Network


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

Hotspot


Dive into the research topics where Wensheng Sun is active.

Publication


Featured researches published by Wensheng Sun.


Journal of Hepatology | 2010

T cell immunoglobulin- and mucin-domain-containing molecule-3 (Tim-3) mediates natural killer cell suppression in chronic hepatitis B

Ying Ju; Nan Hou; Jing Meng; Xiaoyan Wang; Xiaoning Zhang; Di Zhao; Ying Liu; Faliang Zhu; Lining Zhang; Wensheng Sun; Xiaohong Liang; Lifen Gao; Chunhong Ma

BACKGROUND & AIMS T cell immunoglobulin- and mucin-domain-containing molecule-3 (Tim-3) has been shown to influence autoimmune diseases; however, its function in viral infection has not been well-defined. We therefore investigated the expression and regulatory function of Tim-3 in natural killer (NK) cells in chronic Hepatitis B (CHB) infection. METHODS Seventy-six CHB patients, 38 healthy controls, and 18 patients with fatty liver disease (FLD) were tested for Tim-3 expression on peripheral blood mononuclear cells (PBMCs) and in the liver tissue by flow cytometry and immunohistochemical stainning. The effects of HBV infection on Tim-3 expression in NK cells and the roles of Tim-3 in regulation of NK-cell function were also studied. RESULTS There was a significant increase of Tim-3 expression in PBMCs, circulating NK cells and liver infiltrating lymphocytes (LILs) from CHB patients compared to that of healthy controls and FLD patients. Increased Tim-3 expression was also detected in NK92 cells that had been transfected with a HBV expression vector and NK cells isolated from the liver of HBV transgenic mice. Importantly, blockage of Tim-3 signaling with anti-Tim-3 antibodies or Tim-3-Fc fusion proteins resulted in an increased cytotoxicity for NK92 cells compared to HepG2 and HepG2.2.15 cells, as well as an elevated interferon-gamma (IFN-gamma) production. Similarly, enhanced cytotoxicity was also observed in PBMCs or NK cells from CHB patients treated with the Tim-3 blockade ex vivo. CONCLUSION HBV infection can up-regulate Tim-3 expression in NK cells, which may in turn suppress NK-cell functions in CHB patients.


Biochemical and Biophysical Research Communications | 2011

miR-29c targets TNFAIP3, inhibits cell proliferation and induces apoptosis in hepatitis B virus-related hepatocellular carcinoma.

Chunmei Wang; Yan Wang; Chunguang Fan; Fei-Fei Xu; Wensheng Sun; Yugang Liu; Ji-Hui Jia

Recent studies have revealed that microRNA-29c (miR-29c) is involved in a variety of biological processes including carcinogenesis. Here, we report that miR-29c was significantly downregulated in hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) cell lines as well as in clinical tissues compared with their corresponding controls. Tumor necrosis factor alpha-induced protein 3 (TNFAIP3), a key regulator in inflammation and immunity, was found to be inversely correlated with miR-29c levels and was identified as a target of miR-29c. Overexpression of miR-29c in HepG2.2.15 cells effectively suppressed TNFAIP3 expression and HBV DNA replication as well as inhibited cell proliferation and induced apoptosis. We conclude that miR-29c may play an important role as a tumor suppressive microRNA in the development and progression of HBV-related HCC by targeting TNFAIP3. Thus miR-29c and TNFAIP3 represent key diagnostic markers and potential therapeutic targets for the prevention and treatment of HBV infection.


Journal of Immunology | 2007

Hepatitis B Virus Sensitizes Hepatocytes to TRAIL-Induced Apoptosis through Bax

Xiaohong Liang; Yugang Liu; Qiu Zhang; Lifen Gao; Lihui Han; Chunhong Ma; Lining Zhang; Youhai H. Chen; Wensheng Sun

Hepatitis B virus (HBV) infection afflicts >300 million people worldwide and is a leading cause of hepatocyte death, cirrhosis, and hepatocellular carcinoma. While the morphological characteristics of dying hepatocytes are well documented, the molecular mechanisms leading to the death of hepatocytes during HBV infection are not well understood. TRAIL, the TNF-related apoptosis-inducing ligand, has recently been implicated in the death of hepatocytes under certain inflammatory but not normal conditions. To determine the potential roles of TRAIL in HBV-induced hepatitis, we examined the effects of HBV and its X protein (HBx) on TRAIL-induced hepatocyte apoptosis both in vivo and in vitro. We found that hepatitis and hepatic cell death in HBV transgenic mice were significantly inhibited by a soluble TRAIL receptor that blocks TRAIL function. We also found that HBV or HBx transfection of a hepatoma cell line significantly increased its sensitivity to TRAIL-induced apoptosis. The increase in TRAIL sensitivity were associated with a dramatic up-regulation of Bax protein expression. Knocking down Bax expression using Bax-specific small interference RNA blocked HBV-induced hepatitis and hepatocyte apoptosis. The degradation of caspases 3 and 9, but not that of Bid or caspase-8, was preferentially affected by Bax knockdown. These results establish that HBV sensitizes hepatocytes to TRAIL-induced apoptosis through Bax and that Bax-specific small interference RNA can be used to inhibit HBV-induced hepatic cell death.


Oncology Reports | 2011

miR-122 inhibits viral replication and cell proliferation in hepatitis B virus-related hepatocellular carcinoma and targets NDRG3.

Chunguang Fan; Chunmei Wang; Chuan Tian; Yan Wang; Li Li; Wensheng Sun; Rui-Feng Li; Yu-Gang Liu

microRNAs (miRNAs) are short, non-coding RNAs with post-transcriptional regulatory functions that participate in diverse biological pathways. miR-122, a liver-specific miRNA, has been found to be down-regulated in hepatocellular carcinoma (HCC) and HCC-derived cell lines. In this study, miR-122 was down-regulated in the hepatitis B virus (HBV)-related HCC cell line HepG2.2.15 compared to HepG2. NDRG3, a member of the N-myc downstream-regulated gene (NDRG) family, was up-regulated in HepG2.2.15 and was identified as a target gene of miR-122. An inverse correlation between the expression of miR-122 and the NDRG3 protein was noted in HBV-related HCC specimens. The transfection of the miR-122 expression vector into the HepG2.2.15 cell line repressed the transcription and expression of NDRG3, which subsequently reversed the malignant phenotype of the cells. The replication of HBV, expression of viral antigens and proliferation of cells were significantly inhibited by restoration of miR-122. The data demonstrate that miR-122 plays an important role in HBV-related hepatocarcinogenesis by targeting NDRG3. Thus, miR-122 and NDRG3 represent key diagnostic markers and potential therapeutic targets for HBV-related HCC.


Cell Death & Differentiation | 2009

Hepatitis B virus core protein inhibits TRAIL-induced apoptosis of hepatocytes by blocking DR5 expression

Juan Du; Xiaohong Liang; Yugang Liu; Zhonghua Qu; Lifen Gao; Lihui Han; Shujing Liu; Min Cui; Yongyu Shi; Zhiyong Zhang; L Yu; L Cao; Chunhong Ma; Lining Zhang; Youhai H. Chen; Wensheng Sun

Hepatitis B virus (HBV) causes chronic hepatitis in hundreds of millions of people worldwide, which can eventually lead to hepatocellular carcinoma (HCC). The molecular mechanisms underlying HBV persistence are not well understood. TRAIL, the TNF-related apoptosis-inducing ligand, has recently been implicated in hepatocyte death during HBV infection. We report here that the HBV core protein (HBc) is a potent inhibitor of TRAIL-induced apoptosis. Overexpressing HBc significantly decreased TRAIL-induced apoptosis of human hepatoma cells, whereas knocking-down HBc expression in hepatoma cells transfected with HBV genome enhanced it. When present in the same cell, HBc blocked the pro-apoptotic effect of the HBV X protein (HBx). The resistance of HBc-expressing cells to TRAIL-induced apoptosis was associated with a significant reduction in death receptor 5 (DR5) expression. Upon transfection, HBc significantly repressed the promoter activity of the human DR5 gene. Importantly, HBc gene transfer inhibited hepatocyte death in a mouse model of HBV-induced hepatitis; and in patients with chronic hepatitis, DR5 expression in the liver was significantly reduced. These results indicate that HBc may prevent hepatocytes from TRAIL-induced apoptosis by blocking DR5 expression, which in turn contributes to the development of chronic hepatitis and HCC. They also call into question the potential side effects of HBc-based vaccines.


Scandinavian Journal of Immunology | 2007

Expression of Human TIM‐1 and TIM‐3 on Lymphocytes from Systemic Lupus Erythematosus Patients

Y. Wang; Jing Meng; Xiaoyan Wang; Suxia Liu; Q. Shu; Lifen Gao; Ying Ju; Lining Zhang; Wensheng Sun; Chunhong Ma

The T‐cell immunoglobulin‐ and mucin‐domain‐containing molecules (TIMs) comprise a new family of cell surface molecules expressed on T cells. TIM‐3 is expressed on T helper type 1 (Th1) cells and implicated in the pathogenesis of Th1‐driven auto‐ and allo‐immune diseases. TIM‐1 is suggested to act as a co‐stimulatory molecule for all T cells, but with potentially stronger effects on Th2 than Th1 cells and is associated with Th2‐related immune diseases. However, the TIM molecules have not been investigated in the systemic lupus erythematosus (SLE). In this study, we examined the expression of TIM‐1 and TIM‐3 on peripheral blood mononuclear cells from SLE patients using quantitative real‐time RT‐PCR. An increased TIM‐1 expression was detected in SLE patients, which correlates with interleukin‐10 expression. We also found that there was a significant increase in the expression of TIM‐1 in SLE patients with quite active disease (SLE disease activity index > 6), indicating that TIM‐1 expression might be related to active clinical phases. In contrast, TIM‐3 expression remained normal in SLE patients with low statistical power (34.89%). However, the expression of TIM‐3 ligand, galectin‐9 increased in SLE patients indicating an enhanced engagement of TIM‐3 with its ligand in SLE, which may result in a decreased regulatory T‐cell function as shown by the decreased expression of FoxP3 and TGF‐β1 in SLE. These data suggest that TIM‐1 and TIM‐3/TIM‐3L are involved in the pathogenesis of SLE.


Cellular & Molecular Immunology | 2009

Blockade of Tim-3 Pathway Ameliorates Interferon- γ Production from Hepatic CD8 + T Cells in a Mouse Model of Hepatitis B Virus Infection

Ying Ju; Nan Hou; Xiaoning Zhang; Di Zhao; Ying Liu; Jinjin Wang; Fang Luan; Wei Shi; Faliang Zhu; Wensheng Sun; Lining Zhang; Chengjiang Gao; Lifen Gao; Xiaohong Liang; Chunhong Ma

T cell immunoglobulin- and mucin-domain-containing molecule-3 (Tim-3) has been reported to participate in the pathogenesis of inflammatory diseases. However, whether Tim-3 is involved in hepatitis B virus (HBV) infection remains unknown. Here, we studied the expression and function of Tim-3 in a hydrodynamics-based mouse model of HBV infection. A significant increase of Tim-3 expression on hepatic T lymphocytes, especially on CD8+ T cells, was demonstrated in HBV model mice from day 7 to day 18. After Tim-3 knockdown by specific shRNAs, significantly increased IFN-γ production from hepatic CD8+ T cells in HBV model mice was observed. Very interestingly, we found Tim-3 expression on CD8+ T cells was higher in HBV model mice with higher serum anti-HBs production. Moreover, Tim-3 knockdown influenced anti-HBs production in vivo. Collectively, our data suggested that Tim-3 might act as a potent regulator of antiviral T-cell responses in HBV infection.


Journal of Cellular and Molecular Medicine | 2009

PDCD4 gene silencing in gliomas is associated with 5′CpG island methylation and unfavourable prognosis

Fei Gao; Xiaoyan Wang; Faliang Zhu; Qun Wang; Xia Zhang; Chun Guo; Chengjun Zhou; Chunhong Ma; Wensheng Sun; Yun Zhang; Youhai H. Chen; Lining Zhang

Programmed cell death 4 (PDCD4) is a newly described tumour suppressor that inhibits oncogenesis by suppressing gene transcription and translation. Loss of PDCD4 expression has been found in several types of human cancers including the most common cancer of the brain, the gliomas. However, the molecular mechanisms responsible for PDCD4 gene silencing in tumour cells remain unclear. Here we report the identification of 5′CpG island methylation as the predominant cause of PDCD4 mRNA silencing in gliomas. The methylation of the PDCD4 5′CpG island was found in 47% (14/30) of glioma tissues, which was significantly associated with the loss of PDCD4 mRNA expression (γ=−1.000, P < 0.0001). Blocking methylation in glioma cells using a DNA methyltransferase inhibitor, 5‐aza‐2′‐deoxycytidine, restored the PDCD4 gene expression, inhibited their proliferation and reduced their colony formation capacity. Longitudinal studies of a cohort of 84 patients with gliomas revealed that poor prognosis of patients with high‐grade tumours were significantly associated with loss of PDCD4 expression. Thus, our current study suggests, for the first time, that PDCD4 5′CpG island methylation blocks PDCD4 expression at mRNA levels in gliomas. These results also indicate that PDCD4 reactivation might be an effective new strategy for the treatment of gliomas.


Cancer Science | 2009

PDCD4 inhibits the malignant phenotype of ovarian cancer cells.

Zengtao Wei; Xia Zhang; Xiaoyan Wang; Fei Gao; Chengjun Zhou; Faliang Zhu; Qun Wang; Qi Gao; Chunhong Ma; Wensheng Sun; Qing-Zhao Fu; Youhai H. Chen; Lining Zhang

Programmed cell death 4 (PDCD4) is a newly identified tumor suppressor that can inhibit activator protein (AP)‐1 activation and protein translation. Our previous studies indicate that lost or reduced PDCD4 expression is associated with the progression of ovarian carcinoma. However, direct evidence that PDCD4 inhibits malignant phenotype of human cancer cells is limited. In the present study, we found that PDCD4 expression in ovarian cancer cell lines (SKOV3, 3AO, and CAOV3) inhibited significantly their proliferation and cell cycle progression, and induced apoptosis. More importantly, up‐regulation of PDCD4 expression decreased the colony‐forming capacity of ovarian cancer cells in vitro and tumorigenic capacity in mice. These results demonstrate that PDCD4 can suppress the malignant phenotype of ovarian cancer cells, and may represent a novel therapeutic target for the treatment of ovarian cancer. (Cancer Sci 2009)


Cancer Letters | 2008

Acquisition of anoikis resistance reveals a synoikis-like survival style in BEL7402 hepatoma cells.

Zhiyong Zhang; Lili Cao; Jie Li; Xiaohong Liang; Yugang Liu; Hua Liu; Juan Du; Zhonghua Qu; Min Cui; Suxia Liu; Lifen Gao; Chunhong Ma; Lining Zhang; Lihui Han; Wensheng Sun

Resistance to anoikis is a hallmark of human malignancies. Our results showed that hepatoma cells resisted anoikis by non-proliferation, non-apoptosis and cell cycle arrest which were termed synoikis-like. These synoikis-like cells are more resistant to extracellular stimuli and could spontaneously attach and proliferate again under suitable conditions, which indicate a reversible property of these cells. Microarray expression profile reveals the change of molecules involved in the synoikis-like hepatoma cells and our data indicated that ANGPTL4 contributed to anoikis resistance of hepatoma cells. These results demonstrated that hepatoma cells might resist anoikis through a synoikis-like survival style, which may facilitate tumor metastasis.

Collaboration


Dive into the Wensheng Sun's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge