Cargando…

Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway

INTRODUCTION: Oridonin, which is isolated from the Chinese herb Rabdosia rubescens, has been reported to exhibit an anti-tumorous effect on different cancers. In this study, we investigated the molecular mechanism by which oridonin suppresses human ovarian cancer. MATERIAL AND METHODS: The inhibitio...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Ye, Zhu, Zhiling
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Termedia Publishing House 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6657258/
https://www.ncbi.nlm.nih.gov/pubmed/31360196
http://dx.doi.org/10.5114/aoms.2018.77068
_version_ 1783438780395421696
author Wang, Ye
Zhu, Zhiling
author_facet Wang, Ye
Zhu, Zhiling
author_sort Wang, Ye
collection PubMed
description INTRODUCTION: Oridonin, which is isolated from the Chinese herb Rabdosia rubescens, has been reported to exhibit an anti-tumorous effect on different cancers. In this study, we investigated the molecular mechanism by which oridonin suppresses human ovarian cancer. MATERIAL AND METHODS: The inhibition of oridonin on cell proliferation was assessed by CCK8 assay. Cell cycle and apoptosis were analyzed by flow cytometry, staining with propidium iodide (PI) or annexin-V/PI respectively. The metastasis rate was evaluated using a transwell migration assay. The expression of metastasis-associated genes and mTOR pathway related genes were detected by western blot. RESULTS: We demonstrated that oridonin suppressed the proliferation and blocked the cell cycle in G1/S phage and induced apoptosis in SKOV3 and A2780 cells (p < 0.01). We further found that the mTOR signaling pathway was suppressed by the treatment with oridonin, and the activation of the mTOR pathway attenuated the anti-tumorous effect of oridonin in human ovarian cancer cells, suggesting that the mTOR pathway was involved in the anti-tumorous process of oridonin. Additionally, the activation of the mTOR pathway by an exogenous activator reduced the expression level of FOXP3 (p < 0.01), thus providing evidence that FOXP3 is a factor that is necessary for the anti-tumorous effect of oridonin, and is negatively regulated by the mTOR pathway. CONCLUSIONS: These results suggested that oridonin suppressed the mTOR signaling pathway, up-regulated the FOXP3 level, and inhibited metastasis of human ovarian cancer cells.
format Online
Article
Text
id pubmed-6657258
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Termedia Publishing House
record_format MEDLINE/PubMed
spelling pubmed-66572582019-07-29 Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway Wang, Ye Zhu, Zhiling Arch Med Sci Basic Research INTRODUCTION: Oridonin, which is isolated from the Chinese herb Rabdosia rubescens, has been reported to exhibit an anti-tumorous effect on different cancers. In this study, we investigated the molecular mechanism by which oridonin suppresses human ovarian cancer. MATERIAL AND METHODS: The inhibition of oridonin on cell proliferation was assessed by CCK8 assay. Cell cycle and apoptosis were analyzed by flow cytometry, staining with propidium iodide (PI) or annexin-V/PI respectively. The metastasis rate was evaluated using a transwell migration assay. The expression of metastasis-associated genes and mTOR pathway related genes were detected by western blot. RESULTS: We demonstrated that oridonin suppressed the proliferation and blocked the cell cycle in G1/S phage and induced apoptosis in SKOV3 and A2780 cells (p < 0.01). We further found that the mTOR signaling pathway was suppressed by the treatment with oridonin, and the activation of the mTOR pathway attenuated the anti-tumorous effect of oridonin in human ovarian cancer cells, suggesting that the mTOR pathway was involved in the anti-tumorous process of oridonin. Additionally, the activation of the mTOR pathway by an exogenous activator reduced the expression level of FOXP3 (p < 0.01), thus providing evidence that FOXP3 is a factor that is necessary for the anti-tumorous effect of oridonin, and is negatively regulated by the mTOR pathway. CONCLUSIONS: These results suggested that oridonin suppressed the mTOR signaling pathway, up-regulated the FOXP3 level, and inhibited metastasis of human ovarian cancer cells. Termedia Publishing House 2018-07-10 2019-07 /pmc/articles/PMC6657258/ /pubmed/31360196 http://dx.doi.org/10.5114/aoms.2018.77068 Text en Copyright: © 2018 Termedia & Banach http://creativecommons.org/licenses/by-nc-sa/4.0/ This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License, allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
spellingShingle Basic Research
Wang, Ye
Zhu, Zhiling
Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway
title Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway
title_full Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway
title_fullStr Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway
title_full_unstemmed Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway
title_short Oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mTOR pathway
title_sort oridonin inhibits metastasis of human ovarian cancer cells by suppressing the mtor pathway
topic Basic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6657258/
https://www.ncbi.nlm.nih.gov/pubmed/31360196
http://dx.doi.org/10.5114/aoms.2018.77068
work_keys_str_mv AT wangye oridonininhibitsmetastasisofhumanovariancancercellsbysuppressingthemtorpathway
AT zhuzhiling oridonininhibitsmetastasisofhumanovariancancercellsbysuppressingthemtorpathway