Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Med Rep ; 17(4): 5258-5264, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29363716

RESUMEN

Potentilla discolor has been used in traditional Chinese medicine for the treatment of hyperglycemia. However, the potential role of Potentilla discolor against cancer and its mode of action remain to be fully elucidated. The present study explored the apoptotic effect of methanol extract of Potentilla discolor (MEPD) in human mucoepidermoid carcinoma (MEC) cell lines of salivary glands. MEPD markedly suppressed the growth and induced apoptotic cell death in MC3 and YD15 cells. MEPD treatment significantly upregulated the expression of PUMA and reduced STAT3 phosphorylation. Overexpression of STAT3 partially recovered the growth of MEC cells inhibited by MEPD. In addition, dephosphorylation of STAT3 by cryptotanshinone (a potent STAT3 inhibitor) was sufficient to inhibit the growth of MEC cells and induce apoptosis via affecting PUMA protein. These results suggest that MEPD has a potential anticancer property via the STAT3/PUMA signaling axis in human MEC cells of salivary gland.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Carcinoma Mucoepidermoide/metabolismo , Extractos Vegetales/farmacología , Potentilla/química , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos
2.
Cell Oncol (Dordr) ; 40(3): 235-246, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28401485

RESUMEN

PURPOSE: Approximately 20% of all salivary gland cancer patients who are treated with current treatment modalities will ultimately develop metastases. Its most common form, mucoepidermoid carcinoma (MEC) is a highly aggressive tumor with an overall 5-year survival rate of ~30%. Until now, several chemotherapeutic drugs have been tested for the treatment of salivary gland tumors, but the results have been disappointing and the drugs often cause unwanted side effects. Therefore, several recent studies have focused on the potential of alternative and/or complementary therapeutic options, including the use of silymarin. METHODS: The effects of silymarin and its active component silibinin on salivary gland cancer-derived MC3 and HN22 cells and their underlying molecular mechanisms were examined using trypan blue exclusion, 4'-6-diamidino-2-phenylindole (DAPI) staining, Live/Dead, Annexin V/PI staining, mitochondrial membrane potential (ΔΨm) measurement, quantitative RT-PCR, soft agar colony formation and Western blotting analyses. RESULTS: We found that silymarin and silibinin dramatically increased the expression of the pro-apoptotic protein Bim in a concentration- and time-dependent manner and, concomitantly, induced apoptosis in MC3 and HN22 cells. We also found that ERK1/2 signaling inhibition successfully sensitized these cells to the apoptotic effects of silymarin and silibinin, which indicates that the ERK1/2 signaling pathway may act as an upstream regulator that modulates the silymarin/silibinin-induced Bim signaling pathway. CONCLUSIONS: Taken together, we conclude that ERK1/2 signaling pathway inhibition by silymarin and silibinin increases the expression of the pro-apoptotic Bcl-2 family member Bim which, subsequently, induces mitochondria-mediated apoptosis in salivary gland cancer-derived cells.


Asunto(s)
Proteína 11 Similar a Bcl2/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias de las Glándulas Salivales/patología , Silimarina/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Silibina
3.
Cell Biochem Funct ; 32(3): 229-35, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24037733

RESUMEN

In the present study, we examined the effects of methanol extracts of Picrasma quassioides (MEPQ) on apoptosis in human cervical cancer cells. The results showed that MEPQ decreased the viability and induced caspase-dependent apoptosis in HEp-2 cells. MEPQ decreased specificity protein 1 (Sp1) in HEp-2 cells, whereas Sp1 mRNA was not changed. We found that MEPQ reduced Sp1 protein through proteasome-dependent protein degradation, but not the inhibition of protein synthesis. Also, MEPQ increased the expressions of Bad and truncated Bid (t-Bid) but did not alter other Bcl-2 family members. The knock-down of Sp1 by both Sp1 interfering RNA and Mithramycin A, Sp1 specific inhibitor clearly increased Bad and t-Bid expression to decrease cell viability and induce apoptosis. In addition, MEPQ inhibited cell viability and induced apoptotic cell death through the modulation of Sp1 in KB cells. These results suggest that MEPQ may be a potential anticancer agent for human cervical cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Picrasma , Extractos Vegetales/farmacología , Factor de Transcripción Sp1/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Metanol , Plicamicina/análogos & derivados , Plicamicina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Interferente Pequeño/genética , Solventes , Factor de Transcripción Sp1/genética , Neoplasias del Cuello Uterino/patología , Proteína Letal Asociada a bcl/metabolismo
4.
Head Neck ; 35(7): 992-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22733715

RESUMEN

BACKGROUND: The aims of this study were to evaluate the apoptotic activities and molecular mechanisms of methanol extracts of Dianthus chinensis (MEDC) and Acalypha australis L. (MEAL) in human oral cancer cells. METHODS: The apoptotic effects and related molecular mechanisms of MEDC and MEAL on oral cancer cells were evaluated using MTS assay, DAPI staining, immunostaining, Western blotting, and reverse transcriptase-polymerase chain reaction. RESULTS: Sp1 was overexpressed in oral tumor tissues compared with normal oral mucosa. Downregulation of Sp1 inhibited the growth of SCC-15 and YD-15 oral cancer cells. MEDC and MEAL inhibited cell growth and induced apoptosis in both cell lines by decreasing the expression of Sp1. In addition, treatment of cells with MEDC and MEAL decreased Mcl-1 expression, which is a downstream target of Sp1. CONCLUSION: Our results indicate that MEDC and MEAL are bioactive natural products that can potentially induce apoptosis of tumor cells that overexpress the Sp1 protein.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Dianthus/química , Euphorbiaceae/química , Neoplasias de la Boca/patología , Extractos Vegetales/farmacología , Factor de Transcripción Sp1/genética , Adulto , Western Blotting , Carcinoma de Células Escamosas/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Metanol/química , Neoplasias de la Boca/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción Sp1/metabolismo , Células Tumorales Cultivadas
5.
Mol Med Rep ; 6(4): 843-7, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22767187

RESUMEN

For thousands of years in Asia, Althaea rosea Cavanil (ARC) and Plantago major L. (PML) have been used as powerful non-toxic therapeutic agents that inhibit inflammation. However, the anticancer mechanisms and molecular targets of ARC and PML are poorly understood, particularly in epidermal growth factor (EGF)-induced neoplastic cell transformation. The aim of this study was to evaluate the chemopreventive effects and mechanisms of the methanol extracts from ARC (MARC) and PML (MPML) in EGF-induced neoplastic cell transformation of JB6 P+ mouse epidermal cells using an MTS assay, anchorage-independent cell transformation assay and western blotting. Our results showed that MARC and MPML significantly suppressed neoplastic cell transformation by inhibiting the kinase activity of the EGF receptor (EGFR). The activation of EGFR by EGF was suppressed by MARC and MPML treatment in EGFR(+/+) cells, but not in EGFR(-/-) cells. In addition, MARC and MPML inhibited EGF-induced cell proliferation in EGFR-expressing murine embryonic fibroblasts (EGFR(+/+)). These results strongly indicate that EGFR targeting by MARC and MPML may be a good strategy for chemopreventive or chemotherapeutic applications.


Asunto(s)
Althaea/química , Transformación Celular Neoplásica/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Extractos Vegetales/farmacología , Plantago/química , Animales , Línea Celular , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Metanol/química , Ratones , Fosforilación , Extractos Vegetales/química
6.
Mol Med Rep ; 6(3): 670-4, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22710351

RESUMEN

Sanguisorba officinalis is a natural plant that has been traditionally used for the treatment of inflammatory and metabolic diseases. Several studies have reported that its extracts exhibit anticancer, antioxidative and anti-lipid peroxidation activities. However, the effects of this plant on human prostate cancer cells have not yet been investigated. In the present study, we investigated the inhibitory effects and underlying mechanisms of a methanol extract of Sanguisorba officinalis (MESO) in PC3 human prostate cancer cells. MESO significantly decreased cell growth and induced apoptosis through the intrinsic apoptosis pathway. MESO decreased the expression levels of myeloid cell leukemia-1 (Mcl-1), a Bcl­2­like anti-apoptotic protein that is highly expressed in various cancer cell lines. Expression levels of the pro-apoptotic protein Bax were increased by MESO whereas those of Bak and Bcl-xL were unchanged. In addition, MESO induced the oligomerization of Bax in the mitochondrial outer membrane. These results suggest that MESO inhibits the growth of prostate cancer cells and induces apoptotic cell death by the downregulation of Mcl-1 protein expression and the oligomerization of Bax. Therefore, MESO has potential as a drug candidate for the treatment of prostate cancer.


Asunto(s)
Metanol/química , Extractos Vegetales/toxicidad , Sanguisorba/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Humanos , Masculino , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Extractos Vegetales/química , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína bcl-X/metabolismo
7.
Oncol Lett ; 4(3): 489-494, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23741248

RESUMEN

Sanguisorba officinalis L. has been used in traditional Asian medicine to treat diseases including diarrhea, chronic intestinal infections, duodenal ulcers and bleeding. This study examined the antiproliferative effects and apoptotic activity of hot water extract of S. officinalis L. (HESO) on HSC4 and HN22 human oral cancer cells. The effects of HESO were evaluated by the 3-(4,5-dimethylthiazol-20yl)-(3-carboxymethoxyphenyl)-2-(4-sulphophenyl)-2H-tetrazolium (MTS) assay, 4'-6-diamidino-2-phenylindole (DAPI) staining and western blot analysis. HESO was found to inhibit cell growth and induce apoptosis in HSC4 and HN22 oral cancer cells. HESO downregulated myeloid cell leukemia-1 (Mcl-1) in HSC4 cells and was associated with the activation of Bak, resulting in Bak oligomerization on the mitochondrial outer membrane. HESO did not alter Mcl-1 expression in HN22 cells, but it decreased Sp1 expression. The downregulation of Sp1 by HESO in HN22 cells resulted in a decrease in survivin, a downstream target protein of Sp1. These results suggested that HESO inhibited the growth of oral cancer through either Mcl-1 or Sp1, indicating that HESO may serve as a potential drug candidate against oral cancer.

8.
J Agric Food Chem ; 59(15): 8124-31, 2011 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-21702500

RESUMEN

Cruciferous vegetables have been shown to have the possibility to protect against multistep carcinogenesis. ß-Phenylethyl isothiocyanate (PEITC) is one component of these vegetables demonstrated to help fight many types of cancer. The present study examined the apoptotic effects of PEITC and its molecular mechanism in human cervical cancer cell lines (HEp-2 and KB). PEITC induced apoptosis to inhibit cell proliferation. According to the protein chip assay, PEITC increased the expression of the death receptors (DR4 and DR5) and cleaved caspase-3 compared to the DMSO treatment group. PEITC also induced caspase-8 and truncated BID. PEITC down-regulated the phosphorylation of extracellular-related kinase (ERK)1/2, whereas neither phospho-c-Jun NH(2)-terminal kinases (JNK) nor phospho-p38 MAPK was changed. The role of ERK in PEITC-induced apoptosis was also investigated using MEK inhibitor (PD98059). PD98059 increased the expression of DR4 and DR5, activated caspase-3, and cleaved PARP. In addition, PEITC decreased the phosphorylation of MEK. Therefore, the apoptotic mechanism of PEITC in cervical cancer cells involves the induction of DR4 and DR5 through the inactivation of ERK and MEK.


Asunto(s)
Anticarcinógenos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Isotiocianatos/farmacología , Extractos Vegetales/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Neoplasias del Cuello Uterino/fisiopatología , Brassicaceae/química , Línea Celular Tumoral , Femenino , Humanos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Verduras/química
9.
Eur J Cancer Prev ; 19(1): 23-30, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19910795

RESUMEN

In a previous study, we demonstrated that cyclooxygenase-2 (COX-2) is overexpressed in Korean patients having oral cancer. The goal of this study was to study whether KO-202125 (KO), a sauristolactam derivative in KB human oral squamous carcinoma cells, inhibits the activity of COX-2 enzyme and induces apoptotic cell death. In this study, it was shown that KO inhibited COX-2 mRNA and protein and its catalytic activity (prostaglandin E2), but not COX-1. The antiproliferative effect of KO on KB cells was also examined. The results showed that KO significantly decreased the number of viable cells and showed morphological changes in a concentration-dependent manner. The decrease in cell number was associated with apoptotic cell death evidenced by cleaved poly ADP ribose polymerase (PARP), nuclear fragmentation, sub-G1 population and annexin V positivity. Interestingly, KO is more potent than celecoxib, which is a well-known selective COX-2 inhibitor, although more studies are needed to prove it. Altogether, these results show that KO can act as a potent antioral cancer drug candidate by regulating COX-2 activity.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Ciclooxigenasa 2/genética , Isoindoles/farmacología , Neoplasias de la Boca/patología , Alcaloides/química , Alcaloides/farmacología , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Celecoxib , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Células KB , Lactamas/química , Lactamas/farmacología , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Fenantrenos/química , Fenantrenos/farmacología , Pirazoles/farmacología , Sulfonamidas/farmacología
10.
Biochim Biophys Acta ; 1726(3): 309-16, 2005 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-16213662

RESUMEN

Many studies have suggested that dietary flavonoids are anticancer agents that induce the apoptosis of cancer cells. However, the effects of flavonoids on the induction of apoptosis in osteosarcoma cells are unclear. Previously, a flavonoid fraction, consisting mainly of protocatechuic acid, fustin, fisetin, sulfuretin, and butein, herein named RCMF (the RVS chloroform-methanol fraction), was prepared from a crude acetone extract of Rhus verniciflua Stokes (RVS). This study evaluated the effects of RCMF on the proliferation and apoptosis using human osteosarcoma (HOS) cells. The mechanism of growth inhibition of the HOS cells by the flavonoid fraction, RCMF, was also assessed. The results demonstrated that RCMF exhibited sensitive growth inhibition and induced apoptosis in HOS cells. PARP cleavage was closely associated with the RCMF-induced apoptosis of the HOS cells. Furthermore, the activation of caspase 8 and Bax, the inhibition of Bcl-2 expression, and the release of cytochrome c are believed to be involved in the RCMF-mediated apoptosis. Collectively, these findings suggest that RCMF is an agent which may be capable of inducing sensitive growth inhibition and apoptosis in HOS cells.


Asunto(s)
Anticarcinógenos/toxicidad , Apoptosis , Flavonoides/toxicidad , Anticarcinógenos/aislamiento & purificación , Neoplasias Óseas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Flavonoides/aislamiento & purificación , Humanos , Mitocondrias/metabolismo , Osteosarcoma/patología , Extractos Vegetales/toxicidad , Poli(ADP-Ribosa) Polimerasas/metabolismo , Rhus/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA