Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Phytomedicine ; 82: 153465, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33486268

RESUMEN

BACKGROUND: Ovarian cancer is one of the most common gynecological malignancies in the world. Daphnetin (Daph) was previously reported to possess antitumor potential, but its potential and molecular mechanisms in ovarian cancer remain poorly understood. PURPOSE: In the current study, we aimed to explore the antitumor effect and detailed mechanisms of Daph in ovarian cancer cells. METHODS: The cytotoxic effect of Daph on ovarian cells was determined in vitro and in vivo. Cell growth, proliferation, apoptosis and ROS generation were measured by CCK8 assays, colony formation assays and flow cytometry. Western blotting was used to evaluate the related signal proteins. Immunofluorescence and transmission electron microscopy were used to evaluate markers of autophagy and autophagic flux. The antitumor effects were observed in the A2780 xenograft model. Moreover, Daph-induced autophagy was observed by enhanced LC3-II accumulation and endogenous LC3 puncta, and an autophagy inhibitor further enhanced the antitumor efficacy of Daph, which indicated that the cytoprotective role of autophagy in ovarian cancer. RESULTS: We found that Daph exhibited antitumor effects by inducing ROS-dependent apoptosis in ovarian cancer, which could be reversed by N-acetyl cysteine (NAC). The AMPK/Akt/mTOR pathway was involved in Daph-mediated cytoprotective autophagy, and when Daph-mediated the expression level of AMPK and autophagy were blocked, there was robust inhibition of cell proliferation and induction of apoptosis. In addition, in the A2780 xenograft model, combined treatment with Daph and an autophagy inhibitor showed obvious synergetic effects on the inhibition of cell viability and promotion of apoptosis, without any side effects. CONCLUSION: Our results suggest that Daph triggers ROS-induced cell apoptosis and induces cytoprotective autophagy by modulating the AMPK/Akt/mTOR pathway. Moreover, the combination of Daph and autophagy inhibitor may be a potential therapeutic strategy for ovarian cancer.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia/efectos de los fármacos , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Umbeliferonas/farmacología , Apoptosis/efectos de los fármacos , Muerte Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/metabolismo , Transducción de Señal/efectos de los fármacos
2.
Clin Adv Hematol Oncol ; 18(9): 550-556, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33006584

RESUMEN

Poly(ADP-ribose) polymerase (PARP) inhibitors have been rapidly integrated into clinical practice for women with ovarian cancer. Currently, PARP inhibitors are approved as frontline maintenance treatment for patients with and without BRCA-associated cancers, and they are listed by the National Comprehensive Cancer Network (NCCN) as a treatment option for all high-grade serous and endometrioid cancers with or without bevacizumab. PARP inhibitors are also approved as maintenance treatment following a response to platinum-based therapy in the recurrent setting, irrespective of biomarker status. Additionally, PARP inhibitors are approved as third-line treatment and beyond in lieu of chemotherapy for patients with BRCA-associated cancers, and as fourth-line treatment and beyond for patients with platinum-sensitive homologous recombination-deficient tumors. They are also listed by the NCCN in combination with bevacizumab for the treatment of patients who have platinum-sensitive recurrent disease. The first part of this 2-part review focuses on the changing paradigm of frontline therapy options resulting from the recent approvals of PARP inhibitors; the second part considers the role of PARP inhibition in recurrent ovarian cancer.


Asunto(s)
Bevacizumab/uso terapéutico , Carcinoma Epitelial de Ovario , Proteínas de Neoplasias , Neoplasias Ováricas , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Poli(ADP-Ribosa) Polimerasas/metabolismo , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/enzimología , Carcinoma Epitelial de Ovario/patología , Femenino , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología
3.
Free Radic Res ; 53(8): 882-891, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31290351

RESUMEN

To define the role of glutathione peroxidase (GPx) in modulating the oxygen radical-related cytotoxicity of doxorubicin and H2O2 in cells that overexpress P-glycoprotein (Pgp), the GPx activity of NCI/ADR-RES cancer cells was altered by growth in 0.5% serum with (MR-30 subline) or without (MR-0 subline) selenium supplementation. GPx activity increased from 2.2 nmol/min/mg (MR-0) to 22.5 nmol/min/mg (MR-30) when cells were grown in 30-nM selenium, p < .01; the activities of other antioxidant enzymes were unchanged by selenium. By reverse transcriptase polymerase chain reaction, MR-30 and MR-0 cells expressed similar levels of the MDR1, GPx-1, BCL2 and TOP2A mRNA. The IC50 concentration for H2O2 in MR-0 cells was 10-fold lower than in the MR-30 subline, p < .01. Despite identical anthracycline accumulation and efflux in these two lines that expressed equivalent levels of Pgp, the doxorubicin IC50 decreased fivefold in MR-0 versus MR-30 cells, p < .01. Log-linear tumour cell killing by doxorubicin was observed only in selenium-deficient MR-0 cells. Doxorubicin exposure also produced substantially more apoptosis in MR-0 than MR-30 cells; this was not related to the presence of selenium per se. MR-0 cells generated ≈5-times more methane from dimethyl sulfoxide (a measure of reactive oxygen metabolism) than MR-30 cells in the presence of equimolar doxorubicin concentrations (p < .05). These studies suggest that GPx-mediated detoxification of peroxides can modulate the antitumor activity of doxorubicin in the presence of high levels of Pgp.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Apoptosis , Doxorrubicina/farmacología , Glutatión Peroxidasa/metabolismo , Neoplasias Ováricas/enzimología , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Peróxido de Hidrógeno/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/fisiopatología , Peróxidos/metabolismo , Células Tumorales Cultivadas
4.
Redox Biol ; 25: 101051, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30509602

RESUMEN

Ovarian cancer remains the most lethal gynecologic malignancy, and is primarily diagnosed at late stage when considerable metastasis has occurred in the peritoneal cavity. At late stage abdominal cavity ascites accumulation provides a tumor-supporting medium in which cancer cells gain access to growth factors and cytokines that promote survival and metastasis. However, little is known about the redox status of ascites, or whether antioxidant enzymes are required to support ovarian cancer survival during transcoelomic metastasis in this medium. Gene expression cluster analysis of antioxidant enzymes identified two distinct populations of high-grade serous adenocarcinomas (HGSA), the most common ovarian cancer subtype, which specifically separated into clusters based on glutathione peroxidase 3 (GPx3) expression. High GPx3 expression was associated with poorer overall patient survival and increased tumor stage. GPx3 is an extracellular glutathione peroxidase with reported dichotomous roles in cancer. To further examine a potential pro-tumorigenic role of GPx3 in HGSA, stable OVCAR3 GPx3 knock-down cell lines were generated using lentiviral shRNA constructs. Decreased GPx3 expression inhibited clonogenicity and anchorage-independent cell survival. Moreover, GPx3 was necessary for protecting cells from exogenous oxidant insult, as demonstrated by treatment with high dose ascorbate. This cytoprotective effect was shown to be due to GPx3-dependent removal of extracellular H2O2. Importantly, GPx3 was necessary for clonogenic survival when cells were cultured in patient-derived ascites fluid. While oxidation reduction potential (ORP) of malignant ascites was heterogeneous in our patient cohort, and correlated positively with ascites iron content, GPx3 was required for optimal survival regardless of ORP or iron content. Collectively, our data suggest that HGSA ovarian cancers cluster into distinct groups of high and low GPx3 expression. GPx3 is necessary for HGSA ovarian cancer cellular survival in the ascites tumor environment and protects against extracellular sources of oxidative stress, implicating GPx3 as an important adaptation for transcoelomic metastasis.


Asunto(s)
Progresión de la Enfermedad , Espacio Extracelular/metabolismo , Glutatión Peroxidasa/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Antioxidantes/metabolismo , Ascitis/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular , Células Clonales , Femenino , Humanos , Peróxido de Hidrógeno/toxicidad , Estadificación de Neoplasias , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos
5.
Molecules ; 23(8)2018 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-30044423

RESUMEN

Two tetrahydrofurofurano lignans (1 and 2), four phenylpropanoids (3⁻6), and two alkamides (7 and 8) were isolated from the EtOAc-soluble fraction of the roots of Asarum sieboldii. The chemical structures of the isolates were identified by analysis of spectroscopic data measurements, and by a comparison of their data with published values. The isolates (1, 2, 4⁻8) were evaluated for their cytotoxicity against human ovarian cancer cells (A2780 and SKOV3) and immortalized ovarian surface epithelial cells (IOSE80PC) using a MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide) assay. Of the isolates, (-)-asarinin (1) exhibited the most potent cytotoxicity to both A2780 and SKOV3 cells. A propidium iodide/annexin V-fluorescein isothiocyanate (V-FITC) double staining assay showed that (-)-asarinin (1) induces apoptotic cell death in ovarian cancer cells. In addition, (-)-asarinin (1) increased the activation of caspase-3, caspase-8, and caspase-9 in ovarian cancer cells. Pretreatment with caspase inhibitors attenuated the cell death induced by (-)-asarinin (1). In conclusion, our findings show that (-)-asarinin (1) from the roots of A. sieboldii may induce caspase-dependent apoptotic cell death in human cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Asarum/química , Caspasas/metabolismo , Dioxoles/farmacología , Lignanos/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Extractos Vegetales/farmacología , Raíces de Plantas/química , Antineoplásicos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas/farmacología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Dioxoles/aislamiento & purificación , Activación Enzimática , Femenino , Humanos , Lignanos/aislamiento & purificación , Estructura Molecular , Neoplasias Ováricas/enzimología , Relación Estructura-Actividad
6.
J Tradit Chin Med ; 36(3): 353-9, 2016 Jun.
Artículo en Chino | MEDLINE | ID: mdl-27468551

RESUMEN

OBJECTIVE: To explore the effects of evodiamine on ovarian cancer cells and the mechanisms underlying such effects. METHODS: Human. ovarian cancer cells HO-8910PM were treated with evodiamine at 0, 1.25, 2.5, and 5 µM for 1-4 d. 3-(4,5-Dimethiylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to detect the growth inhibition rate of evodiamine-treated HO-8910PM cells. The cell cycle was observed via propidium iodide (PI) staining. Apoptosis induction was assessed via Annexin V-fluorescein isothiocyanate/propidium iodide (Annexin V-FITC/PI) double staining assay. To verify the mechanism of apoptosis, caspase-dependent apoptotic pathway-related protein was detected by Western blot analysis. The expression levels of mitogen-activated protein kinase (MAPK) and/or phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) pathway-related proteins were also investigated. RESULTS: Evodiamine significantly inhibited the proliferation of HO-8910PM cells in a dose- and time-dependent manner. Evodiamine induced G2/M arrest with an increase of cyclin B1 level, and promoted cell apoptosis with a decrease of B cell lymphoma/lewkmia-2 (Bcl-2) and an increase of Bcl-2-associated X protein (Bax) level. In addition, evodiamine treatment led to the activation of caspase-8, caspase-9, and caspase-3 and the cleavage of poly (ADP-ribose)-polymerase (PARP). Evodiamine targeted the MAPK and/or PI3K/Akt pathways by reducing the expression and activity of PI3K, Akt, and extracellular signal-regulated kinase mitogen-activated protein kinase (ERK1/2 MAPK) and the activity of p38 MAPK. CONCLUSION: Evodiamine can inhibit the growth of ovarian cancer cells by G2/M arrest and intrinsic and extrinsic apoptosis. In addition, evodiamine-induced PI3K/Akt, ERK1/2 MAPK, and p38 MAPK signaling may be involved in cell death.


Asunto(s)
Apoptosis/efectos de los fármacos , Evodia/química , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias Ováricas/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Medicamentos Herbarios Chinos , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética
7.
Biomed Pharmacother ; 83: 130-140, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27470560

RESUMEN

Over the last four centuries, cocoa and chocolate have been described as having potential medicinal value. As of today, Theobroma cacao L. (Sterculiaceae) and its products are consumed worldwide. They are of great research interest because of the concentration dependent antioxidant as well as pro-oxidant properties of some of their polyphenolic constituents, specially procyanidins and flavan-3-ols such as catechin. This study was aimed at investigating the cellular and molecular changes associated with cytotoxicity, caused due pro-oxidant activity of cocoa catechins and procyanidins, in ovarian cancer cell lines. Extract of non-alkalized cocoa powder enriched with catechins and procyanidins was used to treat human epithelial ovarian cancer cell lines OAW42 and OVCAR3 at various concentrations ≤1000µg/mL. The effect of treatment on intracellular reactive oxygen species (ROS) levels was determined. Apoptotic cell death, post treatment, was evaluated microscopically and using flow cytometry by means of annexin-propidium iodide (PI) dual staining. Levels of active caspase-3 as a pro-apoptotic marker and matrix metalloproteinase 2 (MMP2) as an invasive potential marker were detected using Western blotting and gelatin zymography. Treatment with extract caused an increase in intracellular ROS levels in OAW42 and OVCAR3 cell lines. Bright field and fluorescence microscopy of treated cells revealed apoptotic morphology and DNA damage. Increase in annexin positive cell population and dose dependent upregulation of caspase-3 confirmed apoptotic cell death. pro-MMP2 was found to be downregulated in a dose dependent manner in cells treated with the extract. Treated cells also showed a reduction in MMP2 activity. Our data suggests that cocoa catechins and procyanidins are cytotoxic to epithelial ovarian cancer, inducing apoptotic morphological changes, DNA damage and caspase-3 mediated cell death. Downregulation of pro-MMP2 and reduction in active MMP2 levels imply a decrease in invasive potential of the cells. Apoptosis and MMP2 downregulation appear to be linked to the increase in intracellular ROS levels, caused due to the prooxidant effect of cocoa procyanidin extract.


Asunto(s)
Apoptosis , Biflavonoides/uso terapéutico , Cacao/química , Caspasa 3/metabolismo , Catequina/uso terapéutico , Metaloproteinasa 2 de la Matriz/metabolismo , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Extractos Vegetales/uso terapéutico , Proantocianidinas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Biflavonoides/farmacología , Western Blotting , Carcinoma Epitelial de Ovario , Catequina/farmacología , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Forma de la Célula/efectos de los fármacos , Femenino , Humanos , Espacio Intracelular/metabolismo , Neoplasias Glandulares y Epiteliales/enzimología , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Extractos Vegetales/farmacología , Polvos , Proantocianidinas/farmacología
8.
Pharm Biol ; 54(10): 2026-32, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26911246

RESUMEN

Context Curcumin is widely used in China and India as a traditional herb but additional work is required to ascertain the folkloric claim of its antitumour and antioxidant activities. Objective The present study determines the antitumour effect of curcumin against SKOV3 cell growth. Materials and methods SKOV3 cells were incubated with curcumin (0, 20, 30 and 40 µM) for 72 h. The antiproliferative activity and the apoptosis rate were measured by MTT and flow cytometry. Expression of PI3K, T-Akt and p-Akt proteins was measured by western blotting. Results The administration of curcumin (0, 20, 30 and 40 µM) inhibits SKOV3 cell growth (IC50 value= 24.8 µM) and increased apoptosis (32.5 and 85.7%). The activity of SKOV3 cell invasion (98.2 and 19.4%) was also decreased by curcumin administration (p < 0.05). Results of western blot analysis confirmed that the expression of p-Akt protein was decreased by curcumin (p < 0.05). It was also found that a high dose of curcumin (40 µM) can cause stronger antitumour activity (80.4%). Conclusion Our results suggest that the curcumin induced SKOV3 apoptosis via modulation of the PI3K/Akt-signalling pathway.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Curcumina/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Invasividad Neoplásica , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Fosforilación , Factores de Tiempo
9.
Acta Biochim Biophys Sin (Shanghai) ; 48(4): 301-17, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26764240

RESUMEN

The development and strategic application of effective anticancer therapies have turned out to be one of the most critical approaches of managing human cancers. Nevertheless, drug resistance is the major obstacle for clinical management of these diseases especially ovarian cancer. In the past years, substantial studies have been carried out with the aim of exploring alternative therapeutic approaches to enhance efficacy of current chemotherapeutic regimes and reduce the side effects caused in order to produce significant advantages in overall survival and to improve patients' quality of life. Targeting cancer cell metabolism by the application of AMP-activated protein kinase (AMPK)-activating agents is believed to be one of the most plausible attempts. AMPK activators such as 5-aminoimidazole-4-carboxamide 1-ß-d-ribofuranoside, A23187, metformin, and bitter melon extract not only prevent cancer progression and metastasis but can also be applied as a supplement to enhance the efficacy of cisplatin-based chemotherapy in human cancers such as ovarian cancer. However, because of the undesirable outcomes along with the frequent toxic side effects of most pharmaceutical AMPK activators that have been utilized in clinical trials, attentions of current studies have been aimed at the identification of replaceable reagents from nutraceuticals or traditional medicines. However, the underlying molecular mechanisms of many nutraceuticals in anticancer still remain obscure. Therefore, better understanding of the functional characterization and regulatory mechanism of natural AMPK activators would help pharmaceutical development in opening an area to intervene ovarian cancer and other human cancers.


Asunto(s)
Adenilato Quinasa/metabolismo , Neoplasias Ováricas/prevención & control , Transducción de Señal , Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/metabolismo , Compuestos de Bifenilo , Activadores de Enzimas/farmacología , Femenino , Humanos , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/metabolismo , Pironas/farmacología , Tiofenos/farmacología
10.
Phytother Res ; 27(7): 1036-41, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22949403

RESUMEN

In the present study we analysed the effects of isothiocyanates (ITCs)--plant-derived sulphur-containing constituents known for their potential chemotherapeutic activity--on growth inhibition and programmed death in primary ovarian carcinoma cells from ascites of human patients. Twenty-four hour exposure of carcinoma cells to 5-50 µM erucin or benzyl ITC led to a concentration-dependent viability loss, as determined by erytrosin B cell staining. This concurred with an increase in internucleosomal DNA fragmentation, mitochondrial membrane depolarization and downregulation of Akt as indicator for apoptosis induction. Cell accumulation at the G2/M phase was evident after 48 h of erucin treatment. Telomerase, a selective target of cancer cells, was suppressed by erucin. Although pre-treatment of cells with the thiol antioxidant N-acetylcysteine could completely prevent initialization of the apoptotic process, it failed to abolish ITC-mediated telomerase suppression. Taken together, in our study, ITC exerted comparable cytotoxic efficacy against primary ovarian cancer cells as reported for corresponding cell lines. The clinical significance of this observation should be addressed in future studies and the role of telomerase further investigated.


Asunto(s)
Apoptosis/efectos de los fármacos , Isotiocianatos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Sulfuros/uso terapéutico , Telomerasa/efectos de los fármacos , Tiocianatos/uso terapéutico , Acetilcisteína/farmacología , Apoptosis/fisiología , Línea Celular Tumoral , Supervivencia Celular , Fragmentación del ADN , Regulación hacia Abajo , Femenino , Fase G2 , Humanos , Membranas Mitocondriales/efectos de los fármacos , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo
11.
Biosci Trends ; 6(4): 201-11, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23006967

RESUMEN

Rhizoma Paridis (dried root and rhizome) has been an essential ingredient in traditional Chinese herbal medicine. In the past decade, active components of Rhizoma Paridis - the Paris saponins have emerged as promising anti-cancer agents. Among these saponins, polyphyllin D (Paris saponin (PS) I), has been extensively studied and proposed to be a potent antitumor agent. In this study, we continue to establish the efficacy and mechanisms underlying the cytotoxic effects of the steroidal PS members, namely formosanin C (PSII) in ovarian cancer treatment. We isolated PSII and evaluated its effects on a panel of ten human cell lines. Isolated PSII has potent inhibitory effects on the growth of tumor cells without deleterious effects to different normal cell types or benign neoplastic derived cells. While PSII, PSI, and etoposide are effective promoting agents for cell cycle arrest and apoptosis, PSII appeared to be marginally more potent than the later two in inhibiting SKOV3 cell growth. In PSII-treated SKOV3 cells, there was an elevation in proapoptotic elements including Bax, cytosolic cytochrome c, activated-caspase-3, and activated-caspase-9. The treatment also reduced extracellular signal-regulated kinase (ERK1/2) phosphorylation and anti-apoptotic Bcl-2 expression. We also assessed the antitumor efficacy of intraperitoneal administration of PSII in human SKOV3 ovarian cancer xenografts in athymic mice. PSII treatment significantly inhibited the growth of xenograft tumors relative to controls by 70% (p < 0.05). These findings demonstrated that, in addition to the unique selectivity against cancer cells, PSII is a potent antitumor molecule that may be developed as a cancer therapeutic agent.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Ováricas/patología , Rizoma/química , Saponinas/farmacología , Animales , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Diosgenina/análogos & derivados , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Citometría de Flujo , Humanos , Ratones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/ultraestructura , Fosforilación/efectos de los fármacos , Saponinas/química , Saponinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Int J Gynecol Cancer ; 21(7): 1172-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21897275

RESUMEN

INTRODUCTION: Antrodia camphorata is a Chinese herb. Recently, several reports demonstrated that it had growth-inhibiting effects on some cancer cells. In this study, we investigated whether the crude extract of A. camphorata could inhibit the growth of ovarian cancer cells and examined the possible mechanisms involved. We also examined whether the cytotoxic effect of paclitaxel on ovarian cancer cells would be affected by A. camphorata. MATERIALS AND METHODS: Two human ovarian cancer cell lines, SKOV-3 and TOV-21G, were treated with A. camphorata (3-300 µg/mL). An MTT assay was used to test its cytotoxic effect. The apoptosis-related factors including the activity of caspase-3, -8, and -9 and the cytochrome c level released from mitochondria were analyzed. The expression of Bcl-2 family proteins (Bcl-2, Bcl-xL, Bax, Bim, Bad, and Bak) was examined by Western blot analysis. Cell lines were further treated with paclitaxel or paclitaxel plus A. camphorata to examine the cytotoxic efficiency. RESULTS: The MTT assay revealed that A. camphorata was cytotoxic to both the ovarian cancer cells in a dose- and time-dependent manner. Activities of caspase-3, -8, and -9 and release of mitochondrial cytochrome c increased in both ovarian cancer cell lines with increased dose of A. camphorata. Western blot analysis of Bcl-2 family proteins revealed an increased expression of Bad in SKOV-3 cells, whereas increased expression of Bim and Bak and decreased expression of Bcl-xL were noted in TOV-21G cells. In addition, the cytotoxic effect of paclitaxel on SKOV-3 and TOV-21G cells was increased significantly with the addition of A. camphorata (P < 0.01) by MTT assay. CONCLUSIONS: These in vitro results suggest that A. camphorata causes a cytotoxic effect on ovarian cancer cells through the induction of apoptosis. It may also enhance the antitumor effect of paclitaxel. Further studies with the ultimate goal of conducting clinical trials are warranted.


Asunto(s)
Antrodia , Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Antineoplásicos Fitogénicos/análisis , Antineoplásicos Fitogénicos/uso terapéutico , Antrodia/química , Caspasas/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Quimioterapia Combinada , Medicamentos Herbarios Chinos/farmacología , Femenino , Humanos , Neoplasias Ováricas/enzimología , Paclitaxel/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
13.
J Med Food ; 14(1-2): 46-52, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21186985

RESUMEN

The current study was focused on the induction of apoptotic effects of resveratrol along with the combination treatments of nutlin-3 and transforming growth factor-ß (TGF-ß) against the human ovarian cancer cell line A2780/CP70. To determine the extent of apoptosis following the above-mentioned treatments, we assessed the execution of apoptotic events that proceed via caspase activation and cytochrome c release. We estimated the caspase-3 and -9 activities using a direct enzymatic assay that measures the cleavage of synthetic peptide substrate (N-acetyl-Asp-Glu-Val-Asp-p-nitroanilide). Our experiments showed an increase in caspase-3 and -9 activities in the cells that were treated with the combination of resveratrol (5 µM) with nutlin-3 (5 µM) or TGF-ß (1 µg/mL). Since activation of procaspase-3 by caspase-9 requires the release of cytochrome c into the cytoplasm, we measured the levels of cytochrome c in the cytoplasm by western blot experiments. The data indicated a considerable increase in caspase-3 and cytochrome c levels when cells were treated with drugs for 24 hours. Experiments with 4,6'-diamino-2-phenylindole dihydrochloride (DAPI) staining also confirmed the induction of apoptosis in all the above-mentioned treatments done at 24 and 48 hours. These results support our hypothesis that resveratrol combination can induce programmed cell death at doses that are less than half of what is typically needed for nutlin-3 and TGF-ß to induce apoptosis.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Imidazoles/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/enzimología , Piperazinas/farmacología , Estilbenos/farmacología , Línea Celular Tumoral , Quimioterapia Combinada , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Neoplasias Ováricas/fisiopatología , Resveratrol
14.
J Steroid Biochem Mol Biol ; 118(4-5): 211-8, 2010 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-19800002

RESUMEN

Estradiol (E2), estrogen receptor (ER), ER-coregulators have been implicated in the development and progression of breast cancer. In situ E2 synthesis is implicated in tumor cell proliferation through autocrine or paracrine mechanisms, especially in post-menopausal women. Several recent studies demonstrated activity of aromatase P450 (Cyp19), a key enzyme that plays critical role in E2 synthesis in breast tumors. The mechanism by which tumors enhance aromatase expression is not completely understood. Recent studies from our laboratory suggested that PELP1 (Proline, Glutamic acid, Leucine rich Protein 1), a novel ER-coregulator, functions as a potential proto-oncogene and promotes tumor growth in nude mice models without exogenous E2 supplementation. In this study, we found that PELP1 deregulation contributes to increased expression of aromatase, local E2 synthesis and PELP1 cooperates with growth factor signaling components in the activation of aromatase. PELP1 deregulation uniquely up-regulated aromatase expression via activation of aromatase promoter I.3/II. Analysis of PELP1 driven mammary tumors in xenograft as well as in transgenic mouse models revealed increased aromatase expression. PELP1-mediated induction of aromatase requires functional Src and PI3K pathways. Chromatin immuno precipitation (ChIP) assays revealed that PELP1 is recruited to the Aro 1.3/II aromatase promoter. HER2 signaling enhances PELP1 recruitment to the aromatase promoter and PELP1 plays a critical role in HER2-mediated induction of aromatase expression. Mechanistic studies revealed that PELP1 interactions with orphan receptor ERRalpha, and histone demethylases play a role in the activation of aromatase promoter. Accordingly, ChIP analysis showed alterations in histone modifications at the aromatase promoter in the model cells that exhibit local E2 synthesis. Immunohistochemical analysis of breast tumor progression tissue arrays suggested that deregulation of aromatase expression occurs in advanced-stage and node-positive tumors, and that cooverexpression of PELP1 and aromatase occur in a sub set of tumors. Collectively, our results suggest that PELP1 regulation of aromatase represent a novel mechanism for in situ estrogen synthesis leading to tumor proliferation by autocrine loop and open a new avenue for ablating local aromatase activity in breast tumors.


Asunto(s)
Aromatasa/genética , Neoplasias de la Mama/enzimología , Estrógenos/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Transactivadores/metabolismo , Animales , Aromatasa/metabolismo , Neoplasias de la Mama/genética , Proteínas Co-Represoras , Endometriosis/enzimología , Endometriosis/genética , Epigénesis Genética , Femenino , Humanos , Ratones , Ratones Transgénicos , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/genética , Proto-Oncogenes Mas , Receptores de Estrógenos/metabolismo , Transactivadores/genética , Factores de Transcripción , Receptor Relacionado con Estrógeno ERRalfa
15.
Br J Cancer ; 100(12): 1903-11, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19455141

RESUMEN

Prodrug activation gene therapy is a developing approach to cancer treatment, whereby prodrug-activating enzymes are expressed in tumour cells. After administration of a non-toxic prodrug, its conversion to cytotoxic metabolites directly kills tumour cells expressing the activating enzyme, whereas the local spread of activated metabolites can kill nearby cells lacking the enzyme (bystander cell killing). One promising combination that has entered clinical trials uses the nitroreductase NfsB from Escherichia coli to activate the prodrug, CB1954, to a potent bifunctional alkylating agent. NfsA, the major E. coli nitroreductase, has greater activity with nitrofuran antibiotics, but it has not been compared in the past with NfsB for the activation of CB1954. We show superior in vitro kinetics of CB1954 activation by NfsA using the NADPH cofactor, and show that the expression of NfsA in bacterial or human cells results in a 3.5- to 8-fold greater sensitivity to CB1954, relative to NfsB. Although NfsB reduces either the 2-NO(2) or 4-NO(2) positions of CB1954 in an equimolar ratio, we show that NfsA preferentially reduces the 2-NO(2) group, which leads to a greater bystander effect with cells expressing NfsA than with NfsB. NfsA is also more effective than NfsB for cell sensitisation to nitrofurans and to a selection of alternative, dinitrobenzamide mustard (DNBM) prodrugs.


Asunto(s)
Antineoplásicos/uso terapéutico , Aziridinas/uso terapéutico , Proteínas de Escherichia coli/genética , Terapia Genética , Nitrorreductasas/genética , Neoplasias Ováricas/terapia , Profármacos/uso terapéutico , Adenoviridae/genética , Supervivencia Celular , Cromatografía Líquida de Alta Presión , Terapia Combinada , Escherichia coli/genética , Femenino , Vectores Genéticos , Humanos , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Células Tumorales Cultivadas
16.
Cancer Lett ; 271(2): 251-9, 2008 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-18639975

RESUMEN

Metal-binding compounds have been shown to have anticancer activity and are being evaluated clinically as anticancer agents. We have recently found that a zinc-binding compound, 5-chloro-7-iodo-8-hydroxyquinoline (clioquinol), kills cancer cells by transporting zinc into the cells. We therefore compared the action of clioquinol with two other cytotoxic zinc-binding compounds, N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN) and pyrrolidine dithiocarbamate (PDTC). We demonstrate that metal-binding compounds can be subclassified based upon the reversibility of their cytotoxicity by metal supplementation and their modes of action. Understanding the mechanisms whereby metal-binding compounds affect cell behavior may aid in their optimization for clinical use.


Asunto(s)
Apoptosis/efectos de los fármacos , Clioquinol/farmacología , Zinc/metabolismo , Western Blotting , Línea Celular Tumoral , Clioquinol/metabolismo , Etilenodiaminas/farmacología , Femenino , Humanos , FN-kappa B/metabolismo , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Triazenos/farmacología
17.
Cancer Lett ; 266(2): 249-62, 2008 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-18400375

RESUMEN

Over-expression of EGFR, as in most cases of ovarian cancer, is associated with advanced-stage disease and poor prognosis. Activation of EGFR signaling pathway is involved in increased cell proliferation, angiogenesis, metastasis and decreased apoptosis. Tyrosine kinase activity is essential for signal transduction and receptor down-regulation. However, we found in this study that tyrosine kinase activity is not necessary in ligand-induced EGFR down-regulation in ovarian cancer cell line CaOV3 cells. EGFR tyrosine kinase inhibitors, such as PD153035, AG1478, as well as non-specific tyrosine kinase inhibitor PP2 cannot reverse EGF-induced down-regulation of EGFR. These findings thus permit us to develop the following exciting but unconventional strategy to sensitize cancer cells, namely, by priming ovarian cancer cells with EGF and EGFR inhibitor PD153035, before chemotherapy. This priming procedure down-regulates EGFR without induction of mitogenic signals such as ERK and PI3K/AKT. EGF plus EGFR inhibitor-primed ovarian cancer cells display increased sensitivity to taxol-induced cell death, resistant to EGF-induced cell migration and cell proliferation as well as ERK and PI3K/AKT activation. Further studies showed that PD153035, which does not reverse ligand-induced EGFR down-regulation, blocks EGF-induced EGFR activation as well as EGFR's binding to c-cbl and Grb2. Taken together, we contend that priming with EGFR inhibitors plus EGF inhibits cell signaling pathways leading to cell proliferation and survival, while down-regulating EGFR. This priming approach sensitizes ovarian cancer cells and would ultimately result in better chemotherapeutical outcome.


Asunto(s)
Antineoplásicos/farmacología , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Neoplasias Ováricas/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Receptores ErbB/metabolismo , Femenino , Ratones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Paclitaxel/toxicidad , Transducción de Señal
18.
Clin Cancer Res ; 13(18 Pt 1): 5314-21, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875760

RESUMEN

PURPOSE: The phosphatidylinositol 3'-kinase (PI3K) family plays a key regulatory role in various cancer-associated signal transduction pathways. Here, we investigated the genomic alterations and gene expression of most known PI3K family members in human epithelial ovarian cancer. EXPERIMENTAL DESIGN: The DNA copy number of PI3K family genes was screened by a high-resolution array comparative genomic hybridization in 89 human ovarian cancer specimens. The mRNA expression level of PI3K genes was analyzed by microarray retrieval approach, and further validated by real-time reverse transcription-PCR. The expression of p55gamma protein in ovarian cancer was analyzed on tissue arrays. Small interfering RNA was used to study the function of PIK3R3 in ovarian cancer. RESULTS: In ovarian cancer, 6 of 12 PI3K genes exhibited significant DNA copy number gains (>20%), including PIK3CA (23.6%), PIK3CB (27.0%), PIK3CG (25.8%), PIK3R2 (29.2%), PIK3R3 (21.3%), and PIK3C2B (40.4%). Among those, only PIK3R3 had significantly up-regulated mRNA expression level in ovarian cancer compared with normal ovary. Up-regulated PIK3R3 mRNA expression was also observed in liver, prostate, and breast cancers. The PIK3R3 mRNA expression level was significantly higher in ovarian cancer cell lines (n = 18) than in human ovarian surface epithelial cells (n = 6, P = 0.002). Overexpression of p55gamma protein in ovarian cancer was confirmed by tissue array analysis. In addition, we found that knockdown of PIK3R3 expression by small interfering RNA significantly increased the apoptosis in cultured ovarian cancer cell lines. CONCLUSION: We propose that PIK3R3 may serve as a potential therapeutic target in human ovarian cancer.


Asunto(s)
Carcinoma/enzimología , Neoplasias Ováricas/enzimología , Fosfatidilinositol 3-Quinasas/genética , Carcinoma/tratamiento farmacológico , Carcinoma/genética , Femenino , Dosificación de Gen , Genómica , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Fosfatidilinositol 3-Quinasas/análisis , Inhibidores de las Quinasa Fosfoinosítidos-3 , ARN Mensajero/análisis , ARN Mensajero/metabolismo
19.
J Clin Endocrinol Metab ; 92(12): 4802-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17895316

RESUMEN

INTRODUCTION: In polycystic ovary syndrome (PCOS), there is increased formation of androgens by thecal cells. Moreover, PCOS ovaries have been shown to have decreased levels of c-fos transcription factor. We hypothesize that c-fos expression inhibits 17alpha-hydroxylase 17,20 lyase (CYP17) activity in the human ovary, and its decreased expression seen in PCOS may lead to elevated CYP17 transcription, resulting in increased androgen production. OBJECTIVE: Our objective was to define the role of the activator protein-1 transcription factors, namely c-fos, in the regulation of CYP17 expression in theca cells. METHODS: Human ovarian thecal-like tumor cells were used for all experiments. The following techniques were used: steroid quantification, mRNA extraction, microarray analysis, transfection, small interfering RNA, and immunohistochemistry. RESULTS: Stimulation of human ovarian thecal-like tumor cells with the protein kinase A pathway activator forskolin resulted in stimulation of C19 androgen production. In contrast, treatment with the protein kinase C pathway activator tetradecanoylphorbol acetate (TPA) resulted in decreased androgen production with a shift toward C21 progesterone production. TPA also led to complete inhibition of CYP17. Microarray data showed a 37-fold increase in c-fos after treatment with TPA. Transfection with steroidogenic factor 1 resulted in an increase in CYP17 promoter activity, which was significantly inhibited in the presence of c-fos. c-fos gene silencing led to an increase in CYP17 mRNA levels. Immunohistochemical staining for c-fos in ovaries demonstrated strong staining in granulosa cells, but not theca. CONCLUSIONS: The activator protein-1 transcription factor c-fos plays a role in the inhibition of CYP17 expression. The decreased levels of c-fos expression in polycystic ovaries may be responsible for increased CYP17 levels in PCOS.


Asunto(s)
Andrógenos/biosíntesis , Neoplasias Ováricas/metabolismo , Proteína Quinasa C/fisiología , Esteroide 17-alfa-Hidroxilasa/biosíntesis , Neoplasia Tecoma/metabolismo , Western Blotting , Línea Celular Tumoral , ADN Complementario/biosíntesis , ADN Complementario/genética , Femenino , Silenciador del Gen , Genes fos/genética , Genes jun/genética , Vectores Genéticos , Humanos , Inmunohistoquímica , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Ováricas/enzimología , Síndrome del Ovario Poliquístico/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño , Neoplasia Tecoma/enzimología , Transfección
20.
Cancer Detect Prev ; 31(2): 119-28, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17335992

RESUMEN

BACKGROUND: Dietary factors influence tumor formation and progression. Vitamin E is a dietary anti-oxidant capable of eliminating free radical damage, inducing apoptosis and decreasing oncogene expression. Therefore, Vitamin E may be a strong candidate for cancer prevention and/or chemotherapeutic intervention. Since telomerase, a ribonucleoprotein uniquely expressed in over 95% of cancers, plays an important role in cellular immortalization, cell growth and tumor progression, the present study investigated the effects of Vitamin E on telomerase activity in human ovarian cancer. METHODS: Normal and malignant ovarian surface epithelial (OSE) cells were cultured with and without D-alpha tocopheryl acetate (Vitamin E). MTS and Western immunoblot assays were used to examine the effect of Vitamin E on cell growth, survival and cytotoxicity. PCR-ELISA, RT-PCR and luciferase reporter assays were performed to determine the effect of Vitamin E on telomerase activity. RESULTS: Vitamin E suppressed endogenous telomerase activity in ovarian cancer cells, but had no similar effects in telomerase-negative normal OSE cells. Vitamin E also reduced hTERT-mRNA transcript levels and reduced hTERT promoter activity maximally targeting the -976 to -578bp promoter regions. In addition, Vitamin E improved cisplatin-mediated cytotoxicity as evidenced by reduced cancer cell growth and increased cleaved caspase 3 activity. In contrast, Vitamin E protected telomerase-negative OSE cells from cisplatin-mediated cytotoxicity as evidenced by decreased cleaved caspase 3 activity. CONCLUSION: Our data suggest that, by suppressing telomerase activity, Vitamin E may be an important protective agent against ovarian cancer cell growth as well as a potentially effective therapeutic adjuvant.


Asunto(s)
Antioxidantes/farmacología , Neoplasias Ováricas/enzimología , Telomerasa/antagonistas & inhibidores , Vitamina E/farmacología , Antineoplásicos/farmacología , Western Blotting , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telomerasa/genética , Células Tumorales Cultivadas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA