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










Base de datos
Intervalo de año de publicación
1.
Int J Nanomedicine ; 14: 5713-5728, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31413571

RESUMEN

Purpose: The levels of reactive oxygen species (ROS) in tumor cells are much higher than that in normal cells, and rise rapidly under the influence of exogenous or endogenous inducing factors, eventually leading to the apoptosis of tumor cells. Therefore, this study prepared a dual pH/reducing-responsive poly (N-isopropylacrylamide-co-Cinnamaldehyde-co-D-α-tocopheryl polyethylene glycol 1000 succinate, PssNCT) nanogels, which employed two exogenous ROS inducers, cinnamaldehyde (CA) and D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS), to selectively induce apoptosis by regulating ROS levels in tumor cells. Methods: The PssNCT nanogels were prepared by the free radical precipitation polymerization under the crosslink between pH-sensitive hydrazone and reducing-sensitive disulfide bonds, followed by the physicochemical and morphological characteristics investigations. Plasma stability, dual pH/reducing responsive degradation and in vitro release were also assessed. In cell experiments, cytotoxicity in different cells were first detected. The intracellular ROS levels and mitochondrial functions of tumor cells were then evaluated. Moreover, the apoptosis and western-blot assays were employed to verify the association between ROS levels elevation and apoptosis in tumor cells. Results: The nanogels exhibited a round-like hollow structure with the diameter smaller than 200nm. The nanogels were stable in plasma, while showed rapid degradation in acidic and reducing environments, thus achieving significant release of CA and TPGS in these media. Furthermore, the sufficient amplification of ROS signals was induced by the synergistically function of CA and TPGS on mitochondria, which resulted in the opening of the mitochondrial apoptotic pathway and enhanced cytotoxicity on MCF-7 cells. However, nanogels barely affected L929 cells owing to their lower intracellular ROS basal levels. Conclusion: The specific ROS regulation method achieved by these nanogels could be explored to selectively kill tumor cells according to the difference of ROS signals in different kinds of cells.


Asunto(s)
Apoptosis , Espacio Intracelular/química , Polietilenglicoles/farmacología , Polietileneimina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Acroleína/análogos & derivados , Acroleína/farmacología , Animales , Apoptosis/efectos de los fármacos , Doxorrubicina/farmacología , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nanogeles , Vitamina E/síntesis química , Vitamina E/química
2.
Mol Pharm ; 16(2): 689-700, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30601012

RESUMEN

The excessive increase of intracellular reactive oxygen species (ROS) makes tumor cells usually in the state of oxidative stress. Although tumor cells can adapt to this state to a certain extent by upregulating antioxidant systems, the further ROS insults disrupt the transient intracellular redox balance, eventually leading to apoptosis and necrosis. Therefore, increasing the intracellular ROS level can effectively amplify the oxidative stress and induce apoptosis, which can be employed as a strategy for tumor treatment. Herein, a unique pH-responsive ROS inducing micellar system was reported in this study to specifically amplify the ROS signal in tumor cells. This micellar system was constructed by a new amphiphilic polymer, PIAThydCA, composed of poly(itaconic acid) (PIA) as the hydrophilic backbone, d-α-tocopherol polyethylene glycol 1000 succinate (TPGS) as the hydrophobic side chain, and cinnamaldehyde (CA) as the ROS-generating agent, which were linked to PIA by the pH-sensitive hydrazone bond. PIAThydCA micelles could be degraded in the intracellular acidic environment through the hydrolysis of hydrazone bond and release CA. CA and TPGS could amplify oxidative stress cooperatively to kill MCF-7 human breast cells preferentially through the mitochondrial apoptosis pathway. Therefore, we anticipate that the PIAThydCA micelles could exert great potential in anticancer therapy.


Asunto(s)
Estrés Oxidativo/efectos de los fármacos , Polímeros/química , Acroleína/análogos & derivados , Acroleína/química , Antineoplásicos/química , Antineoplásicos/farmacología , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Micelas , Oxidación-Reducción/efectos de los fármacos , Polietilenglicoles/química , Polímeros/farmacología , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...