Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Zeyou Wang is active.

Publication


Featured researches published by Zeyou Wang.


Molecular Cancer | 2011

MiR-185 Targets the DNA Methyltransferases 1 and Regulates Global DNA Methylation in human glioma

Zuping Zhang; Hailin Tang; Zeyou Wang; Baoxin Zhang; Wei Liu; Hongmei Lu; Lan Xiao; Xiaoping Liu; Rong Wang; Xiaoling Li; Minghua Wu; Guiyuan Li

BackgroundPerturbation of DNA methylation is frequent in cancers and has emerged as an important mechanism involved in tumorigenesis. To determine how DNA methylation is modified in the genome of primary glioma, we used Methyl-DNA immunoprecipitation (MeDIP) and Nimblegen CpG promoter microarrays to identify differentially DNA methylation sequences between primary glioma and normal brain tissue samples.MethodsMeDIP-chip technology was used to investigate the whole-genome differential methylation patterns in glioma and normal brain tissues. Subsequently, the promoter methylation status of eight candidate genes was validated in 40 glioma samples and 4 cell lines by Sequenoms MassARRAY system. Then, the epigenetically regulated expression of these genes and the potential mechanisms were examined by chromatin immunoprecipitation and quantitative real-time PCR.ResultsA total of 524 hypermethylated and 104 hypomethylated regions were identified in glioma. Among them, 216 hypermethylated and 60 hypomethylated regions were mapped to the promoters of known genes related to a variety of important cellular processes. Eight promoter-hypermethylated genes (ANKDD1A, GAD1, HIST1H3E, PCDHA8, PCDHA13, PHOX2B, SIX3, and SST) were confirmed in primary glioma and cell lines. Aberrant promoter methylation and changed histone modifications were associated with their reduced expression in glioma. In addition, we found loss of heterozygosity (LOH) at the miR-185 locus located in the 22q11.2 in glioma and induction of miR-185 over-expression reduced global DNA methylation and induced the expression of the promoter-hypermethylated genes in glioma cells by directly targeting the DNA methyltransferases 1.ConclusionThese comprehensive data may provide new insights into the epigenetic pathogenesis of human gliomas.


Current Cancer Drug Targets | 2013

The miR-183/96/182 cluster regulates oxidative apoptosis and sensitizes cells to chemotherapy in gliomas.

Hailin Tang; Yanhui Bian; Chaofeng Tu; Zeyou Wang; Zhibin Yu; Qing Liu; Gang Xu; Minghua Wu; Guiyuan Li

Many microRNAs reside in clusters in the genome, are generally similar in sequence, are transcribed in the same direction, and usually function synergistically. The miR-183/96/182 cluster is composed of 3 miRNA genes, and increased expression of miR-183, 96 and 182 are implicated in glioma carcinogenesis. Knockdown of individual components or of the entire miR-183/96/182 cluster inhibits the survival of glioma cells by regulating the ROS-induced apoptosis pathway. Furthermore, inhibition of the miR-183/96/182 cluster induced ROS-mediated AKT/survival independent of three target genes FGF9, CPEB1, and FOXO1, and inhibition of the miRNA cluster induced p53/apoptosis signaling, which was dependent on these same genes. In addition, knockdown of the miR-183/96/182 cluster enhanced the anticancer effect of Temozolomide on glioma cells by the ROS-mediated apoptosis pathway. Therefore, the miR-183/96/182 cluster may be a pleiotropic target for glioma therapy.


Current Cancer Drug Targets | 2012

LRRC4 inhibits glioma cell growth and invasion through a miR-185-dependent pathway.

Hailin Tang; Zeyou Wang; Xiaoping Liu; Qing Liu; Gang Xu; Guiyuan Li; Minghua Wu

Leucine-rich repeat (LRR) genes encode transmembrane proteins that are essential for normal brain development and are often dysregulated in central nervous system tumors. Leucine-rich repeat C4 (LRRC4) is a member of the LRR protein superfamily and specifically expressed in brain tissue. Importantly it acts as a tumor suppressor in the pathogenesis of malignant gliomas. However, the molecular mechanisms by which LRRC4 regulates glioma tumorigenesis are largely unknown. In this report, we found that miR-185 is markedly upregulated by LRRC4. We also found that miR-185 was downregulated in glioma, and overexpression of miR-185 inhibited glioma cell invasion. Low expressions of LRRC4 and miR-185 were associated with a poor outcome in glioma patients. Further investigation revealed that LRRC4 mediated its tumor suppressor function by regulating miR-185 targets CDC42 and RhoA. LRRC4 overexpression inhibited glioma cell invasion through miR-185-mediated CDC42 and RhoA direct regulation and VEGFA indirect regulation. Together, our findings suggest that the altered expression of the tumor suppressor LRRC4 may be an important event that leads to the dysregulation of miR-185 in human gliomas. LRRC4 and miR-185 may also be good prognostic markers and therapeutic targets in glioma.


PLOS ONE | 2014

Disturbing miR-182 and -381 Inhibits BRD7 Transcription and Glioma Growth by Directly Targeting LRRC4

Hailin Tang; Zeyou Wang; Qing Liu; Xiaoping Liu; Minghua Wu; Guiyuan Li

Inactivated LRRC4 has been clinically detected in gliomas, and promoter hypermethylation has been implicated as the mechanism of inactivation in some of those tumors. Our previous researches indicated that LRRC4 is a target gene of miR-381, the interaction of miR-381 and LRRC4 is involved in glioma growth. In this study, we demonstrate that LRRC4 is a target gene of the other microRNA, miR-182. We found that the high expression of miR-182 and miR-381 in gliomas are involved in pathological malignant progression. The silencing of miR-182 and miR-381 inhibited the proliferation in vitro and growth of glioma cell with in vivo magnetic resonance imaging by intracranial transplanted tumor model in rats. We also demonstrated that BRD7, a transcriptional cofactor for p53, is highly expressed and negatively correlated with LRRC4 expression in gliomas. Disturbing miR-182 and miR-381 affected transcriptional regulation of the BRD7 gene. This finding was verified by ectopic overexpression of LRRC4 or restoration of endogenous LRRC4 expression by treatment with the DNA demethylating agent 5-Aza-dC. Taken together, miR-182 and miR-381 may be a useful therapeutic target for treatment of glioma.


Oncology Reports | 2014

miR-128 and miR-149 enhance the chemosensitivity of temozolomide by Rap1B-mediated cytoskeletal remodeling in glioblastoma.

Xiaoling She; Zhibin Yu; Yulong Cui; Qianqian Lei; Zeyou Wang; Gang Xu; Juanjuan Xiang; Minghua Wu; Guiyuan Li

Glioblastoma multiforme (GBM) is one of the most deadly diseases affecting humans, and is often characterized by poor survival and by high resistance to chemotherapy and radiotherapy. Temozolomide (TMZ) is an oral alkylating agent which is widely used in the treatment of GBM following surgery. Although TMZ may restrain GBM growth, TMZ resistance is also common and accounts for numerous cases of treatment failure. Studies indicate that aberrant miRNA expression is associated with hallmark malignant properties of GBM. Thus, miRNA-based anticancer therapeutic approaches have been exploited, either alone or in combination with standard targeted therapies to enhance the efficacy of chemotherapy agents. In the present study, we demonstrated that the expression of miR-128 and miR-149 was downregulated in glioblastoma, and their overexpression inhibited the invasion of glioblastoma cells by targeting Rap1B-mediated cytoskeletal and related molecular alterations. Moreover, miR-128 and miR-149 enhanced the chemosensitivity of glioblastoma cells to TMZ.


Brain Research | 2011

POTEH hypomethylation, a new epigenetic biomarker for glioma prognosis.

Xiaoping Liu; Hailin Tang; Zuping Zhang; Wenjuan Li; Zeyou Wang; Ying Zheng; Minghua Wu; Guiyuan Li

POTE ankyrin domain family, member H (POTEH) belongs to POTE family, which expresses in many cancers. In this study, methylation status of POTEH promoter and its correlation with clinicopathological parameters were evaluated in glioma tissues and cells. Bisulfite sequencing PCR was carried out to investigate the promoter methylation status of POTEH in tumor of 96 glioma patients and glioma cells U251, SF767, and SF126. The effect of promoter hypomethylation on protein expression was evaluated by immunohistochemistry. POTEH was hypomethylated in 81.3% gliomas and none in normal brain tissues, and correlated significantly with its protein expression. But there was no remarkable relationship between sex, age, advanced tumor grade and POTEH hypomethylation. With the grade progressing, POTEH protein expression was enhanced. The correlation between POTEH hypomethylation, protein expression and overall survival was statistically significant. In POTEH hypomethylation group, patients with POTEH high expression had shorter overall survival than those with low expression. Hypomethylation of POTEH promoter in gliomas accounted for POTEH protein overexpression and poor outcome in a subset of patients. Detection of these epigenetic changes in tumors may provide information regarding prognosis.


Omics A Journal of Integrative Biology | 2014

miR-429 Identified by Dynamic Transcriptome Analysis Is a New Candidate Biomarker for Colorectal Cancer Prognosis

Yingnan Sun; Shourong Shen; Hailin Tang; Juanjuan Xiang; Ya Peng; Anliu Tang; Nan Li; Weiwei Zhou; Zeyou Wang; Decai Zhang; Bo Xiang; Jie Ge; Guiyuan Li; Minghua Wu; Li X

Colorectal cancer (CRC) is a common malignant gastrointestinal cancer. Efforts for preventive and personalized medicine have intensified in the last decade with attention to novel forms of biomarkers. In the present study, microRNA and genetic analyses were performed in tandem for differential transcriptome profiling between primary tumors with or without nodes or distant metastases. Serial Test Cluster (STC) analysis demonstrated that 20 genes and two microRNAs showed distinctive expression patterns associated with the tumor, node, and metastasis (TNM) stage. The selected target genes were characterized by GO and Pathway analysis. A microRNA-target gene network analysis showed that miR-429 resided in the center of the network, indicating that miR-429 might serve important roles in the development of CRC. Real-time PCR and tissue microarrays showed that miR-429 had a dynamic expression pattern during the CRC progression stage, and was significantly downregulated in stage II and stage III clinical progression. The low expression of miR-429 was correlated with poor prognosis for CRC. Taken together, miR-429 warrant further clinical translation research as a candidate biomarker for CRC prognosis. Additional downstream targets and attendant gene function also need to be discerned to design a sound critical path to personalized medicine for persons susceptible to, or diagnosed with CRC.


Medical Oncology | 2014

miR-181 subunits enhance the chemosensitivity of temozolomide by Rap1B-mediated cytoskeleton remodeling in glioblastoma cells

Xiaoling She; Zhibin Yu; Yulong Cui; Qianqian Lei; Zeyou Wang; Gang Xu; Zhaohui Luo; Guiyuan Li; Minghua Wu

Glioblastoma multiforme (GBM) is the most malignant and frequent brain tumor, with an aggressive growth pattern and poor prognosis despite best treatment modalities. Although chemotherapy with temozolomide (TMZ) may restrain tumor growth for some months, TMZ resistance is also common and accounts for many treatment failures. Research into microRNA’s role in GBM has shown that microRNAs play a key regulatory role in the GBM, making it a potential therapeutic target. In this study, we demonstrated that the lower expression of miR-181a/b/c/d subunits contributes to astrocytoma tumorigenesis, and their overexpression could inhibit the invasive proliferation of glioblastoma cells by targeting Rap1B-mediated cytoskeleton remodeling and related molecular (Cdc42, RhoA and N-cadherin) changes, suggesting that miR-181 was a critical regulator and might be an important target for glioblastoma treatment. TMZ as a standard chemotherapeutic agent for GBM inhibited the Rap1B expression and actin cytoskeleton remodeling to exert its cell killing by upregulating miR-181a/b/c/d subunits; conversely, each miR-181a/b/c/d subunit enhanced the chemosensitivity of TMZ in glioblastoma cells.


Journal of Cellular Biochemistry | 2011

LRRC4 inhibits the proliferation of human glioma cells by modulating the expression of STMN1 and microtubule polymerization

Rong Wang; Zeyou Wang; Jing Yang; Xiaoping Liu; Li Wang; Xiaofang Guo; Fang Zeng; Minghua Wu; Guiyuan Li

LRRC4 is a tumor suppressor of glioma, and it is epigenetically inactivated commonly in glioma. Our previous study has shown that induction of LRRC4 expression inhibits the proliferation of glioma cells. However, little is known about the mechanisms underlying the action of LRRC4 in glioma cells. We employed two‐dimensional fluorescence differential gel electrophoresis (2‐D DIGE) and MALDI –TOF/TOF‐MS/MS to identify 11 differentially expressed proteins, including the significantly down‐regulated STMN1 expression in the LRRC4‐expressing U251 glioma cells. The levels of STMN1 expression appeared to be positively associated with the pathogenic degrees of human glioma. Furthermore, induction of LRRC4 over‐expression inhibited the STMN1 expression and U251 cell proliferation in vitro, and the glioma growth in vivo. In addition, induction of LRRC4 or knockdown of STMN1 expression induced cell cycle arrest in U251 cells, which was associated with modulating the p21, cyclin D1, and cyclin B expression, and the ERK phosphorylation, and inhibiting the CDK5 and cdc2 kinase activities, but increasing the microtubulin polymerization in U251 cells. LRRC4, at least partially by down‐regulating the STMN1expression, acts as a major glioma suppressor, induces cell cycle arrest and modulates the dynamic process of microtubulin, leading to the inhibition of glioma cell proliferation and growth. Potentially, modulation of LRRC4 or STMN1 expression may be useful for design of new therapies for the intervention of glioma. J. Cell. Biochem. 112: 3621–3629, 2011.


Journal of Hematology & Oncology | 2016

The D Domain of LRRC4 anchors ERK1/2 in the cytoplasm and competitively inhibits MEK/ERK activation in glioma cells

Zeyou Wang; Qin Guo; Rong Wang; Gang Xu; Peiyao Li; Yingnan Sun; Xiaoling She; Qiang Liu; Qiong Chen; Zhibin Yu; Changhong Liu; Jing Xiong; Guiyuan Li; Minghua Wu

BackgroundAs a well-characterized key player in various signal transduction networks, extracellular-signal-regulated kinase (ERK1/2) has been widely implicated in the development of many malignancies. We previously found that Leucine-rich repeat containing 4 (LRRC4) was a tumor suppressor and a negative regulator of the ERK/MAPK pathway in glioma tumorigenesis. However, the precise molecular role of LRRC4 in ERK signal transmission is unclear.MethodsThe interaction between LRRC4 and ERK1/2 was assessed by co-immunoprecipitation and GST pull-down assays in vivo and in vitro. We also investigated the interaction of LRRC4 and ERK1/2 and the role of the D domain in ERK activation in glioma cells.ResultsHere, we showed that LRRC4 and ERK1/2 interact via the D domain and CD domain, respectively. Following EGF stimuli, the D domain of LRRC4 anchors ERK1/2 in the cytoplasm and abrogates ERK1/2 activation and nuclear translocation. In glioblastoma cells, ectopic LRRC4 expression competitively inhibited the interaction of endogenous mitogen-activated protein kinase (MEK) and ERK1/2. Mutation of the D domain decreased the LRRC4-mediated inhibition of MAPK signaling and its anti-proliferation and anti-invasion roles.ConclusionsOur results demonstrated that the D domain of LRRC4 anchors ERK1/2 in the cytoplasm and competitively inhibits MEK/ERK activation in glioma cells. These findings identify a new mechanism underlying glioblastoma progression and suggest a novel therapeutic strategy by restoring the activity of LRRC4 to decrease MAPK cascade activation.

Collaboration


Dive into the Zeyou Wang's collaboration.

Top Co-Authors

Avatar

Minghua Wu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Guiyuan Li

Central South University

View shared research outputs
Top Co-Authors

Avatar

Hailin Tang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Zhibin Yu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Gang Xu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Xiaoping Liu

Central South University

View shared research outputs
Top Co-Authors

Avatar

Rong Wang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Xiaoling She

Central South University

View shared research outputs
Top Co-Authors

Avatar

Qianqian Lei

Central South University

View shared research outputs
Top Co-Authors

Avatar

Changhong Liu

Central South University

View shared research outputs
Researchain Logo
Decentralizing Knowledge