Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Cell Biol ; 2(10): 709-17, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11025661

RESUMEN

Here we show that p53 protein is physically associated with tubulin in vivo and in vitro, and that it localizes to cellular microtubules. Treatment with vincristine or paclitaxel before DNA-damage or before leptomycin B treatment reduces nuclear accumulation of p53 and expression of mdm2 and p21. Overexpression of dynamitin or microinjection of anti-dynein antibody before DNA damage abrogates nuclear accumulation of p53. Our results indicate that transport of p53 along microtubules is dynein-dependent. The first 25 amino acids of p53 contain the residues that are essential for binding to microtubules. We propose that functional microtubules and the dynein motor protein participate in transport of p53 and facilitate its accumulation in the nucleus after DNA damage.


Asunto(s)
Transporte Activo de Núcleo Celular , Dineínas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Complejo Dinactina , Ácidos Grasos Insaturados/farmacología , Técnica del Anticuerpo Fluorescente , Humanos , Proteínas Motoras Moleculares , Paclitaxel/farmacología , Pruebas de Precipitina , Unión Proteica , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas , Vincristina/farmacología
2.
Oncogene ; 26(48): 6954-8, 2007 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-17486059

RESUMEN

Damage-induced G1 checkpoint in mammalian cells involves upregulation of p53, which activates transcription of p21(Waf1) (CDKN1A). Inhibition of cyclin-dependent kinase (CDK)2 and CDK4/6 by p21 leads to dephosphorylation and activation of Rb. We now show that ectopic p21 expression in human HT1080 fibrosarcoma cells causes not only dephosphorylation but also depletion of Rb; this effect was p53-independent and susceptible to a proteasome inhibitor. CDK inhibitor p27 (CDKN1B) also caused Rb dephosphorylation and depletion, but another CDK inhibitor p16 (CDKN2A) induced only dephosphorylation but not depletion of Rb. Rb depletion was observed in both HT1080 and HCT116 colon carcinoma cells, where p21 was induced by DNA-damaging agents. Rb depletion after DNA damage did not occur in the absence of p21, and it was reduced when p21 induction was inhibited by p21-targeting short hairpin RNA or by a transdominant inhibitor of p53. These results indicate that p21 both activates Rb through dephosphorylation and inactivates it through degradation, suggesting negative feedback regulation of damage-induced cell-cycle checkpoint arrest.


Asunto(s)
Neoplasias del Colon/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibrosarcoma/metabolismo , Proteína de Retinoblastoma/metabolismo , Antibióticos Antineoplásicos/farmacología , Neoplasias del Colon/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN/efectos de los fármacos , Doxorrubicina/farmacología , Fibrosarcoma/patología , Humanos , Immunoblotting , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
3.
Cell Death Differ ; 13(9): 1434-41, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16311509

RESUMEN

Selective modulation of cell death is important for rational chemotherapy. By depleting Hsp90-client oncoproteins, geldanamycin (GA) and 17-allylamino-17-demethoxy-GA (17-AAG) (heat-shock protein-90-active drugs) render certain oncoprotein-addictive cancer cells sensitive to chemotherapy. Here we investigated effects of GA and 17-AAG in apoptosis-prone cells such as HL60 and U937. In these cells, doxorubicin (DOX) caused rapid apoptosis, whereas GA-induced heat-shock protein-70 (Hsp70) (a potent inhibitor of apoptosis) and G1 arrest without significant apoptosis. GA blocked caspase activation and apoptosis and delayed cell death caused by DOX. Inhibitors of translation and transcription and siRNA Hsp70 abrogated cytoprotective effects of GA. Also GA failed to protect HL60 cells from cytotoxicity of actinomycin D and flavopiridol (FL), inhibitors of transcription. We next compared cytoprotection by GA-induced Hsp70, caspase inhibitors (Z-VAD-fmk) and cell-cycle arrest. Whereas cell-cycle arrest protected HL60 cells from paclitaxel (PTX) but not from FL and DOX, Z-VAD-fmk prevented FL-induced apoptosis but was less effective against DOX and PTX. Thus, by inducing Hsp70, GA protected apoptosis-prone cells in unique and cell-type selective manner. Since GA does not protect apoptosis-reluctant cancer cells, we envision a therapeutic strategy to decrease side effects of chemotherapy without affecting its therapeutic efficacy.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Benzoquinonas/farmacología , Inhibidores de Caspasas , Doxorrubicina/farmacología , Proteínas HSP70 de Choque Térmico/biosíntesis , Lactamas Macrocíclicas/farmacología , Clorometilcetonas de Aminoácidos/farmacología , Caspasa 9/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Citoprotección , Dactinomicina/farmacología , Activación Enzimática , Flavonoides/farmacología , Humanos , Paclitaxel/farmacología , Piperidinas/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/genética , Activación Transcripcional/efectos de los fármacos
4.
J Clin Invest ; 105(4): 533-9, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10683383

