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












Intervalo de año de publicación
1.
Braz. j. med. biol. res ; 57: e13152, fev.2024. graf
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1534071

RESUMEN

Abstract The cure rates for osteosarcoma have remained unchanged in the past three decades, especially for patients with pulmonary metastasis. Thus, a new and effective treatment for metastatic osteosarcoma is urgently needed. Anlotinib has been reported to have antitumor effects on advanced osteosarcoma. However, both the effect of anlotinib on autophagy in osteosarcoma and the mechanism of anlotinib-mediated autophagy in pulmonary metastasis are unclear. The effect of anlotinib treatment on the metastasis of osteosarcoma was investigated by transwell assays, wound healing assays, and animal experiments. Related proteins were detected by western blotting after anlotinib treatment, ATG5 silencing, or ATG5 overexpression. Immunofluorescence staining and transmission electron microscopy were used to detect alterations in autophagy and the cytoskeleton. Anlotinib inhibited the migration and invasion of osteosarcoma cells but promoted autophagy and increased ATG5 expression. Furthermore, the decreases in invasion and migration induced by anlotinib treatment were enhanced by ATG5 silencing. In addition, Y-27632 inhibited cytoskeletal rearrangement, which was rescued by ATG5 overexpression. ATG5 overexpression enhanced epithelial-mesenchymal transition (EMT). Mechanistically, anlotinib-induced autophagy promoted migration and invasion by activating EMT and cytoskeletal rearrangement through ATG5 both in vitro and in vivo. Our results demonstrated that anlotinib can induce protective autophagy in osteosarcoma cells and that inhibition of anlotinib-induced autophagy enhanced the inhibitory effects of anlotinib on osteosarcoma metastasis. Thus, the therapeutic effect of anlotinib treatment can be improved by combination treatment with autophagy inhibitors, which provides a new direction for the treatment of metastatic osteosarcoma.

2.
Braz J Med Biol Res ; 57: e13152, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38381883

RESUMEN

The cure rates for osteosarcoma have remained unchanged in the past three decades, especially for patients with pulmonary metastasis. Thus, a new and effective treatment for metastatic osteosarcoma is urgently needed. Anlotinib has been reported to have antitumor effects on advanced osteosarcoma. However, both the effect of anlotinib on autophagy in osteosarcoma and the mechanism of anlotinib-mediated autophagy in pulmonary metastasis are unclear. The effect of anlotinib treatment on the metastasis of osteosarcoma was investigated by transwell assays, wound healing assays, and animal experiments. Related proteins were detected by western blotting after anlotinib treatment, ATG5 silencing, or ATG5 overexpression. Immunofluorescence staining and transmission electron microscopy were used to detect alterations in autophagy and the cytoskeleton. Anlotinib inhibited the migration and invasion of osteosarcoma cells but promoted autophagy and increased ATG5 expression. Furthermore, the decreases in invasion and migration induced by anlotinib treatment were enhanced by ATG5 silencing. In addition, Y-27632 inhibited cytoskeletal rearrangement, which was rescued by ATG5 overexpression. ATG5 overexpression enhanced epithelial-mesenchymal transition (EMT). Mechanistically, anlotinib-induced autophagy promoted migration and invasion by activating EMT and cytoskeletal rearrangement through ATG5 both in vitro and in vivo. Our results demonstrated that anlotinib can induce protective autophagy in osteosarcoma cells and that inhibition of anlotinib-induced autophagy enhanced the inhibitory effects of anlotinib on osteosarcoma metastasis. Thus, the therapeutic effect of anlotinib treatment can be improved by combination treatment with autophagy inhibitors, which provides a new direction for the treatment of metastatic osteosarcoma.


Asunto(s)
Neoplasias Óseas , Indoles , Neoplasias Pulmonares , Osteosarcoma , Quinolinas , Animales , Humanos , Proliferación Celular , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Autofagia , Transición Epitelial-Mesenquimal , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Citoesqueleto/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proteína 5 Relacionada con la Autofagia/farmacología , Proteína 5 Relacionada con la Autofagia/uso terapéutico
3.
Front Oncol ; 10: 543562, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33014879

RESUMEN

The survival rate of osteosarcoma, the most prevalent primary bone tumor, has not been effectively improved in the last 30 years. Hence, new treatments and drugs are urgently needed. Antiangiogenic therapy and immunotherapy have good antitumor effects in many kinds of tumors. It is hypothesized that there may be a synergistic effect between immune checkpoint inhibitors and antiangiogenic therapy. Nevertheless, its potential mechanism is still unclear. Vascular endothelial growth factor receptor-2 (VEGFR2) expression was detected by immunohistochemistry in 18 paired osteosarcoma tissues. Moreover, we investigated the effects of apatinib treatment and VEGFR2 knockdown on osteosarcoma as well as the relevant underlying mechanism. Immunohistochemistry assays showed that, compared with that in primary osteosarcoma, VEGFR2 expression was higher in lung metastases. VEGFR2 was positively correlated with PD-L2 expression in osteosarcoma lung metastasis. Transwell assays indicated that VEGFR2 inhibition reduced osteosarcoma cell metastatic abilities in vitro. We also demonstrated that VEGFR2 inhibition downregulated the STAT3 and RhoA-ROCK-LIMK2 pathways, thereby attenuating migration and invasion. Additionally, VEGFR2 inhibition targeted STAT3, through which it reduced PD-L2 expression in osteosarcoma cells. VEGFR2 inhibition markedly attenuated osteosarcoma lung metastatic ability in vivo. In this study, we presented the pro-metastatic functional mechanism of VEGFR2 in osteosarcoma. VEGFR2 inhibition exhibits antitumor effects through antiangiogenic effects and inhibition of immune escape, which possibly provides potential clinical treatment for metastatic osteosarcoma.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...