Cargando…

Cyclovirobuxine D Induces Apoptosis and Mitochondrial Damage in Glioblastoma Cells Through ROS-Mediated Mitochondrial Translocation of Cofilin

BACKGROUND: Cyclovirobuxine D (CVBD), a steroidal alkaloid, has multiple pharmacological activities, including anti-cancer activity. However, the anti-cancer effect of CVBD on glioblastoma (GBM) has seldom been investigated. This study explores the activity of CVBD in inducing apoptosis of GBM cells...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Lin, Fu, Ruoqiu, Duan, Dongyu, Li, Ziwei, Li, Bin, Ming, Yue, Li, Li, Ni, Rui, Chen, Jianhong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8018288/
https://www.ncbi.nlm.nih.gov/pubmed/33816313
http://dx.doi.org/10.3389/fonc.2021.656184
_version_ 1783674186435133440
author Zhang, Lin
Fu, Ruoqiu
Duan, Dongyu
Li, Ziwei
Li, Bin
Ming, Yue
Li, Li
Ni, Rui
Chen, Jianhong
author_facet Zhang, Lin
Fu, Ruoqiu
Duan, Dongyu
Li, Ziwei
Li, Bin
Ming, Yue
Li, Li
Ni, Rui
Chen, Jianhong
author_sort Zhang, Lin
collection PubMed
description BACKGROUND: Cyclovirobuxine D (CVBD), a steroidal alkaloid, has multiple pharmacological activities, including anti-cancer activity. However, the anti-cancer effect of CVBD on glioblastoma (GBM) has seldom been investigated. This study explores the activity of CVBD in inducing apoptosis of GBM cells, and examines the related mechanism in depth. METHODS: GBM cell lines (T98G, U251) and normal human astrocytes (HA) were treated with CVBD. Cell viability was examined by CCK-8 assay, and cell proliferation was evaluated by cell colony formation counts. Apoptosis and mitochondrial superoxide were measured by flow cytometry. All protein expression levels were determined by Western blotting. JC-1 and CM-H(2)DCFDA probes were used to evaluate the mitochondrial membrane potential (MMP) change and intracellular ROS generation, respectively. The cell ultrastructure was observed by transmission electron microscope (TEM). Colocalization of cofilin and mitochondria were determined by immunofluorescence assay. RESULTS: CVBD showed a greater anti-proliferation effect on the GBM cell lines, T98G and U251, than normal human astrocytes in dose- and time-dependent manners. CVBD induced apoptosis and mitochondrial damage in GBM cells. We found that CVBD led to mitochondrial translocation of cofilin. Knockdown of cofilin attenuated CVBD-induced apoptosis and mitochondrial damage. Additionally, the generation of ROS and mitochondrial superoxide was also induced by CVBD in a dose-dependent manner. N-acetyl-L-cysteine (NAC) and mitoquinone (MitoQ) pre-treatment reverted CVBD-induced apoptosis and mitochondrial damage. MitoQ pretreatment was able to block the mitochondrial translocation of cofilin caused by CVBD. CONCLUSIONS: Our data revealed that CVBD induced apoptosis and mitochondrial damage in GBM cells. The underlying mechanism is related to mitochondrial translocation of cofilin caused by mitochondrial oxidant stress.
format Online
Article
Text
id pubmed-8018288
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-80182882021-04-03 Cyclovirobuxine D Induces Apoptosis and Mitochondrial Damage in Glioblastoma Cells Through ROS-Mediated Mitochondrial Translocation of Cofilin Zhang, Lin Fu, Ruoqiu Duan, Dongyu Li, Ziwei Li, Bin Ming, Yue Li, Li Ni, Rui Chen, Jianhong Front Oncol Oncology BACKGROUND: Cyclovirobuxine D (CVBD), a steroidal alkaloid, has multiple pharmacological activities, including anti-cancer activity. However, the anti-cancer effect of CVBD on glioblastoma (GBM) has seldom been investigated. This study explores the activity of CVBD in inducing apoptosis of GBM cells, and examines the related mechanism in depth. METHODS: GBM cell lines (T98G, U251) and normal human astrocytes (HA) were treated with CVBD. Cell viability was examined by CCK-8 assay, and cell proliferation was evaluated by cell colony formation counts. Apoptosis and mitochondrial superoxide were measured by flow cytometry. All protein expression levels were determined by Western blotting. JC-1 and CM-H(2)DCFDA probes were used to evaluate the mitochondrial membrane potential (MMP) change and intracellular ROS generation, respectively. The cell ultrastructure was observed by transmission electron microscope (TEM). Colocalization of cofilin and mitochondria were determined by immunofluorescence assay. RESULTS: CVBD showed a greater anti-proliferation effect on the GBM cell lines, T98G and U251, than normal human astrocytes in dose- and time-dependent manners. CVBD induced apoptosis and mitochondrial damage in GBM cells. We found that CVBD led to mitochondrial translocation of cofilin. Knockdown of cofilin attenuated CVBD-induced apoptosis and mitochondrial damage. Additionally, the generation of ROS and mitochondrial superoxide was also induced by CVBD in a dose-dependent manner. N-acetyl-L-cysteine (NAC) and mitoquinone (MitoQ) pre-treatment reverted CVBD-induced apoptosis and mitochondrial damage. MitoQ pretreatment was able to block the mitochondrial translocation of cofilin caused by CVBD. CONCLUSIONS: Our data revealed that CVBD induced apoptosis and mitochondrial damage in GBM cells. The underlying mechanism is related to mitochondrial translocation of cofilin caused by mitochondrial oxidant stress. Frontiers Media S.A. 2021-03-19 /pmc/articles/PMC8018288/ /pubmed/33816313 http://dx.doi.org/10.3389/fonc.2021.656184 Text en Copyright © 2021 Zhang, Fu, Duan, Li, Li, Ming, Li, Ni and Chen http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Oncology
Zhang, Lin
Fu, Ruoqiu
Duan, Dongyu
Li, Ziwei
Li, Bin
Ming, Yue
Li, Li
Ni, Rui
Chen, Jianhong
Cyclovirobuxine D Induces Apoptosis and Mitochondrial Damage in Glioblastoma Cells Through ROS-Mediated Mitochondrial Translocation of Cofilin
title Cyclovirobuxine D Induces Apoptosis and Mitochondrial Damage in Glioblastoma Cells Through ROS-Mediated Mitochondrial Translocation of Cofilin
title_full Cyclovirobuxine D Induces Apoptosis and Mitochondrial Damage in Glioblastoma Cells Through ROS-Mediated Mitochondrial Translocation of Cofilin
title_fullStr Cyclovirobuxine D Induces Apoptosis and Mitochondrial Damage in Glioblastoma Cells Through ROS-Mediated Mitochondrial Translocation of Cofilin
title_full_unstemmed Cyclovirobuxine D Induces Apoptosis and Mitochondrial Damage in Glioblastoma Cells Through ROS-Mediated Mitochondrial Translocation of Cofilin
title_short Cyclovirobuxine D Induces Apoptosis and Mitochondrial Damage in Glioblastoma Cells Through ROS-Mediated Mitochondrial Translocation of Cofilin
title_sort cyclovirobuxine d induces apoptosis and mitochondrial damage in glioblastoma cells through ros-mediated mitochondrial translocation of cofilin
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8018288/
https://www.ncbi.nlm.nih.gov/pubmed/33816313
http://dx.doi.org/10.3389/fonc.2021.656184
work_keys_str_mv AT zhanglin cyclovirobuxinedinducesapoptosisandmitochondrialdamageinglioblastomacellsthroughrosmediatedmitochondrialtranslocationofcofilin
AT furuoqiu cyclovirobuxinedinducesapoptosisandmitochondrialdamageinglioblastomacellsthroughrosmediatedmitochondrialtranslocationofcofilin
AT duandongyu cyclovirobuxinedinducesapoptosisandmitochondrialdamageinglioblastomacellsthroughrosmediatedmitochondrialtranslocationofcofilin
AT liziwei cyclovirobuxinedinducesapoptosisandmitochondrialdamageinglioblastomacellsthroughrosmediatedmitochondrialtranslocationofcofilin
AT libin cyclovirobuxinedinducesapoptosisandmitochondrialdamageinglioblastomacellsthroughrosmediatedmitochondrialtranslocationofcofilin
AT mingyue cyclovirobuxinedinducesapoptosisandmitochondrialdamageinglioblastomacellsthroughrosmediatedmitochondrialtranslocationofcofilin
AT lili cyclovirobuxinedinducesapoptosisandmitochondrialdamageinglioblastomacellsthroughrosmediatedmitochondrialtranslocationofcofilin
AT nirui cyclovirobuxinedinducesapoptosisandmitochondrialdamageinglioblastomacellsthroughrosmediatedmitochondrialtranslocationofcofilin
AT chenjianhong cyclovirobuxinedinducesapoptosisandmitochondrialdamageinglioblastomacellsthroughrosmediatedmitochondrialtranslocationofcofilin