Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biomater Sci ; 7(6): 2559-2570, 2019 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-30977484

RESUMEN

The photothermal response of nanomaterials provides a basis for many biomedical applications, including diagnosis (e.g., biosensor and photoacoustic imaging) and treatment (e.g., drug delivery and photothermal therapy). The use of nano-materials for cancer phototherapy (solid tumor ablation) can cause cell necrosis and apoptosis. However, photothermal effects using the same material can differ among tumor cell types, and the molecular mechanisms underlying these differences are not clear. We used polydopamine (PDA)-coated branched Au-Ag nanoparticles (Au-Ag@PDA NPs) for the photothermal treatment of two prostate cancer cell lines. The therapeutic effect was evaluated by CCK8, flow cytometry, and expression analyses of related genes by western blotting. Photothermal therapy resulted in oxidative stress in prostate cancer cells and activated the mitochondrial-related apoptosis pathway, increasing the Bax expression. In addition, we observed a greater photothermal treatment effect on the androgen-dependent cells LNCaP than the androgen-independent cells DU145. Pretreatment with an inhibitor of the NF-κB signaling pathway (BAY 11-7082) enhanced the expression of BAX in the DU145 cells and increased the sensitivity of the cells to the heat treatment of Au-Ag@PDA NPs both in vitro and in vivo. Our findings explain the differences in the observed effects of photothermal therapy and provide the direction for further improvements to this strategy.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Fototerapia , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Oro/química , Calor , Humanos , Indoles/química , Indoles/uso terapéutico , Masculino , Nanopartículas del Metal/química , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , FN-kappa B/antagonistas & inhibidores , Nitrilos/farmacología , Estrés Oxidativo/efectos de los fármacos , Polímeros/química , Polímeros/uso terapéutico , Plata/química , Sulfonas/farmacología , Proteína X Asociada a bcl-2/metabolismo
2.
Acta Biomater ; 83: 414-424, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30366131

RESUMEN

Nanoparticles are emerging as a new therapeutic modality due to their high stability, precise targeting, and high biocompatibility. Branched Au-Ag nanoparticles with polydopamine coating (Au-Ag@PDA) have strong near-infrared absorbance and no cytotoxicity but high photothermal conversion efficiency. However, the photothermal activity of Au-Ag@PDA in vivo and in vitro has not been reported yet, and the mechanism underlying the effects of photothermal nanomaterials is not clear. Therefore, in this study, the colorectal cancer cell line HCT-116 and nude mice xenografts were used to observe the photothermal effects of Au-Ag@PDA in vivo and in vitro. The results suggest that Au-Ag@PDA NPs significantly inhibited cell proliferation and induced apoptosis in colorectal cancer cells. Moreover, Au-Ag@PDA NP-mediated photothermal therapy inhibited the growth of tumors at doses of 50 and 100 µg in vivo. The mechanisms through which Au-Ag@PDA NPs induced colorectal cancer cell death involved multiple pathways, including caspase-dependent and -independent apoptosis, mitochondrial damage, lysosomal membrane permeability, and autophagy. Thus, our findings suggest that Au-Ag@PDA NPs could be used as potential antitumor agents for photothermal ablation of colorectal cancer cells.


Asunto(s)
Apoptosis , Materiales Biocompatibles Revestidos , Neoplasias Colorrectales , Oro , Hipertermia Inducida , Indoles , Nanopartículas del Metal , Fototerapia , Polímeros , Plata , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacocinética , Materiales Biocompatibles Revestidos/farmacología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , Oro/química , Oro/farmacocinética , Oro/farmacología , Células HCT116 , Humanos , Indoles/química , Indoles/farmacocinética , Indoles/farmacología , Masculino , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Polímeros/química , Polímeros/farmacocinética , Polímeros/farmacología , Plata/química , Plata/farmacocinética , Plata/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Int J Nanomedicine ; 13: 6413-6428, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30410328

RESUMEN

PURPOSE: Polydopamine-coated branched Au-Ag nanoparticles (Au-Ag@PDA NPs) exhibit good structural stability, biocompatibility, and photothermal performance, along with potential anticancer efficacy. Here, we investigated the cytotoxicity of Au-Ag@PDA NPs against human bladder cancer cells (T24 cells) in vitro and in vivo, as well as the underlying molecular mechanisms of photothermal therapy-induced T24 cell death. MATERIALS AND METHODS: T24 cells were treated with different doses of Au-Ag@PDA NPs followed by 808 nm laser irradiation, and the effects on cell proliferation, cell cycle, apoptosis, and autophagy were analyzed. To confirm the mechanisms of inhibition, real-time PCR and Western blot analysis were used to evaluate markers of cell cycle, apoptosis, autophagy, and the AKT/ERK signaling pathway. Moreover, we evaluated the effects of the treatment on mitochondrial membrane potential and ROS generation to confirm the underlying mechanisms of inhibition. Finally, we tested the T24 tumor inhibitory effects of Au-Ag@PDA NPs plus laser irradiation in vivo using a xenograft mouse model. RESULTS: Au-Ag@PDA NPs, with appropriate laser irradiation, dramatically inhibited the proliferation of T24 cells, altered the cell cycle distribution by increasing the proportion of cells in the S phase, induced cell apoptosis by activating the mitochondria-mediated intrinsic pathway, and triggered a robust autophagy response in T24 cells. Moreover, Au-Ag@PDA NPs decreased the expression of phosphorylated AKT and ERK and promoted the production of ROS that function upstream of apoptosis and autophagy. In addition, Au-Ag@PDA NP-mediated photothermolysis also significantly suppressed tumor growth in vivo. CONCLUSION: This preclinical study can provide a mechanistic basis for Au-Ag@PDA NP-mediated photothermal therapy toward promotion of this method in the clinical treatment of bladder cancer.


Asunto(s)
Apoptosis , Autofagia , Puntos de Control del Ciclo Celular , Hipertermia Inducida , Indoles/química , Nanopartículas del Metal/química , Fototerapia , Polímeros/química , Neoplasias de la Vejiga Urinaria/patología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Oro/química , Humanos , Potencial de la Membrana Mitocondrial , Nanopartículas del Metal/ultraestructura , Ratones Desnudos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Plata/química , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA