Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
PLoS Genet ; 9(4): e1003423, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23593023

RESUMEN

Mammalian chromosomes initiate DNA replication at multiple sites along their length during each S phase following a temporal replication program. The majority of genes on homologous chromosomes replicate synchronously. However, mono-allelically expressed genes such as imprinted genes, allelically excluded genes, and genes on female X chromosomes replicate asynchronously. We have identified a cis-acting locus on human chromosome 6 that controls this replication-timing program. This locus encodes a large intergenic non-coding RNA gene named Asynchronous replication and Autosomal RNA on chromosome 6, or ASAR6. Disruption of ASAR6 results in delayed replication, delayed mitotic chromosome condensation, and activation of the previously silent alleles of mono-allelic genes on chromosome 6. The ASAR6 gene resides within an ∼1.2 megabase domain of asynchronously replicating DNA that is coordinated with other random asynchronously replicating loci along chromosome 6. In contrast to other nearby mono-allelic genes, ASAR6 RNA is expressed from the later-replicating allele. ASAR6 RNA is synthesized by RNA Polymerase II, is not polyadenlyated, is restricted to the nucleus, and is subject to random mono-allelic expression. Disruption of ASAR6 leads to the formation of bridged chromosomes, micronuclei, and structural instability of chromosome 6. Finally, ectopic integration of cloned genomic DNA containing ASAR6 causes delayed replication of entire mouse chromosomes.


Asunto(s)
Cromosomas Humanos Par 6 , Replicación del ADN/genética , Mitosis , ARN Largo no Codificante , Alelos , Animales , Línea Celular , Inestabilidad Cromosómica/genética , Cromosomas Humanos Par 6/genética , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Hibridación Fluorescente in Situ , Ratones , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cromosoma X/genética , Cromosoma X/metabolismo
2.
Cancer Cell ; 10(1): 65-75, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16843266

RESUMEN

Tyrosine kinases are aberrantly activated in numerous malignancies, including acute myeloid leukemia (AML). To identify tyrosine kinases activated in AML, we developed a screening strategy that rapidly identifies tyrosine-phosphorylated proteins using mass spectrometry. This allowed the identification of an activating mutation (A572V) in the JAK3 pseudokinase domain in the acute megakaryoblastic leukemia (AMKL) cell line CMK. Subsequent analysis identified two additional JAK3 alleles, V722I and P132T, in AMKL patients. JAK3(A572V), JAK3(V722I), and JAK3(P132T) each transform Ba/F3 cells to factor-independent growth, and JAK3(A572V) confers features of megakaryoblastic leukemia in a murine model. These findings illustrate the biological importance of gain-of-function JAK3 mutations in leukemogenesis and demonstrate the utility of proteomic approaches to identifying clinically relevant mutations.


Asunto(s)
Leucemia Experimental/genética , Leucemia Megacarioblástica Aguda/genética , Proteínas Tirosina Quinasas/genética , Alelos , Animales , Apoptosis/efectos de los fármacos , Benzamidas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Mesilato de Imatinib , Janus Quinasa 2 , Janus Quinasa 3 , Células K562 , Leucemia Experimental/metabolismo , Leucemia Experimental/patología , Leucemia Megacarioblástica Aguda/metabolismo , Leucemia Megacarioblástica Aguda/patología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , TYK2 Quinasa
3.
Hum Mol Genet ; 20(12): 2366-78, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21459774

RESUMEN

Mammalian DNA replication initiates at multiple sites along chromosomes at different times, following a temporal replication program. Homologous alleles typically replicate synchronously; however, mono-allelically expressed genes such as imprinted genes, allelically excluded genes and genes on the female X chromosome replicate asynchronously. We have used a chromosome engineering strategy to identify a human autosomal locus that controls this replication timing program in cis. We show that Cre/loxP-mediated rearrangements at a discrete locus at 6q16.1 result in delayed replication of the entire chromosome. This locus displays asynchronous replication timing that is coordinated with other mono-allelically expressed genes on chromosome 6. Characterization of this locus revealed mono-allelic expression of a large intergenic non-coding RNA, which we have named asynchronous replication and autosomal RNA on chromosome 6, ASAR6. Finally, disruption of this locus results in the activation of the previously silent alleles of linked mono-allelically expressed genes. We previously found that chromosome rearrangements involving eight different autosomes display delayed replication timing, and that cells containing chromosomes with delayed replication timing have a 30-80-fold increase in the rate at which new gross chromosomal rearrangements occurred. Taken together, these observations indicate that human autosomes contain discrete cis-acting loci that control chromosome-wide replication timing, mono-allelic expression and the stability of entire chromosomes.


