Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
J Pathol ; 238(5): 651-64, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26800240

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) catalyses histone H3 lysine 27 trimethylation (H3K27me3) to silence tumour-suppressor genes in hepatocellular carcinoma (HCC) but the process of locus-specific recruitment remains elusive. Here we investigated the transcription factors involved and the molecular consequences in HCC development. The genome-wide distribution of H3K27me3 was determined by chromatin immunoprecipitation coupled with high-throughput sequencing or promoter array analyses in HCC cells from hepatitis B virus (HBV) X protein transgenic mouse and human cell models. Transcription factor binding site analysis was performed to identify EZH2-interacting transcription factors followed by functional characterization. Our cross-species integrative analysis revealed a crucial link between Yin Yang 1 (YY1) and EZH2-mediated H3K27me3 in HCC. Gene expression analysis of human HBV-associated HCC specimens demonstrated concordant overexpression of YY1 and EZH2, which correlated with poor survival of patients in advanced stages. The YY1 binding motif was significantly enriched in both in vivo and in vitro H3K27me3-occupied genes, including genes for 15 tumour-suppressive microRNAs. Knockdown of YY1 reduced not only global H3K27me3 levels, but also EZH2 and H3K27me3 promoter occupancy and DNA methylation, leading to the transcriptional up-regulation of microRNA-9 isoforms in HCC cells. Concurrent EZH2 knockdown and 5-aza-2'-deoxycytidine treatment synergistically increased the levels of microRNA-9, which reduced the expression and transcriptional activity of nuclear factor-κB (NF-κB). Functionally, YY1 promoted HCC tumourigenicity and inhibited apoptosis of HCC cells, at least partially through NF-κB activation. In conclusion, YY1 overexpression contributes to EZH2 recruitment for H3K27me3-mediated silencing of tumour-suppressive microRNAs, thereby activating NF-κB signalling in hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Silencing , Liver Neoplasms/metabolism , MicroRNAs/metabolism , NF-kappa B/metabolism , YY1 Transcription Factor/metabolism , Animals , Apoptosis , Binding Sites , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Lysine , Methylation , Mice, Nude , Mice, Transgenic , MicroRNAs/genetics , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , RNA Interference , Signal Transduction , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection , Tumor Burden , Up-Regulation , Viral Regulatory and Accessory Proteins , YY1 Transcription Factor/genetics
2.
J Pathol ; 237(1): 38-49, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25925944

ABSTRACT

Hepatocellular carcinoma (HCC) is a worldwide threat to public health, especially in China, where chronic hepatitis B virus (HBV) infection is found in 80-90% of all HCCs. The HBV-encoded X antigen (HBx) is a trans-regulatory protein involved in virus-induced hepatocarcinogenesis. Although the carboxyl-terminus-truncated HBx, rather than the full-length counterpart, is frequently overexpressed in human HCCs, its functional mechanisms are not fully defined. We investigated the molecular function of a naturally occurring HBx variant which has 35 amino acids deleted at the C-terminus (HBxΔ35). Genome-wide scanning analysis and PCR validation identified growth arrest-specific 2 (GAS2) as a direct target of HBxΔ35 at transcriptional level in human immortalized liver cells. HBxΔ35 was found to bind the promoter region of GAS2 and attenuate its expression to promote hepatocellular proliferation and tumourigenicity. Further functional assays demonstrated that GAS2 induces p53-dependent apoptosis and senescence to counteract HBxΔ35-mediated tumourigenesis. Notably, GAS2 expression was significantly down-regulated in HCCs compared with the corresponding normal tissues. In conclusion, our integrated study uncovered a novel viral mechanism in hepatocarcinogenesis, wherein HBxΔ35 deregulates cell growth via direct silencing of GAS2 and thereby provides a survival advantage for pre-neoplastic hepatocytes to facilitate cancer development.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular/metabolism , Cell Cycle , Cell Transformation, Viral , Cellular Senescence , Gene Silencing , Hepatitis B virus/metabolism , Liver Neoplasms/metabolism , Microfilament Proteins/metabolism , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Binding Sites , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Female , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Hepatitis B/complications , Hepatitis B virus/genetics , Hepatitis B virus/pathogenicity , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Mice, Inbred BALB C , Mice, Nude , Microfilament Proteins/genetics , Promoter Regions, Genetic , Signal Transduction , Time Factors , Trans-Activators/genetics , Transcription, Genetic , Transfection , Tumor Burden , Tumor Suppressor Protein p53/genetics , Viral Regulatory and Accessory Proteins
3.
Gastroenterology ; 144(1): 122-133.e9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23058321

ABSTRACT

BACKGROUND & AIMS: Deregulation of forkhead box (Fox) proteins, an evolutionarily conserved family of transcriptional regulators, leads to tumorigenesis. Little is known about their regulation or functions in the pathogenesis of gastric cancer. Promoter hypermethylation occurs during Helicobacter pylori-induced gastritis. We investigated whether the deregulated genes contribute to gastric tumorigenesis. METHODS: We used integrative genome-wide scans to identify concomitant hypermethylated genes in mice infected with H pylori and human gastric cancer samples. We also analyzed epigenetic gene silencing in gastric tissues from patients with H pylori infection and gastritis, intestinal metaplasia, gastric tumors, or without disease (controls). Target genes were identified by chromatin immunoprecipitation microarrays and expression and luciferase reporter analyses. RESULTS: Methylation profile analyses identified the promoter of FOXD3 as the only genomic region with increased methylation in mice and humans during progression of H pylori-associated gastric tumors. FOXD3 methylation also correlated with shorter survival times of patients with gastric cancer. Genome demethylation reactivated FOXD3 expression in gastric cancer cell lines. Transgenic overexpression of FOXD3 significantly inhibited gastric cancer cell proliferation and invasion, and reduced growth of xenograft tumors in mice, at least partially, by promoting tumor cell apoptosis. FOXD3 bound directly to the promoters of, and activated transcription of, genes encoding the cell death regulators CYFIP2 and RARB. Levels of FOXD3, CYFIP2, and RARB messenger RNAs were reduced in human gastric tumor samples, compared with control tissues. CONCLUSIONS: FOXD3-mediated transcriptional control of tumor suppressors is deregulated by H pylori infection-induced hypermethylation; this could perturb the balance between cell death and survival. These findings identify a pathway by which epigenetic changes affect gastric tumor suppression.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Helicobacter Infections/genetics , Helicobacter pylori , Repressor Proteins/genetics , Stomach Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/genetics , DNA Methylation , Epigenesis, Genetic , Gastritis/genetics , Gene Silencing , Humans , Intestines/pathology , Kaplan-Meier Estimate , Male , Metaplasia/genetics , Mice , Mice, Inbred C57BL , Prognosis , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Retinoic Acid/genetics , Stomach Neoplasms/microbiology
4.
Cancer Res ; 71(11): 4028-39, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21512140

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is the catalytic subunit of the Polycomb-repressive complex 2 (PRC2) that represses gene transcription through histone H3 lysine 27 trimethylation (H3K27me3). Although EZH2 is abundantly present in various cancers, the molecular consequences leading to oncogenesis remain unclear. Here, we show that EZH2 concordantly silences the Wnt pathway antagonists operating at several subcellular compartments, which in turn activate Wnt/ß-catenin signaling in hepatocellular carcinomas (HCC). Chromatin immunoprecipitation promoter array and gene expression analyses in HCCs revealed EZH2 occupancy and reduced expression of Wnt antagonists, including the growth-suppressive AXIN2, NKD1, PPP2R2B, PRICKLE1, and SFRP5. Knockdown of EZH2 reduced the promoter occupancy of PRC2, histone deacetylase 1 (HDAC1), and H3K27me3, whereas the activating histone marks were increased, leading to the transcriptional upregulation of the Wnt antagonists. Combinatorial EZH2 and HDAC inhibition dramatically reduced the levels of nuclear ß-catenin, T-cell factor-dependent transcriptional activity, and downstream pro-proliferative targets CCND1 and EGFR. Functional analysis revealed that downregulation of EZH2 reduced HCC cell growth, partially through the inhibition of ß-catenin signaling. Conversely, ectopic overexpression of EZH2 in immortalized hepatocytes activated Wnt/ß-catenin signaling to promote cellular proliferation. In human HCCs, concomitant overexpression of EZH2 and ß-catenin was observed in one-third (61/179) of cases and significantly correlated with tumor progression. Our data indicate that EZH2-mediated epigenetic silencing contributes to constitutive activation of Wnt/ß-catenin signaling and consequential proliferation of HCC cells, thus representing a novel therapeutic target for this highly malignant tumor.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/metabolism , Liver Neoplasms/metabolism , Transcription Factors/metabolism , Wnt Proteins/antagonists & inhibitors , beta Catenin/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/genetics , Down-Regulation , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Polycomb Repressive Complex 2 , Signal Transduction , Transcription Factors/genetics , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...