RESUMEN

Cell-cycle checkpoint mechanisms, including the p53- and p21-dependent G(2) arrest that follows DNA damage, are often lost during tumorigenesis. We have exploited the ability of DNA-damaging drugs to elicit this checkpoint, and we show here that such treatment allows microtubule drugs, which cause cell death secondary to mitotic arrest, to kill checkpoint-deficient tumor cells while sparing checkpoint-competent cells. Low doses of the DNA-damaging drug doxorubicin cause predominantly G(2) arrest without killing HCT116 cells that harbor wt p53. Doxorubicin treatment prevented mitotic arrest, Bcl-2 phosphorylation, and cell death caused by paclitaxel, epothilones, and vinblastine. In contrast, doxorubicin enhanced cytotoxicity of FR901228, an agent that does not affect microtubules. Low doses of doxorubicin did not arrest p21-deficient clones of HCT116 cells and did not protect these cells from cytotoxicity caused by microtubule drugs, but cells in which p21 expression was restored enjoyed partial protection under these conditions. Moreover, in p53-deficient clones of HCT116 cells doxorubicin did not induce either p53 or p21 and provided no protection against paclitaxel-induced cytotoxicity. Therefore, (a) p53-dependent p21 induction caused by doxorubicin protects from microtubule drug-induced cytotoxicity, and (b) pretreatment with cytostatic doses of DNA-damaging drugs before treatment with microtubule drugs results in selective cytotoxicity to cancer cells with defective p53/p21-dependent checkpoint.


Asunto(s)
Antineoplásicos/farmacología , Microtúbulos/efectos de los fármacos , Mutágenos/farmacología , División Celular/efectos de los fármacos , Neoplasias del Colon , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Ciclinas/metabolismo , Doxorrubicina/farmacología , Interacciones Farmacológicas , Compuestos Epoxi/farmacología , Humanos , Paclitaxel/farmacología , Tiazoles/farmacología , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Vinblastina/farmacología
5.
Mol Cell Biol ; 16(10): 5839-45, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8816498

RESUMEN

The serine/threonine kinase Raf-1 functions downstream of Rats in a signal transduction cascade which transmits mitogenic stimuli from the plasma membrane to the nucleus. Raf-1 integrates signals coming from extracellular factors and, in turn, activates its substrate, MEK kinase. MEK activates mitogen-activated protein kinase (MAPK), which phosphorylates other kinases as well as transcription factors. Raf-1 exists in a complex with HSP90 and other proteins. The benzoquinone ansamycin geldanamycin (GA) binds to HSP90 and disrupts the Raf-1-HSP90 multimolecular complex, leading to destabilization of Raf-1. In this study, we examined whether Raf-1 destabilization is sufficient to block the Raf-1-MEK-MAPK signalling pathway and whether GA specifically inactivates the Raf-1 component of this pathway. Using the model system of NIH 3T3 cells stimulated with phorbol 12-myristate 13-acetate (PMA), we show that GA does not affect the ability of protein kinase C alpha to be activated by phorbol esters, but it does block activation of MEK and MAPK. Further, GA does not decrease the activity of constitutively active MEK in transiently transfected cells. Finally, disruption of the Raf-1-MEK-MAPK signalling pathway by GA prevents both the PMA-induced proliferative response and PMA-induced activation of a MAPK-sensitive nuclear transcription factor. Thus, we demonstrate that interaction between HSP90 and Raf-1 is a sine qua non for Raf stability and function as a signal transducer and that the effects observed cannot be attributed to a general impairment of protein kinase function.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Inhibidores Enzimáticos/farmacología , Quinasa 1 de Quinasa de Quinasa MAP , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Quinonas/farmacología , Transducción de Señal/fisiología , Activación Transcripcional/efectos de los fármacos , Células 3T3 , Animales , Benzoquinonas , División Celular/efectos de los fármacos , ADN/biosíntesis , Genes Reporteros , Proteínas HSP90 de Choque Térmico/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Células Jurkat , Lactamas Macrocíclicas , Ratones , Oligonucleótidos Antisentido/farmacología , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-raf , Ratas , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Transfección
6.
Leukemia ; 20(3): 385-91, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16357832

RESUMEN

Tumor stem cells are quiescent and, therefore, resistant to therapy, yet harbor the capacity to replenish a tumor after therapy. Therefore, it is tempting to explain all therapeutic failures by the persistence of tumor stem cells. Yet, this explanation is relevant only to initial stages of stem-cell-dependent tumors (such as chronic myeloid leukemia) that, actually, are well controlled by therapy. In advanced cancers that poorly respond to therapy, quiescent tumor stem cells play a negligible role. Instead, proliferating cells determine disease progression, prognosis, therapeutic failures, and resistance to therapy. And therapy fails not because it eliminates only proliferating tumor cells, but because it does not eliminate them. With noticeable exceptions, it is the proliferating cell that should be targeted, whereas resting cancer cells including stem and dormant cells need to be targeted only when they 'wake up'. Finally, I discuss a strategy of selectively killing dominant proliferating clones, including proliferating stem-like and drug-resistant cancer cells, while sparing normal cells.


Asunto(s)
División Celular , Neoplasias/terapia , Trasplante de Células Madre , Línea Celular , Resistencia a Antineoplásicos , Humanos , Neoplasias/patología , Recurrencia
7.
Cell Death Differ ; 12(6): 592-602, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15818400

RESUMEN

Carcinogenesis and cancer therapy are two sides of the same coin, such that the same cytotoxic agent can cause cancer and be used to treat cancer. This review links carcinogenesis, chemoprevention and cancer therapy in one process driven by cytotoxic agents (carcinoagents) that select either for or against cells with oncogenic alterations. By unifying therapy and cancer promotion and by distinguishing nononcogenic and oncogenic mechanisms of resistance, I discuss anticancer- and chemopreventive agent-induced carcinogenesis and tumor progression and, vice versa, carcinogens as anticancer drugs, anticancer drugs as chemopreventive agents and exploiting oncogene-addiction and drug resistance for chemoprevention and cancer therapy.


Asunto(s)
Carcinógenos/efectos adversos , Quimioprevención , Neoplasias/tratamiento farmacológico , Animales , Carcinógenos/farmacología , Carcinógenos/uso terapéutico , Quimioprevención/tendencias , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Humanos , Neoplasias/patología
8.
Cancer Res ; 61(11): 4301-5, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11389048

RESUMEN

Chemotherapy of cancer is limited by its toxicity to normal cells. On the basis of discoveries in signal transduction and cell cycle regulation, novel mechanism-based therapeutics are being developed. Although these cell cycle modulators were designed to target cancer cells, some of them can also be applied for a different purpose, i.e., to protect normal cells against the lethality of chemotherapy. Loss of sensitivity of cancer cells to cell cycle inhibitors can be exploited for selective protection of normal cells that retain this response. Indeed, inhibition of redundant or overactivated pathways (e.g., growth factor-activated pathways) or stimulation of absent pathways in cancer cells (e.g., p53, Rb, and p16) may not arrest cycling of cancer cells. But growth arrest of normal cells will then permit selective killing of cancer cells by cycle-dependent chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales , Antineoplásicos/efectos adversos , División Celular/efectos de los fármacos , Humanos
9.
Cancer Res ; 60(13): 3375-8, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10910041

RESUMEN

Nickel (Ni2+) and cobalt (Co2+) mimic hypoxia and were used as a tool to study the role of oxygen sensing and signaling cascades in the regulation of hypoxia-inducible gene expression. These metals can produce oxidative stress; therefore, it was conceivable that reactive oxygen species (ROS) may trigger signaling pathways resulting in the activation of the hypoxia-inducible factor (HIF)-1 transcription factor and up-regulation of hypoxia-related genes. We found that the exposure of A549 cells to Co2+ or Ni2+ produced oxidative stress, and although Co2+ was a more potent producer of ROS than Ni2+, both metals equally increased the expression of Cap43, a hypoxia-regulated gene. The coadministration of hydrogen peroxide with metals induced more ROS; however, this did not further increase the expression of Cap43 mRNA. The free radical scavenger 2-mercaptoethanol completely suppressed ROS generation by CoCl2 and NiCl2 but did not diminish the induced Cap43 gene expression. The activity of the HIF-1 transcription factor as assessed in transient transfection assays was stimulated by Ni2+, hypoxia, and desferrioxamine, but this activation was not diminished when oxidative stress was attenuated nor was HIF-dependent transcription enhanced by hydrogen peroxide. We conclude that ROS are produced during the exposure of cells to metals that mimic hypoxia, but the formation of ROS was not involved in the activation of HIF-1-dependent genes.


Asunto(s)
Carcinógenos/toxicidad , Hipoxia de la Célula/fisiología , Cobalto/toxicidad , Proteínas de Unión al ADN/metabolismo , Níquel/toxicidad , Proteínas Nucleares/metabolismo , Proteínas/genética , Transcripción Genética/efectos de los fármacos , Proteínas de Ciclo Celular , Endotelio Vascular , Depuradores de Radicales Libres/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Peróxido de Hidrógeno/toxicidad , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Péptidos y Proteínas de Señalización Intracelular , Neoplasias Pulmonares , Mercaptoetanol/farmacología , Especies Reactivas de Oxígeno/fisiología , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología , Células Tumorales Cultivadas
10.
Cancer Res ; 60(20): 5630-4, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11059752

RESUMEN

Hypoxia limits tumor growth but selects for higher metastatic potential. We tested the functional activity of hypoxia-inducible factor-1 (HIF-1) in prostate cell lines ranging from normal epithelial cells (PrEC), hormone-dependent LNCaP, hormone-independent DU145, PC-3 to highly metastatic PC-3M cancer cell lines. We found that HIF-1-stimulated transcription was the lowest in PrEC and LNCaP cells and the highest in PC-3M cells. The induction by hypoxia of the HIF-1 dependent genes Cap43 and GAPDH was the highest in the most aggressive PC-3M cancer cells. Because these advanced prostate cancer cell lines have lost p53 function, this further shifts a balance from p53 to HIF-1 transcriptional regulation, and a high ratio of HIF-1-dependent:p53-dependent transcription was a marker of the advanced malignant phenotype. Transient transfection of HIF-1alpha expression vector induced transcription from p21 promoter construct in prostate cancer cell lines. Furthermore, hypoxia slightly induced p21 mRNA in these cells. However, neither expression of p21 nor hypoxia caused growth arrest in PC-3M cells. Therefore, high inducibility of HIF-1-dependent genes, loss of p53 functions with high ratio of HIF-1-dependent:p53-dependent transcription, and loss of sensitivity to p21 inhibition is a part of hypoxic phenotype associated with aggressive cancer behavior.


Asunto(s)
Ciclinas/fisiología , Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Neoplasias de la Próstata/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/fisiología , Ciclo Celular/genética , Proteínas de Ciclo Celular , Hipoxia de la Célula/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/biosíntesis , Ciclinas/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/fisiología , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Péptidos y Proteínas de Señalización Intracelular , Masculino , Metástasis de la Neoplasia , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/fisiología , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Biosíntesis de Proteínas , Proteínas/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/fisiología , Transcripción Genética/fisiología , Activación Transcripcional/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
11.
Cancer Res ; 57(2): 320-5, 1997 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9000576

RESUMEN

Growth arrest and differentiation of leukemic cells by phorbol 12-myristate 13-acetate (PMA) is accompanied by p53-independent activation of p21WAF1/CIP1 and c-myc down-regulation. We show that despite p21 induction in 7 of 12 human cancer cell lines treated with PMA, growth inhibition was observed only in two cell lines (SKBr3 breast and LNCaP prostate cancer cells). Treatment of SKBr3 and LNCaP cells with PMA was followed by Raf-1 hyperphosphorylation, p21 induction, Rb hypophosphorylation, c-myc down-regulation and growth inhibition. The 10 remaining PMA-resistant cell lines were comprised of 5 that failed to induce p21 and 5 that induced p21 but had defects in steps putatively downstream of this (Rb hypophosphorylation and c-myc down-regulation). Exogenous expression and subsequent failure to down-regulate c-myc protein expression in SKBr3 and LNCaP cells was correlated with acquisition of resistance to the growth inhibitory effect of PMA. Exogenous p21 expression down-regulated c-myc protein in PMA-sensitive cancer cells. Our findings suggest that induction of p21 and down-regulation of c-myc may be necessary steps in a PMA-induced growth-inhibitory pathway in cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carcinógenos/farmacología , Ciclinas/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteína de Retinoblastoma/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Regulación hacia Abajo , Resistencia a Medicamentos , Femenino , Genes myc , Humanos , Masculino , Fosforilación , Neoplasias de la Próstata/patología , Transducción de Señal , Transfección , Células Tumorales Cultivadas/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
12.
Cancer Res ; 60(1): 38-41, 2000 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-10646848

RESUMEN

Carcinogenic nickel compounds alter the program of gene expression in normal cells and induce a pattern of gene expression similar to that found in nickel-induced cancers. Here we have demonstrated that nickel exposure induced hypoxic signaling pathways by inducing hypoxia-inducible transcription factor-1 (HIF-1), which mediated the induction of genes required by cells to survive hypoxia. We also show that a new gene, Cap43, is dependent upon HIF-1 because only HIF-1-proficient cells induced Cap43 when exposed to either hypoxia or nickel. We also show that glyceraldehyde-3-phosphate dehydrogenase, a gene induced by hypoxia through HIF-1, was similar to Cap43 in that it required HIF-1-proficient cells to be induced by either nickel or hypoxia. These data demonstrate that nickel exposure turns on signaling for hypoxic stress, which may be important in its carcinogenesis.


Asunto(s)
Carcinógenos/toxicidad , Hipoxia de la Célula/genética , Proteínas de Unión al ADN/fisiología , Expresión Génica/efectos de los fármacos , Níquel/toxicidad , Proteínas Nucleares/fisiología , Proteínas/efectos de los fármacos , Factores de Transcripción , Animales , Northern Blotting , Western Blotting , Calcio/metabolismo , Proteínas de Ciclo Celular , Hipoxia de la Célula/fisiología , Proteínas de Unión al ADN/genética , Fibroblastos/metabolismo , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Péptidos y Proteínas de Señalización Intracelular , Ionóforos/farmacología , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
13.
Cancer Res ; 56(8): 1851-4, 1996 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-8620503

RESUMEN

c-Raf-1 (Raf-1) is a central component of signal transduction pathways stimulated by various growth factors, protein kinase C, and other protein kinases. Raf-1 activation is thought to be initiated at the plasma membrane after its recruitment by Ras. Raf-1 activation is associated primarily with proliferation and cell survival, but it has also been implicated in apoptosis. Raf-1 has also been shown to form complexes with both R-Ras and Bcl-2, raising the possibility that this component of cellular Raf-1 plays a role in apoptosis. Recently, taxol was reported to induce Bcl-2 phosphorylation and inactivation. We have previously demonstrated Raf-1 activation following taxol in MCF7 cells. We now present evidence that taxol fails to stimulate either apoptosis or phosphorylation of Bel-2 in the absence of Raf-1. Moreover, Raf-1 activation by taxol coincided with Bel-2 phosphorylation, showing similar dose and time dependence. Thus, our data support a role for a distinct subcellular component of Raf-1, which is taxol but not phorbol myristate acetate sensitive, in mediating an apoptotic pathway involving Bc1-2.


Asunto(s)
Antineoplásicos Fitogénicos/toxicidad , Apoptosis/efectos de los fármacos , Paclitaxel/toxicidad , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Apoptosis/fisiología , Neoplasias de la Mama , Línea Celular , Activación Enzimática , Femenino , Células HL-60 , Humanos , Masculino , Fosforilación , Neoplasias de la Próstata , Proteínas Serina-Treonina Quinasas/aislamiento & purificación , Proteínas Proto-Oncogénicas/aislamiento & purificación , Proteínas Proto-Oncogénicas c-bcl-2 , Proteínas Proto-Oncogénicas c-raf , Acetato de Tetradecanoilforbol/farmacología , Células Tumorales Cultivadas
14.
Cancer Res ; 60(13): 3425-8, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10910051

RESUMEN

Lack of selectivity in the killing of tumor and normal cells is a major obstacle in cancer therapy. By inhibiting normal but not autonomous cell growth, we exploited the differences in cell cycle regulation to achieve a selective protection of nonautonomous cells against paclitaxel and other microtubule-active drugs. Tubulin polymerization, a primary effect of paclitaxel, can be dissociated from Bcl-2 phosphorylation and cytotoxicity in HL-60 cells. Growth arrest prevented paclitaxel-induced Bcl-2 phosphorylation and apoptosis without affecting paclitaxel-induced tubulin polymerization. We abrogated the effects of paclitaxel on MCF-10A immortalized breast cells, while preserving its effects on MCF-7 cancer cells. Unlike MCF-7 cells, MCF-10A cells were arrested by epidermal growth factor withdrawal, precluding paclitaxel-induced Bcl-2 phosphorylation. Furthermore, the inhibition of the epidermal growth factor receptor kinase with low doses of AG1478 arrested growth of MCF-10A but not MCF-7 cells. Pretreatment with AG1478 did not affect paclitaxel-induced Bcl-2/Raf-1 phosphorylation in MCF-7 but abrogated such phosphorylation in MCF-10A. Exploitation of growth factor dependency may allow the protection of normal cells from microtubule-active drugs.


Asunto(s)
Ciclo Celular/fisiología , Microtúbulos/efectos de los fármacos , Paclitaxel/toxicidad , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tirfostinos/toxicidad , Apoptosis/efectos de los fármacos , Neoplasias de la Mama , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/toxicidad , Factor de Crecimiento Epidérmico/farmacología , Femenino , Células HL-60 , Humanos , Mitosis/efectos de los fármacos , Fosforilación , Quinazolinas , Acetato de Tetradecanoilforbol/farmacología , Tubulina (Proteína)/efectos de los fármacos , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas
15.
Cancer Res ; 55(20): 4623-6, 1995 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-7553639

RESUMEN

Taxol stabilizes microtubules, prevents tubulin depolymerization, and promotes tubulin bundling and is one of the most effective drugs for the treatment of metastatic breast and ovarian cancer. Although its interaction with tubulin has been well characterized, the mechanism by which taxol induces growth arrest and cytotoxicity is not well understood. Herein, we show that taxol induced dose- and time-dependent accumulation of the cyclin inhibitor p21WAF1 in both p53 wild-type and p53-null cells, although the degree of induction was greater in cells expressing wild-type p53. In MCF7 cells, wild-type p53 protein was also induced after taxol treatment, and this induction was mediated primarily by increased protein stability. Taxol induced both p21WAF1 and wild-type p53 optimally in MCF7 cells after 20-24-h exposure with an EC50(3) of 5 nM. In p53-null PC3M cells, p21WAF1 was similarly induced after 24-h exposure to taxol. Coincident with these biochemical effects, taxol altered the electrophoretic mobility of c-raf-1 and stimulated mitogen activated protein kinase. Previous depletion of c-raf-1 inhibited both the p21WAF1- and p53-inducing properties of taxol, as well as the activation of MAP kinase. These data suggest that induction of p21WAF1 by taxol requires c-raf-1 activity, but that it is not strictly dependent on wild-type p53. Furthermore, the ability of taxol to both induce wild-type p53 in MCF7 cells and activate MAP kinase is also dependent on c-raf-1 expression.


Asunto(s)
Ciclinas/biosíntesis , Paclitaxel/farmacología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteína p53 Supresora de Tumor/biosíntesis , Células 3T3 , Animales , Benzoquinonas , Proteínas Quinasas Dependientes de Calcio-Calmodulina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidores Enzimáticos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Lactamas Macrocíclicas , Ratones , Microtúbulos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf , Quinonas/farmacología , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas
16.
Cancer Res ; 58(15): 3331-8, 1998 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9699663

RESUMEN

The oncogenic protein Bcl-2 functions as a potent inhibitor of programmed cell death. This survival activity has been shown in some settings to be influenced by the Bcl-2 phosphorylation state. It has been demonstrated that treatment with microtubule-targeted agents results in phosphorylation of both Raf-1 kinase and Bcl-2. The Bcl-2-related family member Bcl-xL also exhibits a death suppressive activity, but its potential for phosphorylation following exposure to drugs that interact with microtubules has not been evaluated. Several tumor cell lines with low or undetectable levels of Bcl-2 protein expression were found to express Bcl-xL. A more slowly migrating Bcl-xL band was observed on immunoblots after cells were treated with microtubule-targeted agents. The appearance of this band was responsive to dose and was absent when the cell lysates were treated with lambda protein phosphatase. Using a Bcl-xL-specific monoclonal antibody, the phosphorylated form of Bcl-xL was immunoprecipitated from cells treated with paclitaxel and metabolically labeled with 32P-labeled inorganic orthophosphate. Herein, we report that Bcl-xL is phosphorylated in malignant cells after incubation with agents that target tubulin, including paclitaxel, vincristine, vinblastine, colchicine, and nocodazole. Moreover, paclitaxel-resistant ovarian carcinoma cell lines that have mutations in tubulin failed to exhibit phosphorylation of Bcl-xL after paclitaxel exposure, but they did demonstrate Bcl-xL phosphorylation in the presence of other tubulin-targeting agents. As observed for Bcl-2, phosphorylation of Bcl-xL was accompanied by phosphorylation of Raf-1. Interestingly, phosphorylation of these three proteins failed to occur or was much less pronounced when cells grown at high density were challenged with drug. Also, reduced Raf-1 expression, observed after treatment of cells with geldanamycin prior to and during incubation with the microtubule-active drugs, correlated with diminished Bcl-xL phosphorylation. Taken together, these results suggest that Bcl-xL, like Bcl-2, is phosphorylated by agents that disrupt microtubule architecture. By analogy with Bcl-2, this phosphorylation may play a critical role in modulating Bcl-xL function and may be an important determinant of microtubule-directed chemotherapeutic efficacy in human tumors.


Asunto(s)
Microtúbulos/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Benzoquinonas , Recuento de Células , Resistencia a Antineoplásicos , Electroforesis en Gel de Poliacrilamida , Humanos , Lactamas Macrocíclicas , Microtúbulos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/ultraestructura , Paclitaxel/farmacología , Fosfoproteínas Fosfatasas/metabolismo , Radioisótopos de Fósforo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Proteínas Proto-Oncogénicas c-raf/metabolismo , Quinonas/farmacología , Dodecil Sulfato de Sodio , Tubulina (Proteína)/efectos de los fármacos , Células Tumorales Cultivadas , Proteína bcl-X
17.
Cancer Res ; 57(1): 130-5, 1997 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-8988053

RESUMEN

Recent studies have shown that paclitaxel leads to activation of Raf-1 kinase and have suggested that this activation is essential for bcl-2 phosphorylation and apoptosis. In the present study, we demonstrate that, in addition to paclitaxel, other agents that interact with tubulin and microtubules also induce Raf-1/bcl-2 phosphorylation, whereas DNA-damaging drugs, antimetabolites, and alkylating agents do not. Activation of Raf-1 kinase by paclitaxel is linked to tubulin polymerization; the effect is blunted in paclitaxel-resistant cells, the tubulin of which does not polymerize following the addition of paclitaxel. In contrast, vincristine and vinblastine, drugs to which the paclitaxel-resistant cells retain sensitivity were able to bring about Raf-1 phosphorylation. The requirement for disruption of microtubules in this signaling cascade was strengthened further using paclitaxel analogues by demonstrating a correlation between tubulin polymerization, Raf-1/bcl-2 phosphorylation, and cytotoxicity. Inhibition of RNA or protein synthesis prevents Raf-1 activation and bcl-2 phosphorylation, suggesting that an intermediate protein(s) acts upstream of Raf-1 in this microtubule damage-activating pathway. A model is proposed that envisions a pathway of Raf-1 activation and bcl-2 phosphorylation following disruption of microtubular architecture, serving a role similar to p53 induction following DNA damage.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Microtúbulos/efectos de los fármacos , Paclitaxel/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Tubulina (Proteína)/efectos de los fármacos , Antineoplásicos/farmacología , Resistencia a Medicamentos , Activación Enzimática , Femenino , Humanos , Fosforilación , Polímeros/metabolismo , Inhibidores de Proteínas Quinasas , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
18.
Oncogene ; 20(3): 395-8, 2001 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-11313969

RESUMEN

Recently it has been shown that the VHL tumor suppressor targets the hypoxia-inducible transcription factor (HIF-1) for ubiquitin-dependent degradation by the proteasome. Past mysteries of the p53 tumor suppressor help to solve the present puzzles of the VHL tumor suppressor. Thus, Mdm-2 targets the p53 tumor suppressor for ubiquitin-dependent degradation by the proteasome, but, in addition, the p53 transcription factor induces Mdm-2, thus, establishing a feedback loop. Hypoxia or DNA damage by abrogating binding of HIF-1 with VHL and p53 with Mdm-2, respectively, leads to stabilization and accumulation transcriptionally active HIF-1 and p53. More detailed analysis depicts the VHL/HIF-1 pair as the p53/mdm-2 pair that is turned upside down, suggesting that VHL may be a HIF-1-inducible gene of the feedback loop. The extended model proposes that an oncoprotein and a tumor suppressor due to transactivation coupled with feedback protein degradation might form functional pairs (Rb/E7, E2F/Rb, E2F/Mdm-2, catenin/APC, p27, cyclin D1, Rb/gankyrin), thus, predicting missing links.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Ligasas , Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas , Animales , Proteínas de Unión al ADN/genética , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Proteínas Nucleares/genética , Proteínas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-mdm2 , Proteína p53 Supresora de Tumor/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
19.
Oncogene ; 15(16): 1889-93, 1997 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-9365234

RESUMEN

Wild-type (wt) p53 protein is rapidly degraded, has a short half-life and low intracellular levels. Stabilization of wt p53 protein following an appropriate stimulus (for example DNA damage) is a physiological regulation to increase function. In contrast, stabilization of p53 protein in the absence of a stimulus is always a hallmark of loss of function secondary to a mutation, or interaction with viral or cellular oncoproteins. It is generally accepted that stability of p53 protein depends on its intrinsic biochemical properties such as conformation or protein/protein interactions. However, I will discuss evidence that the stability of p53 is not a consequence of its intrinsic properties, but instead is determined by feedback control of its function. In the absence of an appropriate stimulus, a cell needs to keep p53 levels low, since increased levels can lead to apoptosis. To precisely regulate p53 levels, a cell must sense its level; and sensing its transactivating function, is the simplest way to sense p53. Following an appropriate stimulus (for example, DNA damage), the cell senses a state of 'relative' p53 deficiency and adapts by reducing p53 degradation. When the state of p53 deficiency is a consequence of a mutation or interaction with viral oncoproteins, the cell does not sense p53, and again attempts to adapt by reducing p53 degradation. However, in the latter case, the increase in levels does not restore function, and the adaptation continues until degradation of p53 protein is maximally inhibited. In this case, no further inhibition of degradation is possible after DNA-damage or pharmacological inhibition of proteasomes. Thus lack of wt p53 function always results in increased p53 levels and nonregulation.


Asunto(s)
Proteína p53 Supresora de Tumor/metabolismo , Hidrólisis , Mutación , Unión Proteica , Proteína p53 Supresora de Tumor/genética
20.
Oncogene ; 11(5): 933-9, 1995 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-7675452

RESUMEN

Mutated p53 proteins interfere in the function of wild type p53 and may also serve as a dominant oncogene. The vast majority of p53 mutations result in a protein of altered conformation and prolonged half-life. We sought to examine whether geldanamycin, a drug capable of destabilizing several oncogene and proto-oncogene products, could alter the stability and DNA binding characteristics of several mutated p53 proteins. Brief exposure to GA destabilized the p53 protein of several breast, prostate and leukemic cell lines harboring mutated p53 alleles, resulting in a significant reduction in p53 steady state level and half-life. In contrast to its effects on mutated p53, GA altered neither steady state level nor inducibility of the wild type protein. In addition to its effects on protein stability, GA also altered the conformation of mutated p53, so that it was no longer detectable with a mutant conformation-specific antibody. Finally, mutated p53 protein isolated from GA-treated cells regained partial ability to bind a wild type-specific p53 DNA consensus sequence. These data indicate the feasibility of pharmacologic intervention for altering the mutated p53 phenotype.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Quinonas/farmacología , Proteína p53 Supresora de Tumor/efectos de los fármacos , Benzoquinonas , Línea Celular , ADN/metabolismo , Semivida , Proteínas de Choque Térmico/fisiología , Humanos , Lactamas Macrocíclicas , Mutación , Conformación Proteica , Proto-Oncogenes Mas , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA