , Reactive Oxygen Species/metabolism , Amino Acid Sequence , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Diterpenes/therapeutic use , Down-Regulation/drug effects , Down-Regulation/physiology , Genes, abl/drug effects , Genes, src/drug effects , HSP90 Heat-Shock Proteins/genetics , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Molecular Sequence Data
12.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 29(1): 19-22, 2012 Feb.
Article in Chinese | MEDLINE | ID: mdl-22311485

ABSTRACT

OBJECTIVE: To investigate the effect of dihydroartemisinin (DHA) on the BCR/ABL fusion gene in leukemia K562 cell. METHODS: K562 cells were cultured in vitro. The rate of proliferation inhibition of cells treated with various concentrations of DHA were determined by using [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (MTT) method. Expression of BCR/ABL fusion gene was analyzed by reverse transcription(RT-PCR) before and after DHA treatment. Apoptosis of K562 cells was detected by flow cytometry. RESULTS: The growth of K562 cells was inhibited when the concentrations of DHA were 10-160 umol/L. With the added dose of DHA, the growth inhibition was remarkable, with the rate of inhibition risen from 52.76% to 94.65%. The expression of BCR/ABL fusion gene, as detected by RT-PCR after incubating the K562 cells with 20 umol/L DHA, measured as ΔCt = 4.45 ± 0.25 after 12 h and ΔCt = 5.23 ± 0.21 after 24 h, which was significantly lower compared with that of the control ( ΔCt = 4.23 ± 0.21, P < 0.05). CONCLUSION: DHA can inhibit the proliferation of leukemia K562 cells and facilitate the induction of apoptosis by downregulating the expression of BCR/ABL fusion gene.


Subject(s)
Artemisinins/pharmacology , Fusion Proteins, bcr-abl/biosynthesis , Genes, abl/drug effects , Leukemia/genetics , Fusion Proteins, bcr-abl/genetics , Gene Expression/drug effects , Humans , K562 Cells , Tumor Cells, Cultured
13.
FASEB J ; 25(10): 3661-73, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21705667

ABSTRACT

Inactivation of glycogen synthase kinase (GSK)-3 has been implicated in cancer progression. Previously, we showed an abundance of inactive GSK-3 in the human chronic myeloid leukemia (CML) cell line. CML is a hematopoietic malignancy caused by an oncogenic Bcr-Abl tyrosine kinase. In Bcr-Abl signaling, the role of GSK-3 is not well defined. Here, we report that enforced expression of constitutively active GSK-3 reduced proliferation and increased Bcr-Abl inhibition-induced apoptosis by nearly 1-fold. Bcr-Abl inhibition activated GSK-3 and GSK-3-dependent apoptosis. Inactivation of GSK-3 by Bcr-Abl activity is, therefore, confirmed. To reactivate GSK-3, we used glucosylceramide synthase (GCS) inhibitor PDMP to accumulate endogenous ceramide, a tumor-suppressor sphingolipid and a potent GSK-3 activator. We found that either PDMP or silence of GCS increased Bcr-Abl inhibition-induced GSK-3 activation and apoptosis. Furthermore, PDMP sensitized the most clinical problematic drug-resistant CML T315I mutant to Bcr-Abl inhibitor GNF-2-, imatinib-, or nilotinib-induced apoptosis by >5-fold. Combining PDMP and GNF-2 eliminated transplanted-CML-T315I-mutants in vivo and dose dependently sensitized primary cells from CML T315I patients to GNF-2-induced proliferation inhibition and apoptosis. The synergistic efficacy was Bcr-Abl restricted and correlated to increased intracellular ceramide levels and acted through GSK-3-mediated apoptosis. This study suggests a feasible novel anti-CML strategy by accumulating endogenous ceramide to reactivate GSK-3 and abrogate drug resistance.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Genes, abl , Glucosyltransferases/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Morpholines/pharmacology , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , Animals , Apoptosis/physiology , Cell Line, Tumor , Ceramides/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , Genes, abl/drug effects , Genes, abl/physiology , Glycogen Synthase Kinase 3/metabolism , Humans , Immunoglobulin G , Melphalan , Mice , Mice, SCID , Mutation , Neoplasms, Experimental , Pyrimidines , Transplantation, Heterologous
14.
Eur J Haematol ; 86(3): 216-25, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21198861

ABSTRACT

BCR/ABL positive cells are known to be resistant to DNA damage induced by chemotherapy while they are sensitive to imatinib (IM), a tyrosine kinase inhibitor (TKI). To evaluate whether this drug can increase the activity of cytotoxic drugs on BCR/ABL positive cells, we measured the toxicity of cytosine arabinoside (ARA-C), hydroxyurea (HU) and melphalan (MEL), after a pretreatment of 24 h with IM on K562 cell line. The highest cytotoxic effect was seen when the TKI was followed by MEL; our results indicate that inhibition of BCR/ABL activity by IM increased the cytotoxicity of MEL by favoring the DNA damage induced by this drug and by shortening the time for DNA repair at the G2/M checkpoint. A stronger activation of some genes involved in both intrinsic and extrinsic apoptotic pathways was also observed with IM/MEL combination compared to IM or MEL alone. The drugs association was further tested in a type of BaF3 cells (TonB.210) where the BCR-ABL expression is inducible by doxycycline; in this model it was confirmed that a reduction of BCR/ABL activity resulted in an increased susceptibility to the cytotoxic effect of MEL. Furthermore, we studied the effect of IM/MEL treatment on the proliferative potential of myeloid progenitors of six CML patients at diagnosis. The analysis of CFU-GM and BFU-E colonies demonstrated that the IM/MEL combination was more effective than IM alone in reducing the overall number of colonies and the number of copies of BCR/ABL. In conclusion, our work shows that inhibition of BCR/ABL activity increases the toxicity of MEL and allows an efficient killing of leukemic cells, suggesting that a clinical development of this approach could have therapeutic advantages for CML patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Melphalan/administration & dosage , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Apoptosis/drug effects , Benzamides , Cell Cycle/drug effects , Cytarabine/administration & dosage , DNA Damage , Drug Synergism , Gene Expression/drug effects , Genes, abl/drug effects , Humans , Hydroxyurea/administration & dosage , Imatinib Mesylate , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Tumor Stem Cell Assay
15.
J Hepatol ; 55(3): 612-625, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21251937

ABSTRACT

BACKGROUND & AIMS: Nilotinib is a novel tyrosine kinase inhibitor of Bcr-Abl and other kinases. In this study, we have examined its activity as an anti-fibrotic agent. METHODS: The in vitro effect of Nilotinib on rat and human HSCs was assessed using proliferation assays and Western blotting. The in vivo antifibrotic efficacy of Nilotinib was assessed in mice with liver fibrosis induced by CCl(4) and bile duct ligation (BDL). RESULTS: Nilotinib inhibited proliferation, migration, and actin filament formation, as well as the expression of α-SMA and collagen in activated HSCs. Nilotinib induced apoptosis of HSCs, which was correlated with reduced bcl-2 expression, increased p53 expression, cleavage of PARP, as well as increased expression of PPARγ and TRAIL-R. Nilotinib also induced cell cycle arrest, accompanied by increased expression of p27 and downregulation of cyclin D1. Interestingly, Nilotinib not only inhibited activation of PDGFR, but also TGFRII through Src. Nilotinib significantly inhibited PDGF and TGFß-simulated phosphorylation of ERK and Akt. Furthermore, PDGF- and TGFß-activated phosphorylated form(s) of Abl in human HSCs were inhibited by Nilotinib. In vivo, Nilotinib reduced collagen deposition and α-SMA expression in CCl(4) and BDL-induced fibrosis. These beneficial effects were associated with suppressed expression of procollagen-(I), TIMP-1, CD31, CD34, VEGF, and VEGFR. Nilotinib could induce HSC undergoing apoptosis in vivo, which was correlated with downregulation of bcl-2. We also observed reduced expression of phosphorylated ERK, Akt, and Abl in the Nilotinib-treated CCl(4) and BDL livers. In addition to its antifibrotic activity, the drug was hepatoprotective and reduced the elevations of ALT and AST after CCl(4) and BDL. CONCLUSIONS: These studies uncover a novel role of Bcr-Abl activity in treatment of liver fibrosis through multiple mechanisms and indicate that Nilotinib represents a potentially effective antifibrotic agent.


Subject(s)
Genes, abl/drug effects , Liver Cirrhosis/pathology , Platelet-Derived Growth Factor/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta/drug effects , Actins/metabolism , Animals , Apoptosis/drug effects , Bile Ducts , Carbon Tetrachloride , Cell Proliferation/drug effects , Collagen/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Ligation , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred BALB C , PPAR gamma/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism
16.
Mt Sinai J Med ; 77(6): 573-86, 2010.
Article in English | MEDLINE | ID: mdl-21105121

ABSTRACT

With an understanding of the molecular changes that accompany cell transformation, cancer drug discovery has undergone a dramatic change in the past few years. Whereas most of the emphasis in the past has been placed on developing drugs that induce cell death based on mechanisms that do not discriminate between normal and tumor cells, recent strategies have emphasized targeting specific mechanisms that have gone awry in tumor cells. However, the identification of cancer-associated mutations in oncogenes and their amplification in tumors has suggested that inhibitors against such proteins might represent attractive substrates for targeted therapy. In the clinic, the success of imatinib (Gleevec®, STI571) and trastuzumab (Herceptin®), both firsts of their kind, spurred further development of new, second-generation drugs that target kinases in cancer. This review highlights a few important examples each of these types of therapies, along with some newer agents that are in various stages of development. Second-generation kinase inhibitors aimed at overriding emerging resistance to these therapies are also discussed.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/drug effects , Antibodies, Monoclonal, Humanized , Dasatinib , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Gefitinib , Genes, abl/drug effects , Genes, abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Molecular Targeted Therapy , Pyrimidines/therapeutic use , Quinazolines/therapeutic use , Thiazoles/therapeutic use , Trastuzumab
17.
Nat Med ; 15(10): 1179-85, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19783996

ABSTRACT

Germ cells are sensitive to genotoxins, and ovarian failure and infertility are major side effects of chemotherapy in young patients with cancer. Here we describe the c-Abl-TAp63 pathway activated by chemotherapeutic DNA-damaging drugs in model human cell lines and in mouse oocytes and its role in cell death. In cell lines, upon cisplatin treatment, c-Abl phosphorylates TAp63 on specific tyrosine residues. Such modifications affect p63 stability and induce a p63-dependent activation of proapoptotic promoters. Similarly, in oocytes, cisplatin rapidly promotes TAp63 accumulation and eventually cell death. Treatment with the c-Abl kinase inhibitor imatinib counteracts these cisplatin-induced effects. Taken together, these data support a model in which signals initiated by DNA double-strand breaks are detected by c-Abl, which, through its kinase activity, modulates the p63 transcriptional output. Moreover, they suggest a new use for imatinib, aimed at preserving oocytes of the follicle reserve during chemotherapeutic treatments.


Subject(s)
Apoptosis/drug effects , Genes, abl/drug effects , Oocytes/metabolism , Phosphoproteins/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors , Animals , Benzamides , Cell Death/drug effects , Cells, Cultured , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , DNA Repair/drug effects , Dose-Response Relationship, Drug , Female , Imatinib Mesylate , In Situ Nick-End Labeling , Mice , Phosphorylation/drug effects , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology
18.
Anat Rec (Hoboken) ; 292(7): 945-50, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19548306

ABSTRACT

Imatinib mesylate is effective against Ph chromosome-positive leukemia; however, resistance has been reported. High expression of bcr-abl in mRNA and protein levels, and other alterations were found in patients who experienced imatinib treatment failures and thus it is important to design alternative treatment strategies. The aim of this study was to evaluate the in vitro effect of berbamine, on imatinib-resistant chronic myelogenous leukemia (CML) K562 (K562-r) cells, and explore the mechanisms. The growth of K562-r cells was examined using the 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay. Morphological analysis and DNA agarose electrophoresis were used to detect apoptosis in K562-r cells, and the extent of the cells in the sub-G1 cell cycle phase was measured using flow cytometry. The expression levels of BCR-ABL, phospho-BCR-ABL, and nuclear factor kappaB (NF-kappaB), IkappaBalpha, phospho-IkappaBalpha, IkappaB kinases alpha(IKKalpha), and Survivin were determined by Western blot. bcr-abl mRNA expression was determined by RT-PCR. MTT assays indicated that berbamine significantly inhibited the proliferation of K562-r cells. Cells with characteristics of apoptosis were confirmed by morphology examination and DNA agarose electrophoresis and percentage of apoptosis were increased after treatment with berbamine. The results also showed that berbamine was able to down-regulate BCR-ABL and phospho-BCR-ABL proteins by affecting bcr-abl mRNA expression and decrease expression of nuclear NF-kappaB, phospho-IkappaBalpha, IKKalpha, and Survivin. Collectively, berbamine could inhibit the proliferation of K562-r cells and induce apoptosis. The mechanisms may be related at least in part, to inhibit BCR-ABL and its downstream NF-kappaB signaling. Berbamine may provide an alternative candidate for the treatment of patients with CML resistant to imatinib therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzylisoquinolines/pharmacology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , NF-kappa B/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis/physiology , Benzamides , Cell Line, Tumor , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Evaluation, Preclinical/methods , G1 Phase/drug effects , G1 Phase/physiology , Genes, abl/drug effects , Genes, abl/genetics , Humans , I-kappa B Proteins/drug effects , I-kappa B Proteins/metabolism , Imatinib Mesylate , Indicators and Reagents , Inhibitor of Apoptosis Proteins , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Phosphorylation/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Survivin , Tetrazolium Salts
19.
Cancer Res ; 69(11): 4904-10, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19458080

ABSTRACT

During therapy for chronic myeloid leukemia (CML), decline of the number of BCR-ABL transcripts has been shown to follow a biphasic pattern, with a fast phase followed by a slower phase. Hence, sustained remission requires a long phase of therapy. Data indicate that a combination of different available targeted drugs might prevent treatment failure due to drug resistance, especially at advanced stages of the disease. However, for long-term multiple-drug treatments, complications can arise from side effects. We investigate whether and how the number of drugs could be reduced during long-term therapy. Using computational models, we show that one or more drugs can be removed once the number of tumor cells is reduced significantly, without compromising the chances of sustained tumor suppression. Which drug to remove first depends on the number of mutations in the BCR-ABL gene that confer resistance to the drugs, as well as on how effectively the drugs inhibit Bcr-Abl protein tyrosine kinase activity and inhibit tumor growth. We further show that the number of CML cells at which the number of drugs can be reduced does not correlate with the two phases of decline of the BCR-ABL transcript numbers. Neither does it depend much on kinetic parameters of CML growth, except for the mutation rates at which resistance is generated. This is a significant finding because even without any information on most parameters, and using only the data on the number of cancer cells and the rate at which resistant mutants are generated, it is possible to predict at which stage of treatment the number of drugs can be reduced.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Genes, abl/drug effects , Genes, abl/genetics , Humans , Kinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Models, Biological , Mutation/physiology , Substrate Specificity , Time Factors , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/physiology
20.
Haematologica ; 93(8): 1145-54, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18519516

ABSTRACT

BACKGROUND: Dasatinib is a multitargeted inhibitor of ABL, the SRC family, and other tyrosine kinases. We sought to evaluate the effects of this drug on cell proliferation, centrosomes, mitotic spindles, and cell cycle progression in vitro and in vivo. DESIGN AND METHODS: Human dermal fibroblasts, Chinese hamster cells, human osteosarcoma cells, and blood and bone marrow mononuclear cells from 32 patients with chronic myeloid leukemia, gastrointestinal stromal tumor, and systemic mastocytosis as well as from six healthy individuals were investigated. The effects of dasatinib were compared with those of the ABL inhibitors imatinib and nilotinib, the SRC inhibitor PP2, and the ABL/LYN inhibitor INNO-406. RESULTS: Dasatinib induced G(1) phase arrest in all cell lines and this was associated with a decline in cyclin D1 levels. In vitro, centrosomal aberrations, a decrease of mitotic spindles, and G(1) phase arrest were observed. In patients, centrosome alterations were found in a median of 17% (range, 10-22%) of cells with a decrease of spindles in 8/18 patients. In comparison, imatinib, nilotinib and PP2 led to centrosome aberrations without G(1) phase arrest. INNO-406 was associated with centrosome aberrations and cell cycle arrest in G(1) phase. CONCLUSIONS: Dasatinib blocks the G(1)/S transition and inhibits cell growth. It induces centrosomal aberrations and a decrease of mitotic spindles. The effects suggest an involvement of SRC and ABL inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Centrosome/drug effects , Fibroblasts/physiology , G1 Phase/genetics , Genes, abl/drug effects , Genes, src/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Animals , Benzamides , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , CHO Cells , Cell Division/drug effects , Cell Line, Tumor , Cricetinae , Cricetulus , G1 Phase/drug effects , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Osteosarcoma , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Skin Physiological Phenomena
SELECTION OF CITATIONS
SEARCH DETAIL
...