Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Pharm ; 17(12): 4483-4498, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33205974

RESUMEN

Mitochondrial malfunction plays a crucial role in cancer development and progression. Cancer cells show a substantially higher mitochondrial activity and greater mitochondrial transmembrane potential than normal cells. This concept can be exploited for targeting cytotoxic drugs to the mitochondria of cancer cells using mitochondrial-targeting compounds. In this study, a polyamidoamine dendrimer-based mitochondrial delivery system was prepared for curcumin using triphenylphosphonium ligands to improve the anticancer efficacy of the drug in vitro and in vivo. For the in vitro evaluations, various methods, such as viability assay, confocal microscopy, flow cytometry, reactive oxygen species (ROS), and real-time polymerase chain reaction analyses, were applied. Our findings showed that the targeted-dendrimeric curcumin (TDC) could successfully deliver and colocalize the drug to the mitochondria of the cancer cells, and selectively induce a potent apoptosis and cell cycle arrest at G2/M. Moreover, at a low curcumin dose of less than 25 µM, TDC significantly reduced adenosine triphosphate and glutathione, and increased the ROS level of the isolated rat hepatocyte mitochondria. The in vivo studies on the Hepa1-6 tumor-bearing mice also indicated a significant tumor suppression effect and the highest median survival days (Kaplan-Meier survival estimation and log-rank test) after treatment with the TDC construct compared to the free curcumin and untargeted construct. Besides its targeted nature and safety, the expected improved solubility and stability represent the prepared targeted-dendrimeric construct as an up-and-coming candidate for cancer treatment. The results of this study emphasize the promising route of mitochondrial targeting as a practical approach for cancer therapy, which can be achieved by optimizing the delivery method.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Curcumina/administración & dosificación , Portadores de Fármacos/química , Neoplasias Hepáticas/tratamiento farmacológico , Poliaminas/química , Animales , Carcinoma Hepatocelular/patología , Fraccionamiento Celular , Línea Celular Tumoral , Curcumina/química , Dendrímeros/química , Estabilidad de Medicamentos , Hepatocitos , Humanos , Neoplasias Hepáticas/patología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Cultivo Primario de Células , Ratas , Solubilidad , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Sci Rep ; 13(1): 8844, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37258563

RESUMEN

Breast cancer is one of the leading causes of cancer deaths in women worldwide. Magnetic fields have shown anti-tumor effects in vitro and in vivo as a non-invasive therapy method that can affect cellular metabolism remotely. Doxorubicin (DOX) is one of the most commonly used drugs for treating breast cancer patients. It can be assumed that combining chemotherapy and magnetotherapy is one of the most effective treatments for breast cancer. This study aimed to investigate the potential cytotoxic effect of DOX at low concentrations in combination with extremely low-frequency electromagnetic fields (ELF-EMF; 50 Hz; 20 mT). The breast cancer cell line MCF-7 was examined for oxidative stress, cell cycle, and apoptosis. MCF-7 cells were treated with various concentrations of DOX as an apoptosis-inducing agent and ELF-EMF. Cytotoxicity was examined using the MTT colorimetric assay at 12, 24, and 48 h. Consequently, concentration- and time-dependent cytotoxicity was observed in MCF-7 cells for DOX within 24 h. The MTT assay results used showed that a 2 µM concentration of DOX reduced cell viability to 50% compared with control, and as well, the combination of ELF-EMF and DOX reduced cell viability to 50% compared with control at > 0.25 µM doses for 24 h. In MCF-7 cells, combining 0.25 µM DOX with ELF-EMF resulted in increased ROS levels and DOX-induced apoptosis. Flow cytometry analysis, on the other hand, revealed enhanced arrest of MCF-7 cells in the G0-G1 phase of the cell cycle, as well as inducing apoptotic cell death in MCF-7 cells, implying that the synergistic effects of 0.25 µM DOX and ELF-EMF may represent a novel and effective agent against breast cancer.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Humanos , Femenino , Células MCF-7 , Campos Electromagnéticos , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Antineoplásicos/farmacología , Neoplasias de la Mama/patología , Apoptosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA