Silencing of glutathione peroxidase 3 through DNA hypermethylation is associated with lymph node metastasis in gastric carcinomas.

Gastric cancer remains the second leading cause of cancer-related death in the world. H. pylori infection, a major risk factor for gastric cancer, generates high levels of reactive oxygen species (ROS). Glutathione peroxidase 3 (GPX3), a plasma GPX member and a major scavenger of ROS, catalyzes the...

Full description

Bibliographic Details
Main Authors: Dun-Fa Peng, Tian-Ling Hu, Barbara G Schneider, Zheng Chen, Ze-Kuan Xu, Wael El-Rifai
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2012-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3468580?pdf=render
id doaj-006dc9702f374e82a40b1075ffa3dcc0
record_format Article
spelling doaj-006dc9702f374e82a40b1075ffa3dcc02020-11-25T01:22:07ZengPublic Library of Science (PLoS)PLoS ONE1932-62032012-01-01710e4621410.1371/journal.pone.0046214Silencing of glutathione peroxidase 3 through DNA hypermethylation is associated with lymph node metastasis in gastric carcinomas.Dun-Fa PengTian-Ling HuBarbara G SchneiderZheng ChenZe-Kuan XuWael El-RifaiGastric cancer remains the second leading cause of cancer-related death in the world. H. pylori infection, a major risk factor for gastric cancer, generates high levels of reactive oxygen species (ROS). Glutathione peroxidase 3 (GPX3), a plasma GPX member and a major scavenger of ROS, catalyzes the reduction of hydrogen peroxide and lipid peroxides by reduced glutathione. To study the expression and gene regulation of GPX3, we examined GPX3 gene expression in 9 gastric cancer cell lines, 108 primary gastric cancer samples and 45 normal gastric mucosa adjacent to cancers using quantitative real-time RT-PCR. Downregulation or silencing of GPX3 was detected in 8 of 9 cancer cell lines, 83% (90/108) gastric cancers samples, as compared to non-tumor adjacent normal gastric samples (P<0.0001). Examination of GPX3 promoter demonstrated DNA hypermethylation (≥ 10% methylation level determined by Bisulfite Pyrosequencing) in 6 of 9 cancer cell lines and 60% of gastric cancer samples (P = 0.007). We also detected a significant loss of DNA copy number of GPX3 in gastric cancers (P<0.001). Treatment of SNU1 and MKN28 cells with 5-Aza-2' Deoxycytidine restored the GPX3 gene expression with a significant demethylation of GPX3 promoter. The downregulation of GPX3 expression and GPX3 promoter hypermethylation were significantly associated with gastric cancer lymph node metastasis (P = 0.018 and P = 0.029, respectively). We also observed downregulation, DNA copy number losses, and promoter hypermethylation of GPX3 in approximately one-third of tumor-adjacent normal gastric tissue samples, suggesting the presence of a field defect in areas near tumor samples. Reconstitution of GPX3 in AGS cells reduced the capacity of cell migration, as measured by scratch wound healing assay. Taken together, the dysfunction of GPX3 in gastric cancer is mediated by genetic and epigenetic alterations, suggesting impairment of mechanisms that regulate ROS and its possible involvement in gastric tumorigenesis and metastasis.http://europepmc.org/articles/PMC3468580?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Dun-Fa Peng
Tian-Ling Hu
Barbara G Schneider
Zheng Chen
Ze-Kuan Xu
Wael El-Rifai
spellingShingle Dun-Fa Peng
Tian-Ling Hu
Barbara G Schneider
Zheng Chen
Ze-Kuan Xu
Wael El-Rifai
Silencing of glutathione peroxidase 3 through DNA hypermethylation is associated with lymph node metastasis in gastric carcinomas.
PLoS ONE
author_facet Dun-Fa Peng
Tian-Ling Hu
Barbara G Schneider
Zheng Chen
Ze-Kuan Xu
Wael El-Rifai
author_sort Dun-Fa Peng
title Silencing of glutathione peroxidase 3 through DNA hypermethylation is associated with lymph node metastasis in gastric carcinomas.
title_short Silencing of glutathione peroxidase 3 through DNA hypermethylation is associated with lymph node metastasis in gastric carcinomas.
title_full Silencing of glutathione peroxidase 3 through DNA hypermethylation is associated with lymph node metastasis in gastric carcinomas.
title_fullStr Silencing of glutathione peroxidase 3 through DNA hypermethylation is associated with lymph node metastasis in gastric carcinomas.
title_full_unstemmed Silencing of glutathione peroxidase 3 through DNA hypermethylation is associated with lymph node metastasis in gastric carcinomas.
title_sort silencing of glutathione peroxidase 3 through dna hypermethylation is associated with lymph node metastasis in gastric carcinomas.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2012-01-01
description Gastric cancer remains the second leading cause of cancer-related death in the world. H. pylori infection, a major risk factor for gastric cancer, generates high levels of reactive oxygen species (ROS). Glutathione peroxidase 3 (GPX3), a plasma GPX member and a major scavenger of ROS, catalyzes the reduction of hydrogen peroxide and lipid peroxides by reduced glutathione. To study the expression and gene regulation of GPX3, we examined GPX3 gene expression in 9 gastric cancer cell lines, 108 primary gastric cancer samples and 45 normal gastric mucosa adjacent to cancers using quantitative real-time RT-PCR. Downregulation or silencing of GPX3 was detected in 8 of 9 cancer cell lines, 83% (90/108) gastric cancers samples, as compared to non-tumor adjacent normal gastric samples (P<0.0001). Examination of GPX3 promoter demonstrated DNA hypermethylation (≥ 10% methylation level determined by Bisulfite Pyrosequencing) in 6 of 9 cancer cell lines and 60% of gastric cancer samples (P = 0.007). We also detected a significant loss of DNA copy number of GPX3 in gastric cancers (P<0.001). Treatment of SNU1 and MKN28 cells with 5-Aza-2' Deoxycytidine restored the GPX3 gene expression with a significant demethylation of GPX3 promoter. The downregulation of GPX3 expression and GPX3 promoter hypermethylation were significantly associated with gastric cancer lymph node metastasis (P = 0.018 and P = 0.029, respectively). We also observed downregulation, DNA copy number losses, and promoter hypermethylation of GPX3 in approximately one-third of tumor-adjacent normal gastric tissue samples, suggesting the presence of a field defect in areas near tumor samples. Reconstitution of GPX3 in AGS cells reduced the capacity of cell migration, as measured by scratch wound healing assay. Taken together, the dysfunction of GPX3 in gastric cancer is mediated by genetic and epigenetic alterations, suggesting impairment of mechanisms that regulate ROS and its possible involvement in gastric tumorigenesis and metastasis.
url http://europepmc.org/articles/PMC3468580?pdf=render
work_keys_str_mv AT dunfapeng silencingofglutathioneperoxidase3throughdnahypermethylationisassociatedwithlymphnodemetastasisingastriccarcinomas
AT tianlinghu silencingofglutathioneperoxidase3throughdnahypermethylationisassociatedwithlymphnodemetastasisingastriccarcinomas
AT barbaragschneider silencingofglutathioneperoxidase3throughdnahypermethylationisassociatedwithlymphnodemetastasisingastriccarcinomas
AT zhengchen silencingofglutathioneperoxidase3throughdnahypermethylationisassociatedwithlymphnodemetastasisingastriccarcinomas
AT zekuanxu silencingofglutathioneperoxidase3throughdnahypermethylationisassociatedwithlymphnodemetastasisingastriccarcinomas
AT waelelrifai silencingofglutathioneperoxidase3throughdnahypermethylationisassociatedwithlymphnodemetastasisingastriccarcinomas
_version_ 1725127633762516992