NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme.

Cycling and chronic tumor hypoxia are involved in tumor development and growth. However, the impact of cycling hypoxia and its molecular mechanism on glioblastoma multiforme (GBM) progression remain unclear.Glioblastoma cell lines, GBM8401 and U87, and their xenografts were exposed to cycling hypoxi...

Full description

Bibliographic Details
Main Authors: Chia-Hung Hsieh, Woei-Cherng Shyu, Chien-Yi Chiang, Jung-Wen Kuo, Wu-Chung Shen, Ren-Shyan Liu
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2011-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3174133?pdf=render
id doaj-8abf36c4dc3c4ba595b75f1e876e14e8
record_format Article
spelling doaj-8abf36c4dc3c4ba595b75f1e876e14e82020-11-25T00:24:08ZengPublic Library of Science (PLoS)PLoS ONE1932-62032011-01-0169e2394510.1371/journal.pone.0023945NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme.Chia-Hung HsiehWoei-Cherng ShyuChien-Yi ChiangJung-Wen KuoWu-Chung ShenRen-Shyan LiuCycling and chronic tumor hypoxia are involved in tumor development and growth. However, the impact of cycling hypoxia and its molecular mechanism on glioblastoma multiforme (GBM) progression remain unclear.Glioblastoma cell lines, GBM8401 and U87, and their xenografts were exposed to cycling hypoxic stress in vitro and in vivo. Reactive oxygen species (ROS) production in glioblastoma cells and xenografts was assayed by in vitro ROS analysis and in vivo molecular imaging studies. NADPH oxidase subunit 4 (Nox4) RNAi-knockdown technology was utilized to study the role of Nox4 in cycling hypoxia-mediated ROS production and tumor progression. Furthermore, glioblastoma cells were stably transfected with a retroviral vector bearing a dual reporter gene cassette that allowed for dynamic monitoring of HIF-1 signal transduction and tumor cell growth in vitro and in vivo, using optical and nuclear imaging. Tempol, an antioxidant compound, was used to investigate the impact of ROS on cycling hypoxia-mediated HIF-1 activation and tumor progression.Glioblastoma cells and xenografts were compared under cycling hypoxic and normoxic conditions; upregulation of NOX4 expression and ROS levels were observed under cycling hypoxia in glioblastoma cells and xenografts, concomitant with increased tumor cell growth in vitro and in vivo. However, knockdown of Nox4 inhibited these effects. Moreover, in vivo molecular imaging studies demonstrated that Tempol is a good antioxidant compound for inhibiting cycling hypoxia-mediated ROS production, HIF-1 activation, and tumor growth. Immunofluorescence imaging and flow cytometric analysis for NOX4, HIF-1 activation, and Hoechst 3342 in glioblastoma also revealed high localized NOX4 expression predominantly in potentially cycling hypoxic areas with HIF-1 activation and blood perfusion within the endogenous solid tumor microenvironment.Cycling hypoxia-induced ROS via Nox4 is a critical aspect of cancer biology to consider for therapeutic targeting of cycling hypoxia-promoted HIF-1 activation and tumor progression in GBM.http://europepmc.org/articles/PMC3174133?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Chia-Hung Hsieh
Woei-Cherng Shyu
Chien-Yi Chiang
Jung-Wen Kuo
Wu-Chung Shen
Ren-Shyan Liu
spellingShingle Chia-Hung Hsieh
Woei-Cherng Shyu
Chien-Yi Chiang
Jung-Wen Kuo
Wu-Chung Shen
Ren-Shyan Liu
NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme.
PLoS ONE
author_facet Chia-Hung Hsieh
Woei-Cherng Shyu
Chien-Yi Chiang
Jung-Wen Kuo
Wu-Chung Shen
Ren-Shyan Liu
author_sort Chia-Hung Hsieh
title NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme.
title_short NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme.
title_full NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme.
title_fullStr NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme.
title_full_unstemmed NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme.
title_sort nadph oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2011-01-01
description Cycling and chronic tumor hypoxia are involved in tumor development and growth. However, the impact of cycling hypoxia and its molecular mechanism on glioblastoma multiforme (GBM) progression remain unclear.Glioblastoma cell lines, GBM8401 and U87, and their xenografts were exposed to cycling hypoxic stress in vitro and in vivo. Reactive oxygen species (ROS) production in glioblastoma cells and xenografts was assayed by in vitro ROS analysis and in vivo molecular imaging studies. NADPH oxidase subunit 4 (Nox4) RNAi-knockdown technology was utilized to study the role of Nox4 in cycling hypoxia-mediated ROS production and tumor progression. Furthermore, glioblastoma cells were stably transfected with a retroviral vector bearing a dual reporter gene cassette that allowed for dynamic monitoring of HIF-1 signal transduction and tumor cell growth in vitro and in vivo, using optical and nuclear imaging. Tempol, an antioxidant compound, was used to investigate the impact of ROS on cycling hypoxia-mediated HIF-1 activation and tumor progression.Glioblastoma cells and xenografts were compared under cycling hypoxic and normoxic conditions; upregulation of NOX4 expression and ROS levels were observed under cycling hypoxia in glioblastoma cells and xenografts, concomitant with increased tumor cell growth in vitro and in vivo. However, knockdown of Nox4 inhibited these effects. Moreover, in vivo molecular imaging studies demonstrated that Tempol is a good antioxidant compound for inhibiting cycling hypoxia-mediated ROS production, HIF-1 activation, and tumor growth. Immunofluorescence imaging and flow cytometric analysis for NOX4, HIF-1 activation, and Hoechst 3342 in glioblastoma also revealed high localized NOX4 expression predominantly in potentially cycling hypoxic areas with HIF-1 activation and blood perfusion within the endogenous solid tumor microenvironment.Cycling hypoxia-induced ROS via Nox4 is a critical aspect of cancer biology to consider for therapeutic targeting of cycling hypoxia-promoted HIF-1 activation and tumor progression in GBM.
url http://europepmc.org/articles/PMC3174133?pdf=render
work_keys_str_mv AT chiahunghsieh nadphoxidasesubunit4mediatedreactiveoxygenspeciescontributetocyclinghypoxiapromotedtumorprogressioninglioblastomamultiforme
AT woeicherngshyu nadphoxidasesubunit4mediatedreactiveoxygenspeciescontributetocyclinghypoxiapromotedtumorprogressioninglioblastomamultiforme
AT chienyichiang nadphoxidasesubunit4mediatedreactiveoxygenspeciescontributetocyclinghypoxiapromotedtumorprogressioninglioblastomamultiforme
AT jungwenkuo nadphoxidasesubunit4mediatedreactiveoxygenspeciescontributetocyclinghypoxiapromotedtumorprogressioninglioblastomamultiforme
AT wuchungshen nadphoxidasesubunit4mediatedreactiveoxygenspeciescontributetocyclinghypoxiapromotedtumorprogressioninglioblastomamultiforme
AT renshyanliu nadphoxidasesubunit4mediatedreactiveoxygenspeciescontributetocyclinghypoxiapromotedtumorprogressioninglioblastomamultiforme
_version_ 1725353929359753216