Cargando…

Knockdown of Nrf2 radiosensitizes glioma cells by inducing redox stress and apoptosis in hypoxia

BACKGROUND: Radiotherapy remains a mainstream treatment for patients with glioma. Yet intrinsic radioresistance has largely compromised the efficacy of the treatment. Increasing concerns have been raised that overexpression of the Nrf2, along with a hypoxic tumor microenvironment, may have contribut...

Descripción completa

Detalles Bibliográficos
Autores principales: Tang, Ting, Jia, Yue, Liang, Hui, Han, Yanling, Cong, Zixiang, Wang, Handong, Ji, Xiangjun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: AME Publishing Company 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9745379/
https://www.ncbi.nlm.nih.gov/pubmed/36523295
http://dx.doi.org/10.21037/tcr-22-1420
_version_ 1784849140538146816
author Tang, Ting
Jia, Yue
Liang, Hui
Han, Yanling
Cong, Zixiang
Wang, Handong
Ji, Xiangjun
author_facet Tang, Ting
Jia, Yue
Liang, Hui
Han, Yanling
Cong, Zixiang
Wang, Handong
Ji, Xiangjun
author_sort Tang, Ting
collection PubMed
description BACKGROUND: Radiotherapy remains a mainstream treatment for patients with glioma. Yet intrinsic radioresistance has largely compromised the efficacy of the treatment. Increasing concerns have been raised that overexpression of the Nrf2, along with a hypoxic tumor microenvironment, may have contributed to the deterioration of radiotherapy in tumors. So, this study investigated the role of Nrf2 in the radiation therapy of glioma cells in hypoxia. METHODS: To determine the expression levels of Nrf2 and HIF-1α, surgical mastectomy specimens from patients with glioma in our institute were analyzed by immunohistochemical staining. Glioblastoma multiforme (GBM) cell lines U251 and U87 with Nrf2 knocked down were produced by transfection with lentiviral particles. Cell lines were treated with ionizing radiation in hypoxia in vitro, with expression and activity of Nrf2 examined by polymerase chain reaction and western blot. Reactive oxygen species (ROS) generation and cell apoptosis analysis were analyzed by flow cytometry. RESULTS: Nrf2 and its downstream pathway were upregulated in surgical specimens after radiotherapy, verified by GBM cell lines treated with in vitro ionizing radiation in hypoxia. Furthermore, knockdown of Nrf2 could induce the ROS generation and cell apoptosis levels after radiation. CONCLUSIONS: Downregulation of Nrf2 could sensitize the lethal effect on GBM cells in vitro by enhancing oxidative stress and apoptosis in hypoxia.
format Online
Article
Text
id pubmed-9745379
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher AME Publishing Company
record_format MEDLINE/PubMed
spelling pubmed-97453792022-12-14 Knockdown of Nrf2 radiosensitizes glioma cells by inducing redox stress and apoptosis in hypoxia Tang, Ting Jia, Yue Liang, Hui Han, Yanling Cong, Zixiang Wang, Handong Ji, Xiangjun Transl Cancer Res Original Article BACKGROUND: Radiotherapy remains a mainstream treatment for patients with glioma. Yet intrinsic radioresistance has largely compromised the efficacy of the treatment. Increasing concerns have been raised that overexpression of the Nrf2, along with a hypoxic tumor microenvironment, may have contributed to the deterioration of radiotherapy in tumors. So, this study investigated the role of Nrf2 in the radiation therapy of glioma cells in hypoxia. METHODS: To determine the expression levels of Nrf2 and HIF-1α, surgical mastectomy specimens from patients with glioma in our institute were analyzed by immunohistochemical staining. Glioblastoma multiforme (GBM) cell lines U251 and U87 with Nrf2 knocked down were produced by transfection with lentiviral particles. Cell lines were treated with ionizing radiation in hypoxia in vitro, with expression and activity of Nrf2 examined by polymerase chain reaction and western blot. Reactive oxygen species (ROS) generation and cell apoptosis analysis were analyzed by flow cytometry. RESULTS: Nrf2 and its downstream pathway were upregulated in surgical specimens after radiotherapy, verified by GBM cell lines treated with in vitro ionizing radiation in hypoxia. Furthermore, knockdown of Nrf2 could induce the ROS generation and cell apoptosis levels after radiation. CONCLUSIONS: Downregulation of Nrf2 could sensitize the lethal effect on GBM cells in vitro by enhancing oxidative stress and apoptosis in hypoxia. AME Publishing Company 2022-11 /pmc/articles/PMC9745379/ /pubmed/36523295 http://dx.doi.org/10.21037/tcr-22-1420 Text en 2022 Translational Cancer Research. All rights reserved. https://creativecommons.org/licenses/by-nc-nd/4.0/Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0 (https://creativecommons.org/licenses/by-nc-nd/4.0/) .
spellingShingle Original Article
Tang, Ting
Jia, Yue
Liang, Hui
Han, Yanling
Cong, Zixiang
Wang, Handong
Ji, Xiangjun
Knockdown of Nrf2 radiosensitizes glioma cells by inducing redox stress and apoptosis in hypoxia
title Knockdown of Nrf2 radiosensitizes glioma cells by inducing redox stress and apoptosis in hypoxia
title_full Knockdown of Nrf2 radiosensitizes glioma cells by inducing redox stress and apoptosis in hypoxia
title_fullStr Knockdown of Nrf2 radiosensitizes glioma cells by inducing redox stress and apoptosis in hypoxia
title_full_unstemmed Knockdown of Nrf2 radiosensitizes glioma cells by inducing redox stress and apoptosis in hypoxia
title_short Knockdown of Nrf2 radiosensitizes glioma cells by inducing redox stress and apoptosis in hypoxia
title_sort knockdown of nrf2 radiosensitizes glioma cells by inducing redox stress and apoptosis in hypoxia
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9745379/
https://www.ncbi.nlm.nih.gov/pubmed/36523295
http://dx.doi.org/10.21037/tcr-22-1420
work_keys_str_mv AT tangting knockdownofnrf2radiosensitizesgliomacellsbyinducingredoxstressandapoptosisinhypoxia
AT jiayue knockdownofnrf2radiosensitizesgliomacellsbyinducingredoxstressandapoptosisinhypoxia
AT lianghui knockdownofnrf2radiosensitizesgliomacellsbyinducingredoxstressandapoptosisinhypoxia
AT hanyanling knockdownofnrf2radiosensitizesgliomacellsbyinducingredoxstressandapoptosisinhypoxia
AT congzixiang knockdownofnrf2radiosensitizesgliomacellsbyinducingredoxstressandapoptosisinhypoxia
AT wanghandong knockdownofnrf2radiosensitizesgliomacellsbyinducingredoxstressandapoptosisinhypoxia
AT jixiangjun knockdownofnrf2radiosensitizesgliomacellsbyinducingredoxstressandapoptosisinhypoxia