Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Blood ; 125(23): 3588-97, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25814533

RESUMEN

Usp9x was recently shown to be highly expressed in myeloma patients with short progression-free survival and is proposed to enhance stability of the survival protein Mcl-1. In this study, we found that the partially selective Usp9x deubiquitinase inhibitor WP1130 induced apoptosis and reduced Mcl-1 protein levels. However, short hairpin RNA-mediated knockdown (KD) of Usp9x in myeloma cells resulted in transient induction of apoptosis, followed by a sustained reduction in cell growth. A compensatory upregulation of Usp24, a deubiquitinase closely related to Usp9x, in Usp9x KD cells was noted. Direct Usp24 KD resulted in marked induction of myeloma cell death that was associated with a reduction of Mcl-1. Usp24 was found to sustain myeloma cell survival and Mcl-1 regulation in the absence of Usp9x. Both Usp9x and Usp24 were expressed and activated in primary myeloma cells whereas Usp24 protein overexpression was noted in some patients with drug-refractory myeloma and other B-cell malignancies. Furthermore, we improved the drug-like properties of WP1130 and demonstrated that the novel compound EOAI3402143 dose-dependently inhibited Usp9x and Usp24 activity, increased tumor cell apoptosis, and fully blocked or regressed myeloma tumors in mice. We conclude that small-molecule Usp9x/Usp24 inhibitors may have therapeutic activity in myeloma.


Asunto(s)
Apoptosis/efectos de los fármacos , Cianoacrilatos/farmacología , Inhibidores Enzimáticos/farmacología , Linfoma de Células del Manto/tratamiento farmacológico , Mieloma Múltiple/tratamiento farmacológico , Piridinas/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Animales , Apoptosis/genética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Linfoma de Células del Manto/enzimología , Linfoma de Células del Manto/genética , Linfoma de Células del Manto/patología , Masculino , Ratones , Mieloma Múltiple/enzimología , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
2.
PLoS Pathog ; 8(7): e1002783, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792064

RESUMEN

Ubiquitin (Ub) is a vital regulatory component in various cellular processes, including cellular responses to viral infection. As obligate intracellular pathogens, viruses have the capacity to manipulate the ubiquitin (Ub) cycle to their advantage by encoding Ub-modifying proteins including deubiquitinases (DUBs). However, how cellular DUBs modulate specific viral infections, such as norovirus, is poorly understood. To examine the role of DUBs during norovirus infection, we used WP1130, a small molecule inhibitor of a subset of cellular DUBs. Replication of murine norovirus in murine macrophages and the human norovirus Norwalk virus in a replicon system were significantly inhibited by WP1130. Chemical proteomics identified the cellular DUB USP14 as a target of WP1130 in murine macrophages, and pharmacologic inhibition or siRNA-mediated knockdown of USP14 inhibited murine norovirus infection. USP14 is a proteasome-associated DUB that also binds to inositol-requiring enzyme 1 (IRE1), a critical mediator of the unfolded protein response (UPR). WP1130 treatment of murine macrophages did not alter proteasome activity but activated the X-box binding protein-1 (XBP-1) through an IRE1-dependent mechanism. In addition, WP1130 treatment or induction of the UPR also reduced infection of other RNA viruses including encephalomyocarditis virus, Sindbis virus, and La Crosse virus but not vesicular stomatitis virus. Pharmacologic inhibition of the IRE1 endonuclease activity partially rescued the antiviral effect of WP1130. Taken together, our studies support a model whereby induction of the UPR through cellular DUB inhibition blocks specific viral infections, and suggest that cellular DUBs and the UPR represent novel targets for future development of broad spectrum antiviral therapies.


Asunto(s)
Antivirales/farmacología , Nitrilos/farmacología , Norovirus/efectos de los fármacos , Piridinas/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Infecciones por Caliciviridae/tratamiento farmacológico , Infecciones por Caliciviridae/virología , Línea Celular , Línea Celular Tumoral , Cianoacrilatos , Proteínas de Unión al ADN/metabolismo , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/patogenicidad , Inhibidores Enzimáticos/farmacología , Humanos , Virus La Crosse/efectos de los fármacos , Virus La Crosse/patogenicidad , Macrófagos/virología , Proteínas de la Membrana/metabolismo , Ratones , Norovirus/fisiología , Virus Norwalk/efectos de los fármacos , Virus Norwalk/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Factores de Transcripción del Factor Regulador X , Virus Sindbis/efectos de los fármacos , Virus Sindbis/patogenicidad , Factores de Transcripción/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/genética , Replicación Viral/efectos de los fármacos , Proteína 1 de Unión a la X-Box
3.
Bioorg Med Chem ; 22(4): 1450-8, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24457091

RESUMEN

A series of degrasyn-like symmetrical compounds have been designed, synthesized, and screened against B cell malignancy (multiple myeloma, mantle cell lymphoma) cell lines. The lead compounds T5165804 and CP2005 showed higher nanomolar potency against these tumor cells in comparison to degrasyn and inhibited Usp9x activity in vitro and in intact cells. These observations suggest that this new class of compounds holds promise as cancer therapeutic agents.