Asunto(s)
Inestabilidad Cromosómica/genética , Cromosomas Humanos Par 6/genética , Replicación del ADN/fisiología , Regulación de la Expresión Génica/genética , Sitios Genéticos/genética , Secuencia de Bases , Bromodesoxiuridina , Línea Celular , Replicación del ADN/genética , ADN Intergénico/genética , Fucosiltransferasas/metabolismo , Humanos , Hibridación Fluorescente in Situ , Proteínas con Dominio LIM , Datos de Secuencia Molecular , ARN no Traducido/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Factores de Tiempo , Factores de Transcripción/metabolismo
4.
Genetics ; 220(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34849849

RESUMEN

The absence of functional BLM DNA helicase, a member of the RecQ family of helicases, is responsible for the rare human disorder Bloom Syndrome, which results in developmental abnormalities, DNA repair defects, genomic instability, and a predisposition to cancer. In Drosophila melanogaster, the orthologous Blm protein is essential during early development when the embryo is under the control of maternal gene products. We show that lack of functional maternal Blm during the syncytial cell cycles of Drosophila embryonic development results in severe nuclear defects and lethality. Amongst the small fraction of embryos from Blm mutant mothers that survive to adulthood, a prominent sex-bias favors the class that inherits less repetitive DNA content, which serves as an endogenous source of replication stress. This selection against repetitive DNA content reflects a role for Blm in facilitating replication through repetitive sequences during the rapid S-phases of syncytial cell cycles. During these syncytial cycles, Blm is not required for complex DNA double-strand break repair; however, the progeny sex-bias resulting from the absence of maternal Blm is exacerbated by repetitive DNA sequences and by the slowing of replication fork progression, suggesting that the essential role for Blm during this stage is to manage replication fork stress brought about by impediments to fork progression. Additionally, our data suggest that Blm is only required to manage this replication stress during embryonic development, and likely only during the early, rapid syncytial cell cycles, and not at later developmental stages. These results provide novel insights into Blm function throughout development.


Asunto(s)
RecQ Helicasas
5.
Blood ; 111(9): 4788-96, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18252861

RESUMEN

To determine whether aberrantly activated tyrosine kinases other than FLT3 and c-KIT contribute to acute myeloid leukemia (AML) pathogenesis, we used high-throughput (HT) DNA sequence ana-lysis to screen exons encoding the activation loop and juxtamembrane domains of 85 tyrosine kinase genes in 188 AML patients without FLT3 or c-KIT mutations. The screen identified 30 nonsynonymous sequence variations in 22 different kinases not previously reported in single-nucleotide polymorphism (SNP) databases. These included a novel FLT3 activating allele and a previously described activating mutation in MET (METT1010I). The majority of novel sequence variants were stably expressed in factor-dependent Ba/F3 cells. Apart from one FLT3 allele, none of the novel variants showed constitutive phosphorylation by immunoblot analysis and none transformed Ba/F3 cells to factor-independent growth. These findings indicate the majority of these alleles are not potent tyrosine kinase activators in this cellular context and that a significant proportion of nonsynonymous sequence variants identified in HT DNA sequencing screens may not have functional significance. Although some sequence variants may represent SNPs, these data are consistent with recent reports that a significant fraction of such sequence variants are "passenger" rather than "driver" alleles and underscore the importance of functional assessment of candidate disease alleles.


