Network


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

Hotspot


Dive into the research topics where Wenjun Hu is active.

Publication


Featured researches published by Wenjun Hu.


Journal of Experimental & Clinical Cancer Research | 2017

Hepatitis C virus NS3 protein enhances hepatocellular carcinoma cell invasion by promoting PPM1A ubiquitination and degradation

Yali Zhou; Yan Zhao; Yaoying Gao; Wenjun Hu; Yan Qu; Ning Lou; Ying Zhu; Xiaoping Zhang; Hongmei Yang

BackgroundGrowing evidence suggests that hepatitis C virus (HCV) contributes to hepatocellular carcinoma (HCC) by directly modulating oncogenic signaling pathways. Protein phosphatase magnesium-dependent 1A (PPM1A) has recently emerged as an important tumor suppressor as it can block a range of tumor-centric signaling pathways through protein dephosphorylation. However, the role and regulatory mechanisms of PPM1A in HCV-infected cells have not been reported.MethodsTotal, cytoplasmic, and nuclear PPM1A protein after HCV infection or overexpression of HCV nonstructural protein 3 (NS3) were detected by western blotting. The expression of PPM1A in normal liver and HCV-related HCC tissues was quantified by immunohistochemistry. The effects of HCV infection and NS3 expression on the PPM1A protein level were systematically analyzed, and the ubiquitination level of PPM1A was determined by precipitation with anti-PPM1A and immunoblotting with either anti-ubiquitin or anti-PPM1A antibody. Finally, the roles of NS3 and PPM1A in hepatoma cell migration and invasion were assessed by wound healing and transwell assays, respectively.ResultsHCV infection and replication decreased PPM1A abundance, mediated by NS3, in hepatoma cells. Compared to normal liver tissues, the expression of PPM1A was significantly decreased in the HCC tumor tissues and adjacent non-tumor tissues. NS3 directly interacted with PPM1A to promote PPM1A ubiquitination and degradation, which was dependent on its protease domain. Blockade of PPM1A through small interfering RNA significantly promoted HCC cell migration, invasion, and epithelial mesenchymal transition (EMT), which were further intensified by TGF-β1 stimulation, in vitro. Furthermore, restoration of PPM1A abrogated the NS3-mediated promotion of HCC migration and invasion to a great extent, which was dependent on its protein phosphatase function.ConclusionsOur findings demonstrate that the HCV protein NS3 can downregulate PPM1A by promoting its ubiquitination and proteasomal degradation, which might contribute to the migration and invasion of hepatoma cells and may represent a new strategy of HCV in carcinogenesis.


Cellular Physiology and Biochemistry | 2017

Mir-144-3p Promotes Cell Proliferation, Metastasis, Sunitinib Resistance in Clear Cell Renal Cell Carcinoma by Downregulating ARID1A

Wen Xiao; Ning Lou; Hailong Ruan; Lin Bao; Zhiyong Xiong; Changfei Yuan; Junwei Tong; Guanghua Xu; Yali Zhou; Yan Qu; Wenjun Hu; Yaoying Gao; Zeyuan Ru; Lei Liu; Haibing Xiao; Ke Chen; Hongmei Yang; Xiaoping Zhang

Background/Aims: We previously performed microRNA (miRNA) microarray to identify effective indicators of clear cell renal cell carcinoma (ccRCC) tissue samples and preoperative/postoperative plasma in which we identified miR-144-3p as an oncomiRNA. However, the molecular mechanism of miR-144-3p remains unclear. This study aims to explore the roles of miR-144-3p in the invasion, migration and Sunitinib-resistance in ccRCC and to elucidate the underlying mechanisms. Methods: Gain and loss of function approaches were used to investigate the cell proliferation, cycle distribution, clonogenicity, migration, invasion, chemosensitivity of miR-144-3p in vitro. The xenograft model was used to assess the effects of miR-144-3p overexpression on tumorigenesis. Bioinformatics analysis and dual-luciferase reporter assay were used to indentify AT-rich interactive domain 1A (ARID1A) as a direct target gene of miR-144-3p. Quantitative RT-PCR, Western blotting, and immunohistochemical (IHC) staining were used to explore ARID1A expression level of the mRNA and protein. Results: We found that miR-144-3p overexpression enhanced cell proliferation, clonogenicity, migration, invasion, and chemoresistance in ccRCC cells. Notably, the oncotumor activities of miR-144-3p were mediated by repressing the expression of ARID1A. The downregulation of ARIDIA could promote the function of miR-144-3p in cell proliferation, metastasis and chemoresistance. Consistently, ARID1A mRNA and protein levels were decreased in ccRCC and in nude mice, and they negatively correlated with miR-144-3p. Conclusion: Higher miR-144-3p may enhance malignancy and resistance to Sunitinib in ccRCC by targeting ARID1A, the observations may uncover novel strategies of ccRCC treatment.


American Journal of Physiology-cell Physiology | 2016

Valproic acid attenuates skeletal muscle wasting by inhibiting C/EBPβ-regulated atrogin1 expression in cancer cachexia

Rulin Sun; Santao Zhang; Wenjun Hu; Xing Lu; Ning Lou; Zhende Yang; Shaoyong Chen; Xiaoping Zhang; Hongmei Yang

Muscle wasting is the hallmark of cancer cachexia and is associated with poor quality of life and increased mortality. Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, has important biological effects in the treatment of muscular dystrophy. To verify whether VPA could ameliorate muscle wasting induced by cancer cachexia, we explored the role of VPA in two cancer cachectic mouse models [induced by colon-26 (C26) adenocarcinoma or Lewis lung carcinoma (LLC)] and atrophied C2C12 myotubes [induced by C26 cell conditioned medium (CCM) or LLC cell conditioned medium (LCM)]. Our data demonstrated that treatment with VPA increased the mass and cross-sectional area of skeletal muscles in tumor-bearing mice. Furthermore, treatment with VPA also increased the diameter of myotubes cultured in conditioned medium. The skeletal muscles in cachectic mice or atrophied myotubes treated with VPA exhibited reduced levels of CCAAT/enhancer binding protein beta (C/EBPβ), resulting in atrogin1 downregulation and the eventual alleviation of muscle wasting and myotube atrophy. Moreover, atrogin1 promoter activity in myotubes was stimulated by CCM via activating the C/EBPβ-responsive cis-element and subsequently inhibited by VPA. In contrast to the effect of VPA on the levels of C/EBPβ, the levels of inactivating forkhead box O3 (FoxO3a) were unaffected. In summary, VPA attenuated muscle wasting and myotube atrophy and reduced C/EBPβ binding to atrogin1 promoter locus in the myotubes. Our discoveries indicate that HDAC inhibition by VPA might be a promising new approach for the preservation of skeletal muscle in cancer cachexia.


Experimental Cell Research | 2017

Enhanced expression of caveolin-1 possesses diagnostic and prognostic value and promotes cell migration, invasion and sunitinib resistance in the clear cell renal cell carcinoma

Hailong Ruan; Xiang Li; Hongmei Yang; Zhengshuai Song; Junwei Tong; Qi Cao; Keshan Wang; Wen Xiao; HaiBin Xiao; XuanYu Chen; Guanghua Xu; Lin Bao; Zhiyong Xiong; Changfei Yuan; Lei Liu; Yan Qu; Wenjun Hu; Yaoying Gao; Zeyuan Ru; Ke Chen; Xiaoping Zhang

ABSTRACT Caveolin‐1 (CAV1) has been identified to be up‐regulated in many cancers, including clear cell renal cell carcinoma (ccRCC). However, its potential function is still unclear in ccRCC. In this study, we demonstrated that CAV1 was frequently overexpressed in renal cell carcinoma tissues and cells, and was significantly associated with various clinicopathological parameters. In addition, high CAV1 expression was associated with poor disease‐free survival (DFS) rate and could serve as a useful diagnostic indicator in ccRCC patients with different clinicopathological stages. Functional experiments demonstrated that CAV1 knockdown inhibited cell migration and invasion, whereas overexpression of CAV1 promoted cell migration and invasion in ccRCC. Moreover, CAV1 expression was up‐regulated in sunitinib‐resistant renal cancer cell lines, and its overexpression promoted sunitinib resistance. In general, our results confirm that CAV1 plays an important role in the metastasis of kidney cancer and induces sunitinib resistance, so CAV1 function suppression may become a promising clinical treatment strategy during renal cell carcinoma metastasis and sunitinib resistance. HIGHLIGHTSCAV1 serves as a potential diagnostic and prognostic molecular marker in ccRCC.CAV1 expression is elevated in the parental and sunitinib‐resistance ccRCC cells.CAV1 overexpression promotes sunitinib resistance in ccRCC.CAV1 promotes migration and invasion in ccRCC cells.


International Journal of Oncology | 2017

Overexpression of SOX4 promotes cell migration and invasion of renal cell carcinoma by inducing epithelial-mesenchymal transition

Hailong Ruan; Hongmei Yang; Haibin Wei; Wen Xiao; Ning Lou; Bin Qiu; Guanghua Xu; Zhengshuai Song; HaiBin Xiao; Lei Liu; Yali Zhou; Wenjun Hu; Ke Chen; X. Chen; Xiaoping Zhang

Incomplete understanding remains in the molecular mechanisms underlying progression and metastasis of renal cancer. The transcription factor SOX4 is upregulated in various human malignancies, including renal cancer, indicating it may be involved in renal tumorigenesis. In this study, we explored this hypothesis by loss-of-function and gain-of-function assays of SOX4 in renal cancer cell lines and renal epithelial cell line. We found that specific knockdown of SOX4 in renal cancer cell lines significantly suppressed the migration and invasion of cancer cells; specific overexpression of SOX4 in renal epithelial cell line markedly promoted the migration and invasion of the cell line. Epithelial-mesenchymal transition (EMT), a fundamental morphogenesis process, is implicated in renal cancer progression and metastasis. Our results demonstrated that SOX4 positively regulated the expression of mesenchymal cell markers and negatively regulated the expression of epithelial cell marker, and was involved in signal transduction pathway of TGFβ-induced EMT. In addition, SOX4 induced EMT probably through modulating the AKT/p-AKT signaling cascade. Finally, we found that SOX4 was significantly upregulated in clinical renal cancer samples compared with corresponding normal tissues and associated with EMT process in clinical samples. Taken together, our findings confirm a crucial function of SOX4 in the metastasis of renal cancer through orchestrating EMT and establish that the function suppression of SOX4-AKT-EMT axis might be an attractive therapeutic intervention during renal cancer metastasis.


BMC Cancer | 2018

Up-regulation of SR-BI promotes progression and serves as a prognostic biomarker in clear cell renal cell carcinoma

Guanghua Xu; Ning Lou; Hangchuan Shi; Yuchen Xu; Hailong Ruan; Wen Xiao; Lei Liu; Xiang Li; Haibing Xiao; Bin Qiu; Lin Bao; Changfei Yuan; Yali Zhou; Wenjun Hu; Ke Chen; Hongmei Yang; Xiaoping Zhang

BackgroundScavenger receptor class B type I (SR-BI) has been reported to be involved in carcinogenesis of several human cancers. However, it is currently unknown whether SR-BI plays a role in clear cell renal cell carcinoma (ccRCC). Here, we aimed to evaluate a tumor promotive mechanism for SR-BI in ccRCC.MethodsThe expression of SR-BI was evaluated by real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blot and immunohistochemistry (IHC) in ccRCC tissues and cell lines. Lipid droplets in ccRCC tissues and normal kidney tissues were examined by Oil Red O (ORO) and hematoxylin-eosin (HE) staining. The correlation between SR-BI mRNA levels and clinicopathological features was analyzed by Pearson’s chi-square test or Fisher’s exact test. Kaplan-Meier analysis and Cox model were used to evaluate the difference in progression-free survival (PFS) associated with expression of SR-BI. Inhibition of SR-BI was conducted by using small interfering RNA (siRNA). In vitro assays were performed to assess the impact of SR-BI knockdown on cell biological behaviors. High density lipoprotein (HDL)-cholesterol content in ccRCC cells and extracellular media was also measured after transfection with siRNA.ResultsThe expression of SR-BI was markedly up-regulated in ccRCC tissues and tumor cell lines. ORO and HE staining revealed huge amounts of lipid droplets accumulation in ccRCC. Clinical analysis showed that over-expression of SR-BI was positively associated with tumor size, grade, distant metastasis and inversely correlated with PFS. Furthermore, SR-BI was proved to be an independent prognostic marker in ccRCC patients. The inhibition of SR-BI attenuated the tumorous behaviors of ccRCC cells, expression of metastasis and AKT pathway related proteins. The content of HDL-cholesterol was reduced in cells while increased in extracellular media after transfection with si-SR-BI.ConclusionsOur results demonstrate that SR-BI functions as an oncogene and promotes progression of ccRCC. SR-BI may serve as a potential prognostic biomarker and therapeutic target for ccRCC.


Oncology Reports | 2016

Comparative molecular analysis of early and late cancer cachexia-induced muscle wasting in mouse models.

Rulin Sun; Santao Zhang; Xing Lu; Wenjun Hu; Ning Lou; Yan Zhao; Jia Zhou; Xiaoping Zhang; Hongmei Yang

Cancer-induced muscle wasting, which commonly occurs in cancer cachexia, is characterized by impaired quality of life and poor patient survival. To identify an appropriate treatment, research on the mechanism underlying muscle wasting is essential. Thus far, studies on muscle wasting using cancer cachectic models have generally focused on early cancer cachexia (ECC), before severe body weight loss occurs. In the present study, we established models of ECC and late cancer cachexia (LCC) and compared different stages of cancer cachexia using two cancer cachectic mouse models induced by colon-26 (C26) adenocarcinoma or Lewis lung carcinoma (LLC). In each model, tumor-bearing (TB) and control (CN) mice were injected with cancer cells and PBS, respectively. The TB and CN mice, which were euthanized on the 24th day or the 36th day after injection, were defined as the ECC and ECC-CN mice or the LCC and LCC-CN mice. In addition, the tissues were harvested and analyzed. We found that both the ECC and LCC mice developed cancer cachexia. The amounts of muscle loss differed between the ECC and LCC mice. Moreover, the expression of some molecules was altered in the muscles from the LCC mice but not in those from the ECC mice compared with their CN mice. In conclusion, the molecules with altered expression in the muscles from the ECC and LCC mice were not exactly the same. These findings may provide some clues for therapy which could prevent the muscle wasting in cancer cachexia from progression to the late stage.


Chinese journal of cancer | 2007

Expression of Sonic Hedgehog, EGFR and PCNA Proteins in Pancreatic Cancer and Their Correlations to Cell Proliferation

Wenjun Hu; Wang Cy; Liu T; Xiong Jx; Yang Zy


Cellular Physiology and Biochemistry | 2018

Upregulation of MIAT Regulates LOXL2 Expression by Competitively Binding MiR-29c in Clear Cell Renal Cell Carcinoma

Yan Qu; Haibing Xiao; Wen Xiao; Zhiyong Xiong; Wenjun Hu; Yaoying Gao; Zeyuan Ru; Cheng Wang; Lin Bao; Kesan Wang; Hailong Ruan; Zhengshuai Song; Ke Chen; Xiaoping Zhang; Hongmei Yang


Experimental Cell Research | 2018

Corrigendum to “Enhanced expression of caveolin-1 possesses diagnostic and prognostic value and promotes cell migration, invasion and sunitinib resistance in the clear cell renal cell carcinoma” [Exp. Cell Res. (2017) 269–278]

Hailong Ruan; Xiang Li; Hongmei Yang; Zhengshuai Song; Junwei Tong; Qi Cao; Keshan Wang; Wen Xiao; HaiBin Xiao; X. Chen; Guanghua Xu; Lin Bao; Zhiyong Xiong; Changfei Yuan; Lei Liu; Yan Qu; Wenjun Hu; Yaoying Gao; Zeyuan Ru; Ke Chen; Xiaoping Zhang

Collaboration


Dive into the Wenjun Hu's collaboration.

Top Co-Authors

Avatar

Hongmei Yang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Xiaoping Zhang

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Wen Xiao

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Hailong Ruan

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Ke Chen

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Ning Lou

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Changfei Yuan

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Guanghua Xu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Lei Liu

Huazhong University of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Lin Bao

Huazhong University of Science and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge