Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancer Sci ; 112(8): 3349-3362, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34014591

ABSTRACT

Epstein-Barr virus (EBV) is associated with approximately 10% of gastric cancers (GCs). We previously showed that EBV infection of gastric epithelial cells induces aberrant DNA methylation in promoter regions, which causes silencing of critical tumor suppressor genes. Here, we analyzed gene expressions and active histone modifications (H3K4me3, H3K4me1, and H3K27ac) genome-widely in EBV-positive GC cell lines and in vitro EBV-infected GC cell lines to elucidate the transcription factors contributing to tumorigenesis through enhancer activation. Genes associated with "signaling of WNT in cancer" were significantly enriched in EBV-positive GC, showing increased active ß-catenin staining. Genes neighboring activated enhancers were significantly upregulated, and EHF motif was significantly enriched in these active enhancers. Higher expression of EHF in clinical EBV-positive GC compared with normal tissue and EBV-negative GC was confirmed by RNA-seq using The Cancer Genome Atlas cohort, and by immunostaining using our cohort. EHF knockdown markedly inhibited cell proliferation. Moreover, there was significant enrichment of critical cancer pathway-related genes (eg, FZD5) in the downstream of EHF. EBV protein LMP2A caused upregulation of EHF via phosphorylation of STAT3. STAT3 knockdown was shown to inhibit cellular growth of EBV-positive GC cells, and the inhibition was rescued by EHF overexpression. Our data highlighted the important role of EBV infection in gastric tumorigenesis via enhancer activation.


Subject(s)
Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/metabolism , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/virology , Transcription Factors/genetics , Viral Matrix Proteins/metabolism , Cell Line, Tumor , DNA Methylation , Epstein-Barr Virus Infections/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Histone Code , Humans , Phosphorylation , Sequence Analysis, RNA , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Up-Regulation
2.
Digestion ; 102(1): 25-32, 2021.
Article in English | MEDLINE | ID: mdl-33070127

ABSTRACT

BACKGROUND: Gastric cancer (GC) is one of the leading causes of cancer-related deaths worldwide. GC is a pathologically and molecularly heterogeneous disease. DNA hypermethylation in promoter CpG islands causes silencing of tumor-suppressor genes and thus contributes to gastric carcinogenesis. In addition, various molecular aberrations, including aberrant chromatin structures, gene mutations, structural variants, and somatic copy number alterations, are involved in gastric carcinogenesis. SUMMARY: Comprehensive DNA methylation analyses revealed multiple DNA methylation patterns in GCs and classified GC into distinct molecular subgroups: extremely high-methylation epigenotype uniquely observed in GC associated with Epstein-Barr virus (EBV), high-methylation epigenotype associated with microsatellite instability (MSI), and low-methylation epigenotype. In The Cancer Genome Atlas classification, EBV and MSI are extracted as independent subgroups of GC, whereas the remaining GCs are categorized into genomically stable (GS) and chromosomal instability (CIN) subgroups. EBV-positive GC, exhibiting the most extreme DNA hypermethylation in the whole human malignancies, frequently shows CDKN2A silencing, PIK3CA mutations, PD-L1/2 overexpression, and lack of TP53 mutations. MSI, exhibiting high DNA methylation, often has MLH1 silencing and abundant gene mutations. GS is generally a diffuse-type GC and frequently shows CDH1/RHOA mutations or CLDN18-ARHGAP fusion. CIN is generally an intestinal-type GC and frequently has TP53 mutations and genomic amplification of receptor tyrosine kinases. Key Messages: The frequency and targets of genetic aberrations vary depending on the epigenotype. Aberrations in the genome and epigenome are expected to synergistically interact and contribute to gastric carcinogenesis and comprehensive analyses of those in GCs may help elucidate the mechanism of carcinogenesis.


Subject(s)
Epstein-Barr Virus Infections , Stomach Neoplasms , Claudins , CpG Islands , DNA Methylation , Herpesvirus 4, Human/genetics , Humans , Stomach Neoplasms/genetics
3.
Cancer Sci ; 111(5): 1818-1828, 2020 May.
Article in English | MEDLINE | ID: mdl-32119176

ABSTRACT

Epstein-Barr virus (EBV) is associated with particular forms of gastric cancer (GC). We previously showed that EBV infection into gastric epithelial cells induced aberrant DNA hypermethylation in promoter regions and silencing of tumor suppressor genes. We here undertook integrated analyses of transcriptome and epigenome alteration during EBV infection in gastric cells, to investigate activation of enhancer regions and related transcription factors (TFs) that could contribute to tumorigenesis. Formaldehyde-assisted isolation of regulatory elements (FAIRE) sequencing (-seq) data revealed 19 992 open chromatin regions in putative H3K4me1+ H3K4me3- enhancers in EBV-infected MKN7 cells (MKN7_EB), with 10 260 regions showing increase of H3K27ac. Motif analysis showed candidate TFs, eg activating transcription factor 3 (ATF3), to possibly bind to these activated enhancers. ATF3 was considerably upregulated in MKN7_EB due to EBV factors including EBV-determined nuclear antigen 1 (EBNA1), EBV-encoded RNA 1, and latent membrane protein 2A. Expression of mutant EBNA1 decreased copy number of the EBV genome, resulting in relative downregulation of ATF3 expression. Epstein-Barr virus was also infected into normal gastric epithelial cells, GES1, confirming upregulation of ATF3. Chromatin immunoprecipitation-seq analysis on ATF3 binding sites and RNA-seq analysis on ATF3 knocked-down MKN7_EB revealed 96 genes targeted by ATF3-activating enhancers, which are related with cancer hallmarks, eg evading growth suppressors. These 96 ATF3 target genes were significantly upregulated in MKN7_EB compared with MKN7 and significantly downregulated when ATF3 was knocked down in EBV-positive GC cells SNU719 and NCC24. Knockdown of ATF3 in EBV-infected MKN7, SNU719, and NCC24 cells all led to significant decrease of cellular growth through an increase of apoptotic cells. These indicate that enhancer activation though ATF3 might contribute to tumorigenesis of EBV-positive GC.


Subject(s)
Activating Transcription Factor 3/metabolism , Enhancer Elements, Genetic , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/physiology , Stomach Neoplasms/genetics , Activating Transcription Factor 3/genetics , Apoptosis/genetics , Binding Sites , Cell Line , Cell Proliferation/genetics , Epigenome , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression , Herpesvirus 4, Human/genetics , Humans , Mutation , Transcriptome
4.
Ann Diagn Pathol ; 44: 151456, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31862523

ABSTRACT

Gastric adenocarcinoma (GA) with enteroblastic differentiation is a subset of gastric cancer with poor prognosis. RNA-Seq data of The Cancer Genome Atlas of GA (TCGA-STAD) revealed a positive correlation between SALL4, a representative enteroblastic marker, and DNMT3A expression. Here, we conducted immunohistochemical analysis of GA to clarify the clinicopathological significance of DNMT3A expression and its correlation with enteroblastic differentiation. Of the 346 cases of solitary GA analyzed, 120 (34.7%) showed unequivocal DNMT3A nuclear expression. DNMT3A expression was associated with Lauren's intestinal type, papillary and tubular architectures, high frequency of lymphatic and vascular invasion, and lymph node metastasis (each, P < 0.01). Log-rank test revealed that DNMT3A-positive cases recurred more frequently with a predilection for liver metastasis (P < 0.01) and showed poorer overall and recurrence-free survival (each, P < 0.05). With respect to surrogate markers of molecular subtypes, DNMT3A-positive cases more frequently showed p53 overexpression (P < 0.001). Consistent with the results of TCGA data analysis, DNMT3A-positive cases exhibited enteroblastic morphology (18.3% vs. 0.9%, P < 0.001) and expressed enteroblastic markers, SALL4 (32.5% vs. 3.1%, P < 0.001) and glypican-3 (22.5% vs. 4.4%, P < 0.001) more frequently than did DNMT3A-negative cases. Additionally, GAs showing enteroblastic differentiation, morphologically or immunohistochemically, expressed DNMT3A with significantly higher frequency and intensity than did conventional GAs (P < 0.001). Our findings suggest DNMT3A as a potential therapeutic target for this conventional therapy-refractory cancer subtype.


Subject(s)
Biomarkers, Tumor/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Neoplastic , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Cell Differentiation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Female , Glypicans/genetics , Glypicans/metabolism , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Stomach Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , DNA Methyltransferase 3B
5.
Oncotarget ; 8(33): 55265-55279, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28903418

ABSTRACT

Aberrant DNA hypermethylation is a major epigenetic mechanism to inactivate tumor suppressor genes in cancer. Epstein-Barr virus positive gastric cancer is the most frequently hypermethylated tumor among human malignancies. Herein, we performed comprehensive analysis of epigenomic alteration during EBV infection, by Infinium HumanMethylation 450K BeadChip for DNA methylation and ChIP-sequencing for histone modification alteration during EBV infection into gastric cancer cell line MKN7. Among 7,775 genes with increased DNA methylation in promoter regions, roughly half were "DNA methylation-sensitive" genes, which acquired DNA methylation in the whole promoter regions and thus were repressed. These included anti-oncogenic genes, e.g. CDKN2A. The other half were "DNA methylation-resistant" genes, where DNA methylation is acquired in the surrounding of promoter regions, but unmethylated status is protected in the vicinity of transcription start site. These genes thereby retained gene expression, and included DNA repair genes. Histone modification was altered dynamically and coordinately with DNA methylation alteration. DNA methylation-sensitive genes significantly correlated with loss of H3K27me3 pre-marks or decrease of active histone marks, H3K4me3 and H3K27ac. Apoptosis-related genes were significantly enriched in these epigenetically repressed genes. Gain of active histone marks significantly correlated with DNA methylation-resistant genes. Genes related to mitotic cell cycle and DNA repair were significantly enriched in these epigenetically activated genes. Our data show that orchestrated epigenetic alterations are important in gene regulation during EBV infection, and histone modification status in promoter regions significantly associated with acquisition of de novo DNA methylation or protection of unmethylated status at transcription start site.

6.
Sci Rep ; 7(1): 7924, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28801683

ABSTRACT

Epstein-Barr virus (EBV) infection is associated with tumours such as Burkitt lymphoma, nasopharyngeal carcinoma, and gastric cancer. We previously showed that EBV(+) gastric cancer presents an extremely high-methylation epigenotype and this aberrant DNA methylation causes silencing of multiple tumour suppressor genes. However, the mechanisms that drive EBV infection-mediated tumorigenesis, including other epigenomic alteration, remain unclear. We analysed epigenetic alterations induced by EBV infection especially at enhancer regions, to elucidate their contribution to tumorigenesis. We performed ChIP sequencing on H3K4me3, H3K4me1, H3K27ac, H3K27me3, and H3K9me3 in gastric epithelial cells infected or not with EBV. We showed that repressive marks were redistributed after EBV infection, resulting in aberrant enhancer activation and repression. Enhancer dysfunction led to the activation of pathways related to cancer hallmarks (e.g., resisting cell death, disrupting cellular energetics, inducing invasion, evading growth suppressors, sustaining proliferative signalling, angiogenesis, and tumour-promoting inflammation) and inactivation of tumour suppressive pathways. Deregulation of cancer-related genes in EBV-infected gastric epithelial cells was also observed in clinical EBV(+) gastric cancer specimens. Our analysis showed that epigenetic alteration associated with EBV-infection may contribute to tumorigenesis through enhancer activation and repression.


Subject(s)
Cell Transformation, Viral , Enhancer Elements, Genetic , Epigenesis, Genetic , Epithelial Cells/virology , Herpesvirus 4, Human/growth & development , Stomach Neoplasms/physiopathology , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation , Histones/metabolism , Humans , Models, Biological , Protein Binding , Sequence Analysis, DNA
7.
J Pathol ; 242(4): 391-399, 2017 08.
Article in English | MEDLINE | ID: mdl-28418084

ABSTRACT

Epstein-Barr virus (EBV)-positive gastric cancer (GC) shows a higher DNA methylation epigenotype. EBV infection can causally induce genome-wide aberrant DNA methylation, as previously demonstrated by in vitro infection experiments in the low-methylation GC cell line MKN7. However, whether EBV exerts DNA methylation remodelling properties in non-neoplastic epithelial cells remains unclear. Here we performed post-infection time-series DNA methylation analyses using the immortalized normal gastric epithelial cell line GES1. Genome-wide analysis using Illumina's Infinium 450 k BeadArray demonstrated global de novo DNA methylation from post-infection day 17, which was completed by 28 days in a manner similar to that observed in MKN7 cells. De novo methylation of all types of GC-specific methylation marker genes was observed, indicating that EBV infection is sufficient for gastric epithelial cells to acquire an EBV-positive GC epigenotype. Pyrosequencing demonstrated that methylation of the viral genome preceded that of the host cellular genome, suggesting the existence of well-ordered mechanisms that induce methylation. Spatiotemporal representation with differential models revealed dynamic alterations of DNA methylation in promoter regions, occurring from lower-CpG peripheral regions and extending to higher-CpG core regions. In summary, EBV infection exerted powerful pressure to induce global de novo DNA methylation in non-neoplastic cells within a month in a spatiotemporally well-ordered manner. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
DNA Methylation , Epstein-Barr Virus Infections/genetics , Gastric Mucosa/virology , Cell Line, Transformed , CpG Islands , Epigenesis, Genetic , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Epstein-Barr Virus Infections/pathology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Genome-Wide Association Study , Humans , Promoter Regions, Genetic
8.
Oncotarget ; 7(49): 81512-81526, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27829228

ABSTRACT

Extensive DNA methylation is observed in gastric cancer with Epstein-Barr virus (EBV) infection, and EBV infection is the cause to induce this extensive hypermethylaton phenotype in gastric epithelial cells. However, some 5' regions of genes do not undergo de novo methylation, despite the induction of methylation in surrounding regions, suggesting the existence of a resistance factor against DNA methylation acquisition. We conducted an RNA-seq analysis of gastric epithelial cells with and without EBV infection and found that TET family genes, especially TET2, were repressed by EBV infection at both mRNA and protein levels. TET2 was found to be downregulated by EBV transcripts, e.g. BARF0 and LMP2A, and also by seven human miRNAs targeting TET2, e.g., miR-93 and miR-29a, which were upregulated by EBV infection, and transfection of which into gastric cells repressed TET2. Hydroxymethylation target genes by TET2 were detected by hydroxymethylated DNA immunoprecipitation sequencing (hMeDIP-seq) with and without TET2 overexpression, and overlapped significantly with methylation target genes in EBV-infected cells. When TET2 was knocked down by shRNA, EBV infection induced de novo methylation more severely, including even higher methylation in methylation-acquired promoters or de novo methylation acquisition in methylation-protected promoters, leading to gene repression. TET2 knockdown alone without EBV infection did not induce de novo DNA methylation. These data suggested that TET2 functions as a resistance factor against DNA methylation in gastric epithelial cells and repression of TET2 contributes to DNA methylation acquisition during EBV infection.


Subject(s)
Cell Transformation, Viral , DNA Methylation , DNA-Binding Proteins/metabolism , Epstein-Barr Virus Infections/virology , Gastric Mucosa/virology , Herpesvirus 4, Human/pathogenicity , Proto-Oncogene Proteins/metabolism , Stomach Neoplasms/virology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Dioxygenases , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Gastric Mucosa/metabolism , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Host-Pathogen Interactions , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Time Factors , Transcription, Genetic , Transcriptome , Transfection , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
9.
Nat Microbiol ; 1: 16026, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-27572445

ABSTRACT

Most if not all gastric cancers are associated with chronic infection of the stomach mucosa with Helicobacter pylori cagA-positive strains(1-4). Approximately 10% of gastric cancers also harbour Epstein-Barr virus (EBV) in the cancer cells(5,6). Following delivery into gastric epithelial cells via type IV secretion(7,8), the cagA-encoded CagA protein undergoes tyrosine phosphorylation on the Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs initially by Src family kinases (SFKs) and then by c-Abl(9,10). Tyrosine-phosphorylated CagA binds to the pro-oncogenic protein tyrosine phosphatase SHP2 and thereby deregulates the phosphatase activity(11,12), which has been considered to play an important role in gastric carcinogenesis(13). Here we show that the SHP2 homologue SHP1 interacts with CagA independently of the EPIYA motif. The interaction potentiates the phosphatase activity of SHP1 that dampens the oncogenic action of CagA by dephosphorylating the CagA EPIYA motifs. In vitro infection of gastric epithelial cells with EBV induces SHP1 promoter hypermethylation, which strengthens phosphorylation-dependent CagA action via epigenetic downregulation of SHP1 expression. Clinical specimens of EBV-positive gastric cancers also exhibit SHP1 hypermethylation with reduced SHP1 expression. The results reveal that SHP1 is the long-sought phosphatase that can antagonize CagA. Augmented H. pylori CagA activity, via SHP1 inhibition, might also contribute to the development of EBV-positive gastric cancer.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Helicobacter pylori/pathogenicity , Herpesvirus 4, Human/growth & development , Host-Pathogen Interactions , Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Carcinogenesis , Cell Line, Tumor , DNA Methylation , Epithelial Cells/microbiology , Epithelial Cells/virology , Humans , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Interaction Mapping , Protein Processing, Post-Translational
10.
J Biochem ; 156(2): 63-72, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24962701

ABSTRACT

Aberant DNA methylation is a common epigenomic alteration in carcinogenesis. Comprehensive analyses of DNA methylation have stratified gastrointestinal cancer into several subgroups according to specific DNA methylation accumulation. In gastric cancer, Helicobacter pylori infection is a cause of methylation accumulation in apparently normal mucosa. Epstein-Barr virus infection is another methylation inducer that causes more genome-wide methylation, resulting in the formation of unique epigenotype with extensive methylation. In colorectal carcinogenesis, accumulation of high levels of methylation in combination with BRAF mutation is characteristic of the serrated pathway, but not of the adenoma-carcinoma sequence through conventional adenoma. In a de novo pathway, laterally spreading tumours generate intermediate- and low-methylation epigenotypes, accompanied by different genetic features and different macroscopic morphologies. These methylation epigenotypes, with specific genomic aberrations, are mostly completed by the adenoma stage, and additional molecular aberration, such as TP53 mutation, is suggested to lead to cancer development with the corresponding epigenotype. Accumulation of DNA methylation and formation of the epigenotype is suggested to occur during the early stages of carcinogenesis and predetermines the future cancer type.


Subject(s)
DNA Methylation , Stomach Neoplasms/genetics , Animals , Colorectal Neoplasms/genetics , Epstein-Barr Virus Infections/genetics , Gene Expression Regulation, Neoplastic , Gene-Environment Interaction , Helicobacter Infections/genetics , Humans , Oncogenes , Phenotype , Stomach Neoplasms/microbiology
11.
World J Gastroenterol ; 20(14): 3916-26, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24744581

ABSTRACT

Gastric cancer is a leading cause of cancer death worldwide, and significant effort has been focused on clarifying the pathology of gastric cancer. In particular, the development of genome-wide analysis tools has enabled the detection of genetic and epigenetic alterations in gastric cancer; for example, aberrant DNA methylation in gene promoter regions is thought to play a crucial role in gastric carcinogenesis. The etiological viewpoint is also essential for the study of gastric cancers, and two distinct pathogens, Helicobacter pylori (H. pylori) and Epstein-Barr virus (EBV), are known to participate in gastric carcinogenesis. Chronic inflammation of the gastric epithelium due to H. pylori infection induces aberrant polyclonal methylation that may lead to an increased risk of gastric cancer. In addition, EBV infection is known to cause extensive methylation, and EBV-positive gastric cancers display a high methylation epigenotype, in which aberrant methylation extends to not only Polycomb repressive complex (PRC)-target genes in embryonic stem cells but also non-PRC-target genes. Here, we review aberrant DNA methylation in gastric cancer and the association between methylation and infection with H. pylori and EBV.


Subject(s)
DNA Methylation , Helicobacter pylori , Herpesvirus 4, Human , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Stomach Neoplasms/virology , Chromatin Assembly and Disassembly , CpG Islands , Epigenesis, Genetic , Epithelium/microbiology , Epithelium/virology , Gastric Mucosa/immunology , Gastric Mucosa/virology , Humans , Inflammation , Methylation , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...