Asunto(s)
Leucemia Mieloide Aguda/enzimología , Polimorfismo de Nucleótido Simple , Proteínas Tirosina Quinasas/genética , Secuencia de Bases , Análisis Mutacional de ADN , Humanos , Leucemia Mieloide Aguda/etiología , Tirosina Quinasa 3 Similar a fms
6.
Mol Cell Biol ; 26(16): 6082-93, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16880519

RESUMEN

Kinase domain (KD) mutations of Bcr-Abl interfering with imatinib binding are the major mechanism of acquired imatinib resistance in patients with Philadelphia chromosome-positive leukemia. Mutations of the ATP binding loop (p-loop) have been associated with a poor prognosis. We compared the transformation potency of five common KD mutants in various biological assays. Relative to unmutated (native) Bcr-Abl, the ATP binding loop mutants Y253F and E255K exhibited increased transformation potency, M351T and H396P were less potent, and the performance of T315I was assay dependent. The transformation potency of Y253F and M351T correlated with intrinsic Bcr-Abl kinase activity, whereas the kinase activity of E255K, H396P, and T315I did not correlate with transforming capabilities, suggesting that additional factors influence transformation potency. Analysis of the phosphotyrosine proteome by mass spectroscopy showed differential phosphorylation among the mutants, a finding consistent with altered substrate specificity and pathway activation. Mutations in the KD of Bcr-Abl influence kinase activity and signaling in a complex fashion, leading to gain- or loss-of-function variants. The drug resistance and transformation potency of mutants may determine the outcome of patients on therapy with Abl kinase inhibitors.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Proteínas de Fusión bcr-abl/metabolismo , Mutación/genética , Fosfotransferasas/metabolismo , Piperazinas/farmacología , Pirimidinas/farmacología , Secuencia de Aminoácidos , Animales , Benzamidas , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Modelos Animales de Enfermedad , Femenino , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/genética , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Células Progenitoras Mieloides/citología , Fosfotirosina/metabolismo , Estructura Terciaria de Proteína , Transducción de Señal , Especificidad por Sustrato
7.
Cancer Res ; 65(11): 4500-5, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15930265

RESUMEN

Imatinib, a Bcr-Abl tyrosine kinase inhibitor, is a highly effective therapy for patients with chronic myelogenous leukemia (CML). Despite durable responses in most chronic phase patients, relapses have been observed and are much more prevalent in patients with advanced disease. The most common mechanism of acquired imatinib resistance has been traced to Bcr-Abl kinase domain mutations with decreased imatinib sensitivity. Thus, alternate Bcr-Abl kinase inhibitors that have activity against imatinib-resistant mutants would be useful for patients who relapse on imatinib therapy. Two such Bcr-Abl inhibitors are currently being evaluated in clinical trials: the improved potency, selective Abl inhibitor AMN107 and the highly potent dual Src/Abl inhibitor BMS-354825. In the current article, we compared imatinib, AMN107, and BMS-354825 in cellular and biochemical assays against a panel of 16 kinase domain mutants representing >90% of clinical isolates. We report that AMN107 and BMS-354825 are 20-fold and 325-fold more potent than imatinib against cells expressing wild-type Bcr-Abl and that similar improvements are maintained for all imatinib-resistant mutants tested, with the exception of T315I. Thus, both inhibitors hold promise for treating imatinib-refractory CML.


Asunto(s)
Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Tiazoles/farmacología , Animales , Antineoplásicos/farmacología , Benzamidas , Línea Celular , Dasatinib , Proteínas de Fusión bcr-abl , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Ratones , Modelos Moleculares , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/genética
8.
Leuk Res ; 30(9): 1097-104, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16464493

RESUMEN

STAT5 is constitutively phosphorylated in leukemic cells in approximately 70% of acute myeloid leukemia (AML) patients. To identify kinase candidates potentially responsible for STAT5 phosphorylation, we used liquid chromatography-tandem mass spectrometry (LC-MS/MS) mass spectrometry to detect phosphoproteins in AML cell lines. We established TEL-ARG and BCR-ABL fusion proteins as the mechanism underlying STAT5 phosphorylation in HT-93 and KBM-3 cells, respectively. In addition, we identified a JAK2 pseudokinase domain mutation in HEL cells and using siRNA downregulation, established JAK2 as the kinase responsible for phosphorylating STAT5. This study illustrates the benefit of LC-MS/MS mass spectrometry and siRNA for the identification of novel targets and mutations.