Asunto(s)
Antineoplásicos/química , Nitrilos/química , Piridinas/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cianoacrilatos , Dimerización , Humanos , Modelos Moleculares , Mieloma Múltiple/tratamiento farmacológico , Nitrilos/farmacología , Nitrilos/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo
4.
Blood ; 117(11): 3151-62, 2011 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-21248063

RESUMEN

Although chronic myelogenous leukemia (CML) is effectively controlled by Bcr-Abl kinase inhibitors, resistance to inhibitors, progressive disease, and incomplete eradication of Bcr-Abl-expressing cells are concerns for the long-term control and suppression of this disease. We describe a novel approach to targeting key proteins in CML cells with a ubiquitin-cycle inhibitor, WP1130. Bcr-Abl is rapidly modified with K63-linked ubiquitin polymers in WP1130-treated CML cells, resulting in its accumulation in aggresomes, where is it unable to conduct signal transduction. Induction of apoptosis because of aggresomal compartmentalization of Bcr-Abl was observed in both imatinib-sensitive and -resistant cells. WP1130, but not Bcr-Abl kinase inhibitors, directly inhibits Usp9x deubiquitinase activity, resulting in the down-regulation of the prosurvival protein Mcl-1 and facilitating apoptosis. These results demonstrate that ubiquitin-cycle inhibition represents a novel and effective approach to blocking Bcr-Abl kinase signaling and reducing Mcl-1 levels to engage CML cell apoptosis. This approach may be a therapeutic option for kinase inhibitor-resistant CML patients.


Asunto(s)
Apoptosis , Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Transducción de Señal , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitinación , Apoptosis/efectos de los fármacos , Benzamidas , Línea Celular Tumoral , Cianoacrilatos , Resistencia a Antineoplásicos/efectos de los fármacos , Endopeptidasas/metabolismo , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Humanos , Mesilato de Imatinib , Modelos Biológicos , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Transporte de Proteínas/efectos de los fármacos , Piridinas/farmacología , Pirimidinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato/efectos de los fármacos , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación/efectos de los fármacos
5.
Infect Immun ; 79(12): 4850-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21911458

RESUMEN

Macrophages are key mediators of antimicrobial defense and innate immunity. Innate intracellular defense mechanisms can be rapidly regulated at the posttranslational level by the coordinated addition and removal of ubiquitin by ubiquitin ligases and deubiquitinases (DUBs). While ubiquitin ligases have been extensively studied, the contribution of DUBs to macrophage innate immune function is incompletely defined. We therefore employed a small molecule DUB inhibitor, WP1130, to probe the role of DUBs in the macrophage response to bacterial infection. Treatment of activated bone marrow-derived macrophages (BMM) with WP1130 significantly augmented killing of the intracellular bacterial pathogen Listeria monocytogenes. WP1130 also induced killing of phagosome-restricted bacteria, implicating a bactericidal mechanism associated with the phagosome, such as the inducible nitric oxide synthase (iNOS). WP1130 had a minimal antimicrobial effect in macrophages lacking iNOS, indicating that iNOS is an effector mechanism for WP1130-mediated bacterial killing. Although overall iNOS levels were not notably different, we found that WP1130 significantly increased colocalization of iNOS with the Listeria-containing phagosome during infection. Taken together, our data indicate that the deubiquitinase inhibitor WP1130 increases bacterial killing in macrophages by enhancing iNOS localization to the phagosome and suggest a potential role for ubiquitin regulation in iNOS trafficking.


Asunto(s)
Macrófagos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nitrilos/farmacología , Fagosomas/enzimología , Inhibidores de Proteasas/farmacología , Piridinas/farmacología , Animales , Línea Celular , Cianoacrilatos , Endopeptidasas/metabolismo , Escherichia coli/fisiología , Listeria monocytogenes/fisiología , Macrófagos/enzimología , Ratones , Óxido Nítrico Sintasa de Tipo II/genética , Transporte de Proteínas/efectos de los fármacos , Ubiquitina/metabolismo
6.
Cancer Res ; 67(8): 3912-8, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17440106

RESUMEN

c-Myc is a highly unstable transcription factor whose deregulation and increased expression are associated with cancer. Degrasyn, a small synthetic molecule, induces rapid degradation of c-Myc protein in MM-1 multiple myeloma and other tumor cell lines. Destruction of c-Myc by degrasyn requires the presence of a region of c-Myc between amino acid residues 316 and 378 that has not previously been associated with c-Myc stability. Degrasyn-induced degradation of c-Myc depends on proteasomes but is independent of the degron regions previously shown to be important for ubiquitin-mediated targeting and proteasomal destruction of the protein. Degrasyn-dependent c-Myc proteolysis is not mediated by any previously identified c-Myc regulatory mechanism, does not require new protein synthesis, and does not depend on the nuclear localization of c-Myc. Degrasyn reduced c-Myc levels in A375 melanoma cells and in A375 tumors in nude mice, and this activity correlated with tumor growth inhibition. Together, these results suggest that degrasyn reduces the stability of c-Myc in vitro and in vivo through a unique signaling process that uses c-Myc domains not previously associated with c-Myc regulation.


Asunto(s)
Melanoma/tratamiento farmacológico , Nitrilos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Piridinas/farmacología , Animales , Línea Celular Tumoral , Cianoacrilatos , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Células HeLa , Humanos , Melanoma/enzimología , Melanoma/metabolismo , Ratones , Ratones Desnudos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/enzimología , Mieloma Múltiple/metabolismo , Mapeo Peptídico , Estructura Terciaria de Proteína , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Clin Cancer Res ; 13(10): 3006-14, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17505003

RESUMEN

PURPOSE: To determine the tolerability and pharmacokinetics of CI-1033 given daily for 7 days of a 21-day cycle. Tumor response and changes in erbB receptor tyrosine kinase activity in tumor and skin tissue were examined, and modulation of potential biomarkers in plasma was explored. DESIGN: This was a dose-finding phase I study in patients with advanced solid malignancies. Patients were evaluated for safety, pharmacokinetics, and tumor response. Pharmacodynamic markers, such as Ki67, p27, and erbB receptor status, were assessed in tumor and skin tissue using immunohistochemical and immunoprecipitation methodologies. Plasma biomarkers HER2, vascular endothelial growth factor, interleukin-8, and matrix metalloproteinase-9 were evaluated using immunologic techniques. RESULTS: Fifty-three patients were enrolled in the study. Dose-limiting toxicity (emesis, persistent rash, and mouth ulcer) was observed at 750 mg. The maximum tolerated dose was 650 mg. There were no confirmed objective responses. CI-1033 treatment showed down-regulation of epidermal growth factor receptor, HER2, and Ki67 in a variety of tumor tissues and up regulation of p27 in skin tissue. Plasma HER2 was reduced following CI-1033 administration, but no consistent change in vascular endothelial growth factor, interleukin-8, or matrix metalloproteinase-9 was noted. CI-1033 plasma concentrations were proportional to dose. CONCLUSION: The safety and pharmacokinetic profile of CI-1033 was favorable for multidose oral administration. Evidence of modulation of erbB receptor activity in tumor and skin tissue was accompanied by changes in markers of proliferation and cell cycle inhibition. Additional clinical trials are warranted in defining the role of CI-1033 in the treatment of cancer and further assessing the utility of antitumor markers.


Asunto(s)
Antineoplásicos/administración & dosificación , Morfolinas/administración & dosificación , Neoplasias/tratamiento farmacológico , Administración Oral , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Biomarcadores de Tumor/análisis , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad , Morfolinas/efectos adversos , Morfolinas/farmacocinética
8.
PLoS One ; 9(8): e104096, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25093325

RESUMEN

The global spread of anti-microbial resistance requires urgent attention, and diverse alternative strategies have been suggested to address this public health concern. Host-directed immunomodulatory therapies represent one approach that could reduce selection for resistant bacterial strains. Recently, the small molecule deubiquitinase inhibitor WP1130 was reported as a potential anti-infective drug against important human food-borne pathogens, notably Listeria monocytogenes and noroviruses. Utilization of WP1130 itself is limited due to poor solubility, but given the potential of this new compound, we initiated an iterative rational design approach to synthesize new derivatives with increased solubility that retained anti-infective activity. Here, we test a small library of novel synthetic molecules based on the structure of the parent compound, WP1130, for anti-infective activity in vitro. Our studies identify a promising candidate, compound 9, which reduced intracellular growth of L. monocytogenes at concentrations that caused minimal cellular toxicity. Compound 9 itself had no bactericidal activity and only modestly slowed Listeria growth rate in liquid broth culture, suggesting that this drug acts as an anti-infective compound by modulating host-cell function. Moreover, this new compound also showed anti-infective activity against murine norovirus (MNV-1) and human norovirus, using the Norwalk virus replicon system. This small molecule inhibitor may provide a chemical platform for further development of therapeutic deubiquitinase inhibitors with broad-spectrum anti-infective activity.


Asunto(s)
Antiinfecciosos/farmacología , Macrófagos/microbiología , Inhibidores de Proteasas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Animales , Antivirales/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular , Cianoacrilatos/química , Cianoacrilatos/farmacología , Humanos , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/crecimiento & desarrollo , Macrófagos/efectos de los fármacos , Ratones , Viabilidad Microbiana/efectos de los fármacos , Peso Molecular , Norovirus/efectos de los fármacos , Piridinas/química , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/química , Proteasas Ubiquitina-Específicas/metabolismo
9.
PLoS One ; 9(4): e94491, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24722666

RESUMEN

Most antiviral treatment options target the invading pathogen and unavoidably encounter loss of efficacy as the pathogen mutates to overcome replication restrictions. A good strategy for circumventing drug resistance, or for pathogens without treatment options, is to target host cell proteins that are utilized by viruses during infection. The small molecule WP1130 is a selective deubiquitinase inhibitor shown previously to successfully reduce replication of noroviruses and some other RNA viruses. In this study, we screened a library of 31 small molecule derivatives of WP1130 to identify compounds that retained the broad-spectrum antiviral activity of the parent compound in vitro but exhibited improved drug-like properties, particularly increased aqueous solubility. Seventeen compounds significantly reduced murine norovirus infection in murine macrophage RAW 264.7 cells, with four causing decreases in viral titers that were similar or slightly better than WP1130 (1.9 to 2.6 log scale). Antiviral activity was observed following pre-treatment and up to 1 hour postinfection in RAW 264.7 cells as well as in primary bone marrow-derived macrophages. Treatment of the human norovirus replicon system cell line with the same four compounds also decreased levels of Norwalk virus RNA. No significant cytotoxicity was observed at the working concentration of 5 µM for all compounds tested. In addition, the WP1130 derivatives maintained their broad-spectrum antiviral activity against other RNA viruses, Sindbis virus, LaCrosse virus, encephalomyocarditis virus, and Tulane virus. Thus, altering structural characteristics of WP1130 can maintain effective broad-spectrum antiviral activity while increasing aqueous solubility.


Asunto(s)
Antivirales/farmacología , Inhibidores Enzimáticos/farmacología , Nitrilos/farmacología , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteasas Ubiquitina-Específicas/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Animales , Antivirales/química , Línea Celular , Cianoacrilatos , Virus de la Encefalomiocarditis/efectos de los fármacos , Virus de la Encefalomiocarditis/fisiología , Inhibidores Enzimáticos/química , Interacciones Huésped-Patógeno , Humanos , Virus La Crosse/efectos de los fármacos , Virus La Crosse/fisiología , Macrófagos/efectos de los fármacos , Macrófagos/virología , Ratones , Nitrilos/química , Norovirus/efectos de los fármacos , Norovirus/fisiología , Virus Norwalk/efectos de los fármacos , Virus Norwalk/fisiología , Cultivo Primario de Células , Piridinas/química , Virus Sindbis/efectos de los fármacos , Virus Sindbis/fisiología , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad , Proteasas Ubiquitina-Específicas/metabolismo
10.
Cell Signal ; 23(12): 2076-85, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21855629

RESUMEN

AG490 is a tyrosine kinase inhibitor with activity against Jak2 and apoptotic activity in specific leukemias. Due to its weak kinase inhibitory activity and poor pharmacology, we conducted a cell-based screen for derivatives with improved Jak2 inhibition and activity in animals. Two hits emerged from an initial small chemical library screen, and more detailed structure-activity relationship studies led to the development of WP1130 with 50-fold greater activity in suppressing Jak2-dependent cytokine signaling than AG490. However, WP1130 did not directly suppress Jak2 kinase activity, but mediated Jak2 ubiquitination resulting in its trafficking through HDAC6 to perinuclear aggresomes without cytokine stimulation or SOCS-1 induction. Jak2 primarily contained K63-linked ubiquitin polymers, and mutation of this lysine blocked Jak2 ubiquitination and mobilization in WP1130-treated cells. Further analysis demonstrated that WP1130, but not AG490, acts as a deubiquitinating enzyme (DUB) inhibitor, possibly through a Michael addition reaction. We conclude that chemical modification of AG490 resulted in development of a DUB inhibitor with activity against a DUB capable of modulating Jak2 ubiquitination, trafficking and signal transduction.


Asunto(s)
Endopeptidasas/metabolismo , Janus Quinasa 2/metabolismo , Inhibidores de Proteasas/farmacología , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología , Ubiquitinación , Sustitución de Aminoácidos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cianoacrilatos , Evaluación Preclínica de Medicamentos , Endopeptidasas/química , Pruebas de Enzimas , Proteínas HSP90 de Choque Térmico/metabolismo , Histona Desacetilasa 6 , Histona Desacetilasas/metabolismo , Humanos , Concentración 50 Inhibidora , Interleucina-6/farmacología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Nitrilos/farmacología , Fosforilación , Inhibidores de Proteasoma , Transporte de Proteínas/efectos de los fármacos , Proteolisis , Piridinas/farmacología , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo
11.
Cancer Res ; 70(22): 9265-76, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21045142

RESUMEN

Recent evidence suggests that several deubiquitinases (DUB) are overexpressed or activated in tumor cells and many contribute to the transformed phenotype. Agents with DUB inhibitory activity may therefore have therapeutic value. In this study, we describe the mechanism of action of WP1130, a small molecule derived from a compound with Janus-activated kinase 2 (JAK2) kinase inhibitory activity. WP1130 induces rapid accumulation of polyubiquitinated (K48/K63-linked) proteins into juxtanuclear aggresomes, without affecting 20S proteasome activity. WP1130 acts as a partly selective DUB inhibitor, directly inhibiting DUB activity of USP9x, USP5, USP14, and UCH37, which are known to regulate survival protein stability and 26S proteasome function. WP1130-mediated inhibition of tumor-activated DUBs results in downregulation of antiapoptotic and upregulation of proapoptotic proteins, such as MCL-1 and p53. Our results show that chemical modification of a previously described JAK2 inhibitor results in the unexpected discovery of a novel DUB inhibitor with a unique antitumor mechanism.


Asunto(s)
Apoptosis/efectos de los fármacos , Cuerpos de Inclusión/efectos de los fármacos , Nitrilos/farmacología , Piridinas/farmacología , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Western Blotting , Carboxipeptidasas/antagonistas & inhibidores , Carboxipeptidasas/genética , Carboxipeptidasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cianoacrilatos , Endopeptidasas/genética , Endopeptidasas/metabolismo , Células HEK293 , Humanos , Microscopía Confocal , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Interferencia de ARN , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
12.
Blood ; 109(8): 3470-8, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17202319

RESUMEN

Imatinib mesylate (Gleevec) is effective therapy against Philadelphia chromosome-positive leukemia, but resistance develops in all phases of the disease. Bcr/Abl point mutations and other alterations reduce the kinase inhibitory activity of imatinib mesylate; thus, agents that target Bcr/Abl through unique mechanisms may be needed. Here we describe the activity of WP1130, a small molecule that specifically and rapidly down-regulates both wild-type and mutant Bcr/Abl protein without affecting bcr/abl gene expression in chronic myelogenous leukemia (CML) cells. Loss of Bcr/Abl protein correlated with the onset of apoptosis and reduced phosphorylation of Bcr/Abl substrates. WP1130 did not affect Hsp90/Hsp70 ratios within the cells and did not require the participation of the proteasomal pathway for loss of Bcr/Abl protein. WP1130 was more effective in reducing leukemic versus normal hematopoietic colony formation and strongly inhibited colony formation of cells derived from patients with T315I mutant Bcr/Abl-expressing CML in blast crisis. WP1130 suppressed the growth of K562 heterotransplanted tumors as well as both wild-type Bcr/Abl and T315I mutant Bcr/Abl-expressing BaF/3 cells transplanted into nude mice. Collectively, our results demonstrate that WP1130 reduces wild-type and T315I mutant Bcr/Abl protein levels in CML cells through a unique mechanism and may be useful in treating CML.


Asunto(s)
Apoptosis/efectos de los fármacos , Crisis Blástica/tratamiento farmacológico , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Animales , Apoptosis/genética , Benzamidas , Crisis Blástica/enzimología , Crisis Blástica/genética , Crisis Blástica/patología , Línea Celular Tumoral , Cianoacrilatos , Ensayos de Selección de Medicamentos Antitumorales , Proteínas de Fusión bcr-abl/biosíntesis , Proteínas de Fusión bcr-abl/genética , Regulación Enzimológica de la Expresión Génica , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Desnudos , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Piperazinas/farmacología , Mutación Puntual , Pirimidinas/farmacología , Ensayo de Tumor de Célula Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA