Critical role of H2O2 generated by NOX4 during cellular response under glucose deprivation.

Glucose is the most efficient energy source, and various cancer cells depend on glycolysis for energy production. For maintenance of survival and proliferation, glucose sensing and adaptation to poor nutritional circumstances must be well organized in cancer cells. While the glucose sensing machiner...

Full description

Bibliographic Details
Main Authors: Satoshi Owada, Yuko Shimoda, Katsuya Tsuchihara, Hiroyasu Esumi
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2013-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3605446?pdf=render
id doaj-a1c7acaf47084bb894639be78f56dfe9
record_format Article
spelling doaj-a1c7acaf47084bb894639be78f56dfe92020-11-25T02:32:28ZengPublic Library of Science (PLoS)PLoS ONE1932-62032013-01-0183e5662810.1371/journal.pone.0056628Critical role of H2O2 generated by NOX4 during cellular response under glucose deprivation.Satoshi OwadaYuko ShimodaKatsuya TsuchiharaHiroyasu EsumiGlucose is the most efficient energy source, and various cancer cells depend on glycolysis for energy production. For maintenance of survival and proliferation, glucose sensing and adaptation to poor nutritional circumstances must be well organized in cancer cells. While the glucose sensing machinery has been well studied in yeasts, the molecular mechanism of glucose sensing in mammalian cells remains to be elucidated. We have reported glucose deprivation rapidly induces AKT phosphorylation through PI3K activation. We assumed that regulation of AKT is relevant to glucose sensing and further investigated the underlying mechanisms. In this study, AKT phosphorylation under glucose deprivation was inhibited by galactose and fructose, but induced by 2-deoxyglucose (2-DG). Both 2-DG treatment and glucose deprivation were found to induce AKT phosphorylation in HepG2 cells. These findings suggested that glucose transporter may not be involved in the sensing of glucose and induction of AKT phosphorylation, and that downstream metabolic events may have important roles. A variety of metabolic stresses reportedly induce the production of reactive oxygen species (ROS). In the present study, glucose deprivation was found to induce intracellular hydrogen peroxide (H2O2) production in HepG2 cells. N-acetylcysteine (NAC), an antioxidant reagent, reduced both the increase in cellular H2O2 levels and AKT phosphorylation induced by glucose deprivation. These results strongly suggest that the glucose deprivation-induced increase of H2O2 in the cells mediated the AKT phosphorylation. RNA interference of NOX4, but not of NOX5, completely suppressed the glucose deprivation-induced AKT phosphorylation as well as increase of the intracellular levels of ROS, whereas exogenous H2O2 could still induce AKT phosphorylation in the NOX4-knockdown cells. In this study, we demonstrated that the ROS generated by NOX4 are involved in the intracellular adaptive responses by recognizing metabolic flux.http://europepmc.org/articles/PMC3605446?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Satoshi Owada
Yuko Shimoda
Katsuya Tsuchihara
Hiroyasu Esumi
spellingShingle Satoshi Owada
Yuko Shimoda
Katsuya Tsuchihara
Hiroyasu Esumi
Critical role of H2O2 generated by NOX4 during cellular response under glucose deprivation.
PLoS ONE
author_facet Satoshi Owada
Yuko Shimoda
Katsuya Tsuchihara
Hiroyasu Esumi
author_sort Satoshi Owada
title Critical role of H2O2 generated by NOX4 during cellular response under glucose deprivation.
title_short Critical role of H2O2 generated by NOX4 during cellular response under glucose deprivation.
title_full Critical role of H2O2 generated by NOX4 during cellular response under glucose deprivation.
title_fullStr Critical role of H2O2 generated by NOX4 during cellular response under glucose deprivation.
title_full_unstemmed Critical role of H2O2 generated by NOX4 during cellular response under glucose deprivation.
title_sort critical role of h2o2 generated by nox4 during cellular response under glucose deprivation.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2013-01-01
description Glucose is the most efficient energy source, and various cancer cells depend on glycolysis for energy production. For maintenance of survival and proliferation, glucose sensing and adaptation to poor nutritional circumstances must be well organized in cancer cells. While the glucose sensing machinery has been well studied in yeasts, the molecular mechanism of glucose sensing in mammalian cells remains to be elucidated. We have reported glucose deprivation rapidly induces AKT phosphorylation through PI3K activation. We assumed that regulation of AKT is relevant to glucose sensing and further investigated the underlying mechanisms. In this study, AKT phosphorylation under glucose deprivation was inhibited by galactose and fructose, but induced by 2-deoxyglucose (2-DG). Both 2-DG treatment and glucose deprivation were found to induce AKT phosphorylation in HepG2 cells. These findings suggested that glucose transporter may not be involved in the sensing of glucose and induction of AKT phosphorylation, and that downstream metabolic events may have important roles. A variety of metabolic stresses reportedly induce the production of reactive oxygen species (ROS). In the present study, glucose deprivation was found to induce intracellular hydrogen peroxide (H2O2) production in HepG2 cells. N-acetylcysteine (NAC), an antioxidant reagent, reduced both the increase in cellular H2O2 levels and AKT phosphorylation induced by glucose deprivation. These results strongly suggest that the glucose deprivation-induced increase of H2O2 in the cells mediated the AKT phosphorylation. RNA interference of NOX4, but not of NOX5, completely suppressed the glucose deprivation-induced AKT phosphorylation as well as increase of the intracellular levels of ROS, whereas exogenous H2O2 could still induce AKT phosphorylation in the NOX4-knockdown cells. In this study, we demonstrated that the ROS generated by NOX4 are involved in the intracellular adaptive responses by recognizing metabolic flux.
url http://europepmc.org/articles/PMC3605446?pdf=render
work_keys_str_mv AT satoshiowada criticalroleofh2o2generatedbynox4duringcellularresponseunderglucosedeprivation
AT yukoshimoda criticalroleofh2o2generatedbynox4duringcellularresponseunderglucosedeprivation
AT katsuyatsuchihara criticalroleofh2o2generatedbynox4duringcellularresponseunderglucosedeprivation
AT hiroyasuesumi criticalroleofh2o2generatedbynox4duringcellularresponseunderglucosedeprivation
_version_ 1724818933369798656