Asunto(s)
Carcinógenos , Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/genética , Carcinógenos/metabolismo , Línea Celular Tumoral , Cromatografía Liquida , Humanos , Leucemia Mieloide Aguda/metabolismo , Espectrometría de Masas , Mutación , Proteínas de Neoplasias/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional/genética , Proteómica
9.
Clin Cancer Res ; 11(19 Pt 1): 6987-93, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16203792

RESUMEN

PURPOSE: Chronic myeloid leukemia (CML) is effectively treated with imatinib. However, reactivation of Bcr-Abl via kinase domain mutations that reduce sensitivity to imatinib can cause relapse. As combination therapy is frequently used to prevent emergence of resistance, the combination of imatinib with an inhibitor of imatinib-resistant Bcr-Abl mutants (e.g., Src/Abl inhibitors AP23848 and BMS-354825) was investigated. EXPERIMENTAL DESIGN: To test this approach, cellular proliferation and Bcr-Abl tyrosine phosphorylation assays were done on Ba/F3 cells expressing wild-type (WT) Bcr-Abl and four common imatinib-resistant mutants (Y253F, E255K, T315I, and M351T). Colony-forming assays with primary CML cells were also done. RESULTS: Both Src/Abl inhibitors retained full inhibitory capacity when coadministered with imatinib at concentrations above typical clinical levels. For cells expressing WT Bcr-Abl or the marginally imatinib-resistant mutant M351T, inclusion of imatinib at therapeutic levels enhanced the effects of the Src/Abl inhibitors. By comparison, for the highly imatinib-resistant mutants Y253F and E255K, inclusion of imatinib at clinical levels resulted in only a slight enhancement beyond the effects of the Src/Abl inhibitors. None of the inhibitors affected Bcr-Abl T315I cells. Colony-forming assays with primary CML cells yielded analogous results. CONCLUSIONS: Our results indicate that Src/Abl inhibitors are compatible with imatinib and suggest that combined Abl inhibitor therapy is a feasible treatment strategy for patients with CML.


Asunto(s)
Resistencia a Antineoplásicos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-abl/metabolismo , Pirimidinas/farmacología , Familia-src Quinasas/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Antineoplásicos/farmacología , Benzamidas , Proliferación Celular , Dasatinib , Relación Dosis-Respuesta a Droga , Vectores Genéticos , Células Madre Hematopoyéticas/citología , Humanos , Mesilato de Imatinib , Immunoblotting , Concentración 50 Inhibidora , Ratones , Mutación , Fosfotirosina/química , Mutación Puntual , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Células Madre/metabolismo , Tiazoles/farmacología , Factores de Tiempo
10.
Cancer Res ; 62(24): 7149-53, 2002 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12499247

RESUMEN

Imatinib mesylate, a selective inhibitor of the Abl tyrosine kinase, is effective as a single-agent therapy for chronic myelogenous leukemia. However, resistance has been reported, particularly in patients with advanced-stage disease. Mutations within the Abl kinase domain are a major cause of resistance, demonstrating that Bcr-Abl remains a critical drug target. Recently, a novel pyrido[2,3-d]pyrimidine derivative, PD180970, has been shown to potently inhibit Bcr-Abl and induce apoptosis in Bcr-Abl-expressing leukemic cells. We analyzed the inhibitory activity of PD180970 against Abl kinase domain mutations and cells expressing clinically relevant mutations. Our data indicate that PD180970 is active against several Bcr-Abl mutations that are resistant to imatinib and support the notion that developing additional Abl kinase inhibitors would be useful as a treatment strategy for chronic myelogenous leukemia.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Piperazinas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Piridonas/farmacología , Pirimidinas/farmacología , Animales , Benzamidas , Línea Celular , Resistencia a Antineoplásicos/genética , Proteínas de Fusión bcr-abl , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/fisiología , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Ratones , Mutagénesis Sitio-Dirigida , Isoformas de Proteínas , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/genética , Transfección
11.
Hematol J ; 4(6): 413-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14671613

RESUMEN

Imatinib mesylate (Gleevec, formerly STI571) has been shown to be a safe and effective treatment for chronic myelogenous leukemia (CML). However, despite high rates of hematologic and cytogenetic remissions, molecular remissions are rare. Recent work has revealed the existence of a population of Bcr-Abl-positive, quiescent hematopoietic CML stem cells that are insensitive to induction of apoptosis by imatinib ex vivo. Thus, quiescence is postulated as a mechanism of molecular resistance to imatinib. To model a cell population with reduced cell cycle activity in vitro, we applied three different established approaches to block the cell cycle at the G1/S boundary using Bcr-Abl-positive cell lines. Subsequently, the cells were exposed to imatinib and apoptosis after 48 h of treatment was determined by analysis of activated caspase-3 and apoptotic DNA strand breaks. In these models, reduced cell cycle activity did not have a significant impact on the ability of imatinib to induce apoptosis. These data suggest that the proapoptotic activity of imatinib in vitro is not dependent on cell cycle transit. We conclude that resistance of primary CML cells that are insensitive to imatinib may be the result of molecular properties causing drug resistance rather than a consequence of quiescence itself.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Fusión bcr-abl/análisis , Piperazinas/farmacología , Pirimidinas/farmacología , Antineoplásicos/farmacología , Benzamidas , Crisis Blástica/genética , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Mesilato de Imatinib , Células K562 , Cinética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Tumorales Cultivadas
12.
Genetics ; 196(1): 107-18, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24172129

RESUMEN

The Bloom syndrome helicase, BLM, has numerous functions that prevent mitotic crossovers. We used unique features of Drosophila melanogaster to investigate origins and properties of mitotic crossovers that occur when BLM is absent. Induction of lesions that block replication forks increased crossover frequencies, consistent with functions for BLM in responding to fork blockage. In contrast, treatment with hydroxyurea, which stalls forks, did not elevate crossovers, even though mutants lacking BLM are sensitive to killing by this agent. To learn about sources of spontaneous recombination, we mapped mitotic crossovers in mutants lacking BLM. In the male germline, irradiation-induced crossovers were distributed randomly across the euchromatin, but spontaneous crossovers were nonrandom. We suggest that regions of the genome with a high frequency of mitotic crossovers may be analogous to common fragile sites in the human genome. Interestingly, in the male germline there is a paucity of crossovers in the interval that spans the pericentric heterochromatin, but in the female germline this interval is more prone to crossing over. Finally, our system allowed us to recover pairs of reciprocal crossover chromosomes. Sequencing of these revealed the existence of gene conversion tracts and did not provide any evidence for mutations associated with crossovers. These findings provide important new insights into sources and structures of mitotic crossovers and functions of BLM helicase.


Asunto(s)
Intercambio Genético/genética , Reparación del ADN por Unión de Extremidades/genética , ADN Helicasas/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Animales , Secuencia de Bases , Intercambio Genético/efectos de los fármacos , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Femenino , Hidroxiurea/farmacología , Masculino , Mitosis/genética , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Análisis de Secuencia de ADN
13.
Blood ; 105(7): 2952-4, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15585651

RESUMEN

FMS-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase that is constitutively activated in approximately 30% of acute myelogenous leukemia (AML) patients and appears to confer an adverse prognosis. Thus, development of inhibitors and/or antibodies that specifically target FLT3 has been of substantial interest. In this regard, phase 1 and 2 trials involving FLT3 inhibitors have recently reported FLT3 inhibition and leukemic blast reduction in some patients. Despite this, issues such as specificity and resistance need to be addressed. Consequently, the development of alternative approaches for targeting FLT3 would be of great consequence. In the present report, we demonstrate that FLT3 siRNA effectively down-regulates FLT3 expression in Ba/F3 cells transfected with FLT3 containing an activating internal tandem duplication (ITD) in the juxtamembrane domain and FLT3-ITD-positive Molm-14 human leukemia cells. Treatment with the FLT3 siRNA results in growth inhibition and apoptosis of these cells. Furthermore, siRNA-induced down-regulation of FLT3 increased the sensitivity of both cell lines to treatment with the FLT3 inhibitor MLN518. This illustrates the potential benefit of combined therapeutic approaches.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Quinazolinas/farmacología , ARN Interferente Pequeño/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Línea Celular Tumoral , Regulación hacia Abajo , Resistencia a Antineoplásicos , Humanos , Leucemia Mieloide Aguda/fisiopatología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Transfección , Tirosina Quinasa 3 Similar a fms
14.
Blood ; 106(6): 2128-37, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15914554

RESUMEN

Mutations in the kinase domain (KD) of BCR-ABL are the leading cause of acquired imatinib resistance. In some cases, identical mutations were detected at relapse and in pretherapeutic specimens, consistent with selection of resistant clones in the presence of drug. However, the incidence of KD mutations in imatinibnaive patients, irrespective of response to therapy, is unknown. We studied mutation frequency in 66 patients with chronic myelogenous leukemia (CML), using cDNA sequencing and allele-specific oligonucleotide-polymerase chain reaction (ASO-PCR) assays for 8 common mutations. Thirteen patients were positive by ASO-PCR only, 1 by ASO-PCR and sequencing, and 1 by sequencing only (overall frequency, 22.7%). T315I was most frequent (12% of patients). Eleven of the 14 patients with positive ASO-PCR had follow-up samples available for sequencing. Wild-type sequence was detected in 6 of 11, 2 different mutations in 1 of 11, and identical mutations in 4 of 11 patients, 2 of whom had achieved major cytogenetic response. In multivariate analysis mutation detection was associated with clonal cytogenetic evolution, exposure to 6-Thioguanine, and a low platelet count, but not with response to imatinib, event-free survival, and overall survival. KD mutants present at low levels do not invariably lead to relapse, and additional factors are required to induce a fully drug-resistant phenotype.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas de Fusión bcr-abl/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Mutación , Piperazinas , Pirimidinas , Adulto , Anciano , Anciano de 80 o más Años , Benzamidas , Células Clonales , Análisis Mutacional de ADN , Femenino , Frecuencia de los Genes , Humanos , Mesilato de Imatinib , Cariotipificación , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/mortalidad , Masculino , Persona de Mediana Edad , Análisis Multivariante , Proteínas Tirosina Quinasas/genética , Factores de Riesgo , Sensibilidad y Especificidad , Resultado del Tratamiento
15.
Blood ; 101(11): 4611-4, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12576318

RESUMEN

Imatinib mesylate is a selective Bcr-Abl kinase inhibitor, effective in the treatment of chronic myelogenous leukemia. Most patients in chronic phase maintain durable responses; however, many in blast crisis fail to respond, or relapse quickly. Kinase domain mutations are the most commonly identified mechanism associated with relapse. Many of these mutations decrease the sensitivity of the Abl kinase to imatinib, thus accounting for resistance to imatinib. The role of other mutations in the emergence of resistance has not been established. Using biochemical and cellular assays, we analyzed the sensitivity of several mutants (Met244Val, Phe311Leu, Phe317Leu, Glu355Gly, Phe359Val, Val379Ile, Leu387Met, and His396Pro/Arg) to imatinib mesylate to better understand their role in mediating resistance. While some Abl mutations lead to imatinib resistance, many others are significantly, and some fully, inhibited. This study highlights the need for biochemical and biologic characterization, before a resistant phenotype can be ascribed to a mutant.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteínas de Fusión bcr-abl/genética , Mutación Missense , Piperazinas/farmacología , Pirimidinas/farmacología , Animales , Benzamidas , Western Blotting , División Celular/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Proteínas de Fusión bcr-abl/efectos de los fármacos , Humanos , Mesilato de Imatinib , Ratones , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , Recurrencia , Transfección
16.
Blood ; 103(1): 208-15, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12933582

RESUMEN

Imatinib mesylate (Gleevec, formerly STI571) is an effective therapy for all stages of chronic myelogenous leukemia (CML). While responses in chronic-phase CML are generally durable, resistance develops in many patients with advanced disease. We evaluated novel antileukemic agents for their potential to overcome resistance in various imatinib-resistant cell lines. Using cell proliferation assays, we investigated whether different mechanisms of resistance to imatinib would alter the efficacy of arsenic trioxide (As2O3) or 5-aza-2-deoxycytidine (decitabine) alone and in combination with imatinib. Our results indicate that resistance to imatinib induced by Bcr-Abl overexpression or by engineered expression of clinically relevant Bcr-Abl mutants does not induce cross-resistance to As2O3 or decitabine. Combined treatment with these agents and imatinib is beneficial in cell lines that have residual sensitivity to imatinib monotherapy, with synergistic growth inhibition achieved only at doses of imatinib that overcome resistance. In some imatinib-resistant cell lines, combination treatments that use low doses of imatinib lead to antagonism. Apoptosis studies suggest that this can be explained in part by the reduced proapoptotic activity of imatinib in resistant cell lines. These data underline the importance of resistance testing and provide a rational approach for dose-adjusted administration of imatinib when combined with other agents.


Asunto(s)
Azacitidina/análogos & derivados , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Piperazinas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Pirimidinas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Trióxido de Arsénico , Arsenicales/administración & dosificación , Azacitidina/administración & dosificación , Secuencia de Bases , Benzamidas , División Celular/efectos de los fármacos , Línea Celular Tumoral , ADN de Neoplasias/genética , Decitabina , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Proteínas de Fusión bcr-abl , Genes abl , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Óxidos/administración & dosificación , Piperazinas/administración & dosificación , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/administración & dosificación
17.
Blood ; 104(8): 2532-9, 2004 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-15256422

RESUMEN

The deregulated, oncogenic tyrosine kinase Bcr-Abl causes chronic myeloid leukemia (CML). Imatinib mesylate (Gleevec, STI571), a Bcr-Abl kinase inhibitor, selectively inhibits proliferation and promotes apoptosis of CML cells. Despite the success of imatinib mesylate in the treatment of CML, resistance is observed, particularly in advanced disease. The most common imatinib mesylate resistance mechanism involves Bcr-Abl kinase domain mutations that impart varying degrees of drug insensitivity. AP23464, a potent adenosine 5'-triphosphate (ATP)-based inhibitor of Src and Abl kinases, displays antiproliferative activity against a human CML cell line and Bcr-Abl-transduced Ba/F3 cells (IC(50) = 14 nM; imatinib mesylate IC(50) = 350 nM). AP23464 ablates Bcr-Abl tyrosine phosphorylation, blocks cell cycle progression, and promotes apoptosis of Bcr-Abl-expressing cells. Biochemical assays with purified glutathione S transferase (GST)-Abl kinase domain confirmed that AP23464 directly inhibits Abl activity. Importantly, the low nanomolar cellular and biochemical inhibitory properties of AP23464 extend to frequently observed imatinib mesylate-resistant Bcr-Abl mutants, including nucleotide binding P-loop mutants Q252H, Y253F, E255K, C-terminal loop mutant M351T, and activation loop mutant H396P. AP23464 was ineffective against mutant T315I, an imatinib mesylate contact residue. The potency of AP23464 against imatinib mesylate-refractory Bcr-Abl and its distinct binding mode relative to imatinib mesylate warrant further investigation of AP23464 for the treatment of CML.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/genética , Mutación/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfato/química , Aminoácidos/genética , Aminoácidos/metabolismo , Apoptosis/efectos de los fármacos , Benzamidas , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/química , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HL-60 , Humanos , Mesilato de Imatinib , Concentración 50 Inhibidora , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas de la Leche/metabolismo , Modelos Moleculares , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Piperazinas/química , Piperazinas/farmacología , Estructura Terciaria de Proteína , Piridonas/química , Piridonas/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Factor de Transcripción STAT5 , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA