Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Am Chem Soc ; 132(9): 3224-8, 2010 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-20155906
2.
Mol Cancer Ther ; 5(1): 60-7, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16432163

RESUMEN

Exisulind (sulindac sulfone) and three highly potent derivatives, OSI-461 (CP461), OSIP486823 (CP248), and OSIP487703, inhibit growth and induce apoptosis in SW480 human colon cancer cells, with IC(50)s of 200, 2, 0.1, and 0.003 micromol/L, respectively. The latter three compounds, but not exisulind, induce marked M-phase cell cycle arrest in these cells. This effect seems to be independent of the known ability of these compounds to cause activation of protein kinase G. When tested at twice their IC(50) concentration for growth inhibition, OSI-461, OSIP486823, and OSIP487703 cause depolymerization of microtubules in interphase cells, inhibit spindle formation in mitotic cells, and induce multinucleated cells. In vitro tubulin polymerization assays indicate that all three compounds interact with tubulin directly to cause microtubule depolymerization and/or inhibit de novo tubulin polymerization. These results suggest that the dual effects of OSI-461, OSIP486823, and OSIP487703 on impairment of microtubule functions and protein kinase G activation may explain the potent antiproliferative and apoptotic effects of these compounds in cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Microtúbulos/efectos de los fármacos , Mitosis/efectos de los fármacos , Sulindac/análogos & derivados , Células 3T3/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Microtúbulos/metabolismo , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo , Sulindac/farmacología , Tionucleótidos/farmacología , Tubulina (Proteína)/efectos de los fármacos , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas
3.
Mol Cancer Ther ; 4(9): 1388-98, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16170031

RESUMEN

We previously reported that the garlic-derived compound S-allylmercaptocysteine (SAMC) causes growth inhibition, mitotic arrest, and induction of apoptosis in SW480 human colon cancer cells by inducing microtubule depolymerization and c-Jun NH(2) terminus kinase-1 activation. In the present study, we compared the aforementioned effects of SAMC to those of a series of garlic-derived and other organosulfur compounds. Among the 10 compounds tested, only SAMC, diallyl disulfide (DADS), and S-trityl-L-cysteine (trityl-cys) cause significant inhibition of cell growth with IC(50) values of 150, 56, and 0.9 micromol/L, respectively. These three compounds also induce G(2)-M cell cycle arrest and apoptosis. Further studies reveal that, like SAMC, the garlic-derived compound DADS exerts antiproliferative effects by binding directly to tubulin and disrupting the microtubule assembly, thus arresting cells in mitosis and triggering mitochondria-mediated signaling pathways that lead to apoptosis. However, the synthetic compound trityl-cys exerts its effect on M-phase arrest and growth inhibition by mechanisms that involve spindle impairment but do not involve disruption of microtubule structure or dynamics. Furthermore, trityl-cys does not induce marked loss of mitochondrial membrane potential or release of cytochrome c, but it does induce caspase-3 activation and poly(ADP-ribose) polymerase cleavage. Structure-function analysis suggests that both the allyl and the disulfide moieties are important features for the antiproliferative effects of SAMC and DADS. These findings may be useful in the identification, synthesis, and development of organosulfur compounds that have anticancer activity.


Asunto(s)
Adenocarcinoma/patología , Apoptosis/efectos de los fármacos , Neoplasias del Colon/patología , Mitosis/efectos de los fármacos , Compuestos de Azufre/farmacología , Adenocarcinoma/metabolismo , Compuestos Alílicos/farmacología , Caspasa 3 , Caspasas/metabolismo , División Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Citocromos c/metabolismo , Activación Enzimática/efectos de los fármacos , Ajo/química , Humanos , Potenciales de la Membrana/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mitocondrias/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas
4.
Clin Cancer Res ; 10(19): 6710-21, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15475462

RESUMEN

Hepatoma is one of the most frequently occurring cancers worldwide. However, effective chemotherapeutic agents for this disease have not been developed. Acyclic retinoid, a novel synthetic retinoid, can reduce the incidence of postsurgical recurrence of hepatoma and improve the survival rate. OSI-461, a potent derivative of exisulind, can increase intracellular levels of cyclic GMP, which leads to activation of protein kinase G and induction of apoptosis in cancer cells. In the present study, we examined the combined effects of acyclic retinoid plus OSI-461 in the HepG2 human hepatoma cell line. We found that the combination of as little as 1.0 micromol/L acyclic retinoid and 0.01 micromol/L OSI-461 exerted synergistic inhibition of the growth of HepG2 cells. Combined treatment with low concentrations of these two agents also acted synergistically to induce apoptosis in HepG2 cells through induction of Bax and Apaf-1, reduction of Bcl-2 and Bcl-xL, and activation of caspase-3, -8, and -9. OSI-461 enhanced the G0-G1 arrest caused by acyclic retinoid, and the combination of these agents caused a synergistic decrease in the levels of expression of cyclin D1 protein and mRNA, inhibited cyclin D1 promoter activity, decreased the level of hyperphosphorylated forms of the Rb protein, induced increased cellular levels of the p21(CIP1) protein and mRNA, and stimulated p21(CIP1) promoter activity. Moreover, OSI-461 enhanced the ability of acyclic retinoid to induce increased cellular levels of retinoic acid receptor beta and to stimulate retinoic acid response element-chloramphenicol acetyltransferase activity. A hypothetical model involving concerted effects on p21(CIP1) and retinoic acid receptor beta expression is proposed to explain these synergistic effects. Our results suggest that the combination of acyclic retinoid plus OSI-461 might be an effective regimen for the chemoprevention and chemotherapy of human hepatoma and possibly other malignancies.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sulindac/análogos & derivados , Tretinoina/análogos & derivados , Tretinoina/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Moléculas de Adhesión Celular/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , GMP Cíclico/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Proteínas del Citoesqueleto/metabolismo , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Fase G1/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteínas de Microfilamentos , Modelos Biológicos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Elementos de Respuesta/genética , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Proteína de Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulindac/farmacología , Transactivadores/metabolismo , Transfección , Proteína p53 Supresora de Tumor/metabolismo , beta Catenina
5.
Breast Cancer Res Treat ; 83(3): 221-31, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14758092

RESUMEN

The purpose of this study was to determine whether black cohosh contains constituents that inhibit the growth of human breast cancer cells, and therefore might eventually be useful in the prevention or treatment of breast cancer. Black cohosh rhizomes were extracted with methanol/water and fractionated by solvent-solvent partitioning to yield three fractions: hexane, ethyl acetate and water. The ethyl acetate fraction displayed the highest potency in two cell-based assays, growth inhibition and cell cycle analysis. This fraction inhibited growth of both the ER+ MCF7 and ER-MDA-MB-453 human breast cancer cell lines with IC50 values of about 20 and 10 micro g/ml, respectively. It also induced cell cycle arrest at G1 when tested at 30 micro g/ml and at G2/M at 60 micro g/ml in MCF7 cells. This suggests that the extract contains a mixture of components with the more active (or more abundant) causing G1 arrest and the less active causing G2/M arrest. We then examined specific components in this extract. The triterpene glycoside fraction obtained by polyamide column chromatography, and the specific triterpene glycosides actein, 23-epi-26-deoxyactein and cimiracemoside A, inhibited growth of the MCF7 human breast cancer cells and induced cell cycle arrest at G1. The most potent compound, actein, decreased the level of cyclin D1, cdk4 and the hyperphosphorylated form of the pRb protein and increased the level of p21cip1 in MCF7 cells, changes that may contribute to the arrest in G1. Further studies are in progress to identify the mechanisms by which actein and related compounds present in black cohosh inhibit growth of human breast cancer cells.


Asunto(s)
Neoplasias de la Mama/prevención & control , Ciclo Celular/efectos de los fármacos , Cimicifuga/química , Fitoterapia , Extractos Vegetales/farmacología , Femenino , Humanos , Raíces de Plantas/química , Células Tumorales Cultivadas
6.
Cancer Res ; 63(20): 6825-37, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14583480

RESUMEN

Epidemiological and experimental carcinogenesis studies provide evidence that components of garlic (Allium sativum) have anticancer activity. We recently reported that the garlic derivative S-allylmercaptocysteine (SAMC) inhibits growth, arrests cells in G(2)-M, and induces apoptosis in human colon cancer cells (Shirin et al., Cancer Res., 61: 725-731, 2001). Because a fraction of the SAMC-treated cells are specifically arrested in mitosis, we examined the mechanism of this effect in the present study. Immunofluorescent microscopy revealed that the treatment of SW480 cells or NIH3T3 fibroblasts with 150 micro M SAMC (the IC(50) concentration) caused rapid microtubule (MT) depolymerization, MT cytoskeleton disruption, centrosome fragmentation and Golgi dispersion in interphase cells. It also induced the formation of monopolar and multipolar spindles in mitotic cells. In vitro turbidity assays indicated that SAMC acted directly on tubulin to cause MT depolymerization, apparently because it interacts with -SH groups on tubulin. To investigate the signaling pathways involved in SAMC-induced apoptosis, we assayed c-Jun NH(2)-terminal kinase (JNK) activity and found that treatment with SAMC caused a rapid and sustained induction of JNK activity. The selective JNK inhibitor SP600125 inhibited the early phase (24 h) but not the late phase (48 h and later) of apoptosis induced by SAMC. Expression of a dominant-negative mutant of JNK1 in SW480 cells inhibited apoptosis induced by SAMC at 24 h but had no protective effect at 48 h. JNK1(-/-) mouse embryonic fibroblasts were resistant to SAMC-induced apoptosis at 24 h but not at 48 h. On the other hand, the inhibition or abrogation of JNK1 activity did not inhibit the G(2)-M arrest induced by SAMC. SAMC also activated caspase-3. The general caspase inhibitor z-VAD-fmk inhibited both early and late phases of apoptosis induced by SAMC. We conclude that the garlic-derived compound SAMC exerts antiproliferative effects by binding directly to tubulin and disrupting the MT assembly, thus arresting cells in mitosis and triggering JNK1 and caspase-3 signaling pathways that lead to apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Cisteína/análogos & derivados , Cisteína/farmacología , Microtúbulos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Animales , Apoptosis/fisiología , Sitios de Unión , Caspasa 3 , Caspasas/metabolismo , Ciclo Celular/fisiología , Línea Celular Tumoral , Centrosoma/efectos de los fármacos , Centrosoma/fisiología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/fisiología , Humanos , Ratones , Microtúbulos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteína Quinasa 8 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Mitosis/efectos de los fármacos , Mitosis/fisiología , Células 3T3 NIH , Tubulina (Proteína)/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
7.
J Exp Ther Oncol ; 3(2): 83-94, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12822514

RESUMEN

We examined the effects of exisulind (sulindac sulfone) and a potent derivative CP248 on the Barrett's esophagus (BE)-related adenocarcinoma cell lines Seg-1 and Bic-1, and on HCE7 esophageal squamous carcinoma cells. Marked growth inhibition and apoptosis occurred in all cell lines with IC50 values of 100-300 microM for exisulind and 100 nM for CP248. Bic-1 and HCE7 cells were more sensitive to the growth inhibitory properties of exisulind. Treatment of all cell lines with CP248 for 24 h increased the proportion of cells in mitosis. Exisulind had no effect on cell-cycle progression. Treatment with either compound induced rapid activation of the c-Jun NH2-terminal kinase 1 (JNK1), suggesting that JNK1 activation plays a role in the induction of apoptosis by these compounds. Only Seg-1 cells expressed a detectable basal level of cyclooxygenase-2 (cox-2), providing further evidence that cox-2 is not the critical target for the growth inhibitory and apoptotic effects of these compounds. Cellular levels of reduced glutathione (GSH) increased approximately five-fold in all cell lines after 24 h of treatment with either compound. These studies provide support for the use of exisulind in BE chemoprevention trials, and of exisulind or CP248 in the therapy of patients with esophageal carcinoma.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Apoptosis , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/patología , Sulindac/uso terapéutico , Animales , Biomarcadores , Western Blotting , Ciclo Celular , División Celular/efectos de los fármacos , Ciclooxigenasa 2 , Relación Dosis-Respuesta a Droga , Glutatión/metabolismo , Humanos , Concentración 50 Inhibidora , Isoenzimas/biosíntesis , Isoenzimas/metabolismo , Proteínas de la Membrana , Proteína Quinasa 8 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mitosis , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Prostaglandina-Endoperóxido Sintasas/metabolismo , Sulindac/análogos & derivados , Factores de Tiempo , Células Tumorales Cultivadas
8.
Mol Cancer Ther ; 1(6): 393-404, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12477052

RESUMEN

Exisulind (sulindac sulfone) and two potent derivatives, CP248 and CP461, have been shown previously to cause growth inhibition and apoptosis in several types of human carcinoma cell lines. These and related compounds have not been previously studied with respect to glioma cell lines. In the present study, we found that these three compounds caused marked growth inhibition in four rat glioma and eight human glioma cell lines, with IC50 values of 150, 1, and 0.075 microm, respectively. When studied at these concentrations exisulind and CP461 had no significant effect on the cell cycle profile of glioma cells, but CP248 caused marked arrest in mitosis. Detailed studies of CP248 in the 9L rat gliosarcoma cell line indicated that treatment with 0.075 microM CP248 caused abnormalities in the spindle apparatus and activation of the spindle assembly check point. In interphase glioma cells, CP248 stabilized microtubules (MTs) at low concentrations (0.075 microM) and depolymerized MTs at higher concentrations (0.2-0.4 microM). In NIH 3T3 fibroblasts, 0.1 microM CP248 caused extensive MT depolymerization. CP248 also caused MT depolymerization when added to assembled MTs in vitro, which indicated that it can directly affect MTs, perhaps because it shares certain structural similarities with Colcemid. In glioma cells, the effects of CP248 on MTs were independent of the previously reported effects of this compound on activation of protein kinase G. Therefore, CP248 is a novel MT-active agent that may be useful in the treatment of glioblastoma, and possibly other types of cancer, because of its dual effects on protein kinase G and MTs.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/patología , Proteínas de Ciclo Celular , Glioma/patología , Microtúbulos/efectos de los fármacos , Huso Acromático/efectos de los fármacos , Sulindac/análogos & derivados , Sulindac/farmacología , 3',5'-AMP Cíclico Fosfodiesterasas/metabolismo , 3',5'-GMP Cíclico Fosfodiesterasas , Células 3T3/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , División Celular/efectos de los fármacos , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2 , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Citometría de Flujo , Técnica del Anticuerpo Fluorescente Indirecta , Glioma/tratamiento farmacológico , Glioma/metabolismo , Humanos , Técnicas para Inmunoenzimas , Técnicas In Vitro , Interfase/efectos de los fármacos , Cinesinas , Ratones , Fosfoproteínas/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Ratas , Timidina/metabolismo
9.
Clin Cancer Res ; 8(3): 893-903, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11895924

RESUMEN

PURPOSE: We examined the effects of the phytochemical resveratrol in six human cancer cell lines (MCF7, SW480, HCE7, Seg-1, Bic-1, and HL60). EXPERIMENTAL DESIGN AND RESULTS: Resveratrol induced marked growth inhibition in five of these cell lines, with IC(50) values of approximately 70-150 microM. However, only partial growth inhibition was seen in Bic-1 cells. After treatment with 300 microM resveratrol for 24 h, most of the cell lines were arrested in the S phase of the cell cycle. In addition, induction of apoptosis was demonstrated by the appearance of a sub-G(1) peak and confirmed using an annexin V-based assay. Cyclin B1 expression levels were decreased in all cell lines after 48 h of treatment. In SW480 cells, cyclin A, cyclin B1, and beta-catenin expression levels were decreased within 24 h. There was a decrease in cyclin D1 expression after only 2 h of treatment, and this persisted for up to 48 h. This decrease was partially blocked by concurrent treatment with the proteasome inhibitor calpain inhibitor I. Using a luciferase-based reporter assay, resveratrol did not inhibit cyclin D1 promoter activity in SW480 cells. Furthermore, using a reverse transcription-PCR-based assay, only a higher dose of resveratrol (300 microM) appeared to decrease cyclin D1 mRNA. Seg-1 cells expressed basal levels of cyclooxygenase-2 (cox-2), which was further induced by resveratrol. Neither basal levels nor induction of cox-2 was detectable in the remaining cell lines. Thus, cox-2 does not appear to be a critical target of this compound. CONCLUSIONS: These studies provide support for the use of resveratrol in chemoprevention and cancer therapy trials. Cyclin D1, cyclin B1, beta-catenin, and apoptotic index could be useful biomarkers to evaluate treatment efficacy.


Asunto(s)
Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Ciclina D1/metabolismo , Fase S/efectos de los fármacos , Estilbenos/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Ciclina D1/genética , Ciclinas/metabolismo , Cartilla de ADN/química , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Immunoblotting , Luciferasas/metabolismo , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Resveratrol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleótido Reductasas/antagonistas & inhibidores , Células Tumorales Cultivadas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...