Network


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

Hotspot


Dive into the research topics where Zhirong Wang is active.

Publication


Featured researches published by Zhirong Wang.


PLOS ONE | 2011

The Effect of hOGG1 Ser326Cys Polymorphism on Cancer Risk: Evidence from a Meta-Analysis

Bingbing Wei; You Zhou; Zhuoqun Xu; Bo Xi; Huan Cheng; Jun Ruan; Ming Zhu; Qiang Hu; Qiang Wang; Zhirong Wang; Zhiqiang Yan; Ke Jin; Deqi Zhou; Feng Xuan; Xing Huang; Jianfeng Shao; Peng Lu

Background Human oxoguanine glycosylase 1 (hOGG1) in base excision repair (BER) pathway plays a vital role in DNA repair. Numerous epidemiological studies have evaluated the association between hOGG1 Ser326Cys polymorphism and the risk of cancer. However, the results of these studies on the association remain conflicting. To derive a more precise estimation of the association, we conducted a meta-analysis. Methodology/Principal Findings A comprehensive search was conducted to identify the eligible studies of hOGG1 Ser326Cys polymorphism and cancer risk. We used odds ratios (ORs) with 95% confidence intervals (CIs) to assess the strength of the association. We found that the hOGG1 Ser326Cys polymorphism was significantly associated with overall cancer risk (Cys/Cys vs. Ser/Ser: OR = 1.19, 95%CI = 1.09–1.30, P<0.001; Cys/Cys vs. Cys/Ser+Ser/Ser: OR = 1.16, 95%CI = 1.08–1.26, P<0.001). Moreover, in subgroup analyses by cancer types, the stronger significant association between hOGG1 Ser326Cys polymorphism and lung cancer risk was found (Cys/Cys vs. Ser/Ser: OR = 1.29, 95%CI = 1.16–1.44, P<0.001; Cys/Cys vs. Cys/Ser+Ser/Ser: OR = 1.22, 95%CI = 1.12–1.33, P<0.001). The significant effects of hOGG1 Ser326Cys polymorphism on colorectal, breast, bladder, prostate, esophageal, and gastric cancer were not detected. In addition, in subgroup analyses by ethnicities, we found that the hOGG1 Ser326Cys polymorphism was associated with overall cancer risk in Asians (Cys/Cys vs. Ser/Ser: OR = 1.21, 95%CI = 1.10–1.33, P<0.001). Conclusions This meta-analysis showed that hOGG1 326Cys allele might be a low-penetrant risk factor for lung cancer.


PLOS ONE | 2012

Association of GSTM1 Null Allele with Prostate Cancer Risk: Evidence from 36 Case-Control Studies

Bingbing Wei; Zhuoqun Xu; You Zhou; Jun Ruan; Huan Cheng; Bo Xi; Ming Zhu; Ke Jin; Deqi Zhou; Qiang Hu; Qiang Wang; Zhirong Wang; Zhiqiang Yan; Feng Xuan; Xing Huang; Jian Zhang; Hongyi Zhou

Background Glutathione S-transferase M1 (GSTM1) is thought to be involved in detoxifying several carcinogens and may play a vital role in tumorigenesis. Numerous studies have evaluated the association between GSTM1 null/present polymorphism and risk of prostate cancer (PCa). However, the results remain inconsistent. To derive a more precise estimation, we performed a meta-analysis. Methodology/Principal Findings A comprehensive search was conducted to identify all eligible case-control studies. We used odds ratios (ORs) with 95% confidence intervals (CIs) to assess the strength of the association. The overall association was significant (OR = 1.28, 95% CI: 1.11–1.48, P = 0.001). Moreover, subgroup analyses showed GSTM1 null genotype significantly associated with PCa risk among Asians (OR = 1.35, 95% CI: 1.03–1.78, P = 0.03) but not among Caucasians (OR = 1.12, 95% CI: 0.96–1.31, P = 0.16). In addition, we did not find that smoking modified the genotype effect on the risk of PCa. Conclusions/Significance The present meta-analysis suggested that GSTM1 null allele was a low-penetrant risk factor for PCa among Asians.


PLOS ONE | 2013

GSTP1 Ile105Val Polymorphism and Prostate Cancer Risk: Evidence from a Meta-Analysis

Bingbing Wei; You Zhou; Zhuoqun Xu; Jun Ruan; Huan Cheng; Ming Zhu; Qiang Hu; Ke Jin; Zhiqiang Yan; Deqi Zhou; Feng Xuan; Hongyi Zhou; Zhirong Wang; Xing Huang; Qiang Wang

Background Glutathione S-transferase P1 (GSTP1) is thought to be involved in the detoxification of reactive carcinogen metabolites. Numerous epidemiological studies have evaluated the association of GSTP1 Ile105Val polymorphism with the risk of prostate cancer. However, the results remain inconclusive. To derive a more precise estimation, a meta-analysis was performed. Methodology/Principal Findings A comprehensive search was conducted to identify the eligible studies. We used odds ratios (ORs) with 95% confidence intervals (CIs) to assess the strength of the relationship. The overall association was not significant (Val/Val vs. Ile/Ile OR = 1.06, 95% CI = 0.90–1.25, P = 0.50; Val/Val vs. Val/Ile+Ile/Ile: OR = 1.07, 95% CI = 0.91–1.25, P = 0.44). In subgroup analyses by ethnicity and prostate cancer grade, the similar results were observed. However, in stratified analysis by clinical stage, we found a significant association with low-stage prostate cancer (Val/Val vs. Ile/Ile: OR = 2.70, 95% CI = 1.73–4.22, P<0.001; Val/Val vs. Val/Ile+Ile/Ile: OR = 2.14, 95% CI = 1.38–3.33, P = 0.001). Moreover, there was no statistically significant evidence of multiplicative interactions neither between the GSTP1 Ile105Val polymorphism and GSTM1, nor between smoking status and GSTP1 on prostate cancer risk. Conclusions This meta-analysis showed that GSTP1 Ile105Val polymorphism might not be significantly associated with overall prostate cancer risk. Further stratified analyses showed a significant association with low-stage prostate cancer.


Medical Science Monitor | 2017

CPNE1 Is a Useful Prognostic Marker and Is Associated with TNF Receptor-Associated Factor 2 (TRAF2) Expression in Prostate Cancer

Jiabei Liang; Jian Zhang; Jun Ruan; Yuanyuan Mi; Qiang Hu; Zhirong Wang; Bingbing Wei

Background CPNE1 plays a vital role in regulating cell differentiation. The clinical and biological values of CPNE1 in prostate cancer are still unclear. The aim of this study was to investigate the clinicopathological value of CPNE1 and the association of CPNE1 with TRAF2 expression in patients with prostate cancer. Material/Methods CPNE1 expression in prostate cancer was analyzed using Gene Expression Omnibus (GEO) databases. The Cancer Genome Atlas (TCGA) dataset was used to investigate the association of CPNE1 expression with TRAF2 expression in prostate cancer. The association of CPNE1 expression with recurrence-free survival in patients was also analyzed using the TCGA dataset. Immunohistochemistry assay was performed to examine CPNE1 expression in 65 normal prostate samples and 114 prostate cancer samples. The recurrence-free survival in patients was evaluated using Kaplan-Meier curves and log-rank test. In addition, multivariate and univariate analyses of prognostic factors were investigated by Cox regression. The effect of CPNE1 on TRAF2 expression was explored in human prostate cancer DU-145 cells. Results Our results showed that expression level of CPNE1 is higher in prostate cancer than in normal prostate tissues (P=0.006). In the GSE35988 dataset, CPNE1 expression was found to be upregulated in castration-resistant prostate cancer compared with non-castration-resistant prostate cancer (P<0.001). Furthermore, we found that CPNE1 high expression was significantly related to tumor stage, Gleason score, and poorer biochemical recurrence-free survival in prostate cancer patients. Co-expression analysis of TCGA data showed that CPNE1 is significantly associated with TRAF2 expression. CPNE1 overexpression can upregulate TRAF2 expression in prostate cancer DU-145 cells as determined by Western blotting and immunofluorescence assays. Conclusions Overall, our findings suggest that CPNE1 is a valuable prognostic marker for evaluating recurrence-free survival and is positively related to TRAF2 expression in prostate cancer.


Medical Science Monitor | 2017

TRAF2 is a Valuable Prognostic Biomarker in Patients with Prostate Cancer

Bingbing Wei; Jiabei Liang; Jimeng Hu; Yuanyuan Mi; Jun Ruan; Jian Zhang; Zhirong Wang; Qiang Hu; Haowen Jiang; Qiang Ding

Background TRAF2 exerts important functions in regulating the development and progression of cancer. The aim of this study is to investigate whether TRAF2 is a valuable prognostic biomarker and to determine if it regulates TRAIL-induced apoptosis in prostate cancer. Material/Methods Microarray gene expression data from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were used to determine TRAF2 expression in prostate cancer. TRAF2 expression in prostate cancer was further investigated by immunohistochemistry assay. Kaplan–Meier curves and log-rank test were used to assess the recurrence-free rate. Cox regression was used to analyze prognostic factors. Effects of TRAF2 on regulating TRAIL-induced apoptosis in DU-145 cells were further investigated. Results We found that TRAF2 was significantly upregulated in prostate cancer compared with normal prostate samples (P<0.001). In addition, compared with primary prostate cancer, TRAF2 was upregulated in metastatic prostate cancer (P=0.006). Furthermore, our results showed that high expression of TRAF2 was significantly associated with tumor stage of prostate cancer (P=0.035). TRAF2 high expression was associated with poorer recurrence-free survival in prostate cancer patients (P=0.013). TRAF2 was found to be a valuable independent prognostic factor for predicting recurrence-free survival (P=0.026). In addition, the present results indicate that TRAF2 affects TRAIL-induced apoptosis in prostate cancer DU-145 cells via regulating cleaved Caspase-8 and c-Flip expression. Conclusions TRAF2 could be a novel prognostic biomarker for predicting recurrence-free survival in patients with prostate cancer, which might be associated with the effects of TRAF2 in regulating TRAIL-induced apoptosis in prostate cancer cells via c-Flip/Caspase-8 signalling.


Biomedicine & Pharmacotherapy | 2017

Knockdown of TNF receptor-associated factor 2 (TRAF2) modulates in vitro growth of TRAIL-treated prostate cancer cells

Bingbing Wei; Jun Ruan; Yuanyuan Mi; Jimeng Hu; Jian Zhang; Zhirong Wang; Qiang Hu; Haowen Jiang; Qiang Ding

TNF receptor-associated factor 2 (TRAF2) is documented to regulate tumor development and progression. Currently, the effect of TRAF2 on growth of androgen-refractory prostate cancer in response to TRAIL and the molecular mechanisms are not well understood. Here, we aim to investigate the effect of TRAF2 on in vitro growth of human androgen-insensitive prostate cancer DU-145 cells in the presence of TRAIL. Bioinformatics analysis of the Cancer Genome Atlas (TCGA) data was performed to examine TRAF2 expression and the prognostic value in prostate cancer. Microarray data of GSE21032 dataset were downloaded from Gene Expression Omnibus (GEO) to explore TRAF2 expression in metastatic prostate cancer. Bioinformatics analysis was further conducted to investigate the association of TRAF2 expression with recurrence-free survival in prostate cancer patients. Colony formation, cell viability, and Annexin V/PI apoptosis assays were performed to investigate the effect of TRAF2 on in vitro growth and apoptosis in TRAIL-treated DU-145 cells. The expression levels of mRNA and protein were detected by quantitative RT-PCR and immunoblotting assays. Bioinformatics analysis indicated that TRAF2 expression is significantly upregulated in prostate cancer patients with high Gleason scores (GS>7) compared with those with low Gleason scores (GS≤7). Upregulation of TRAF2 expression is significantly associated with recurrence-free survival in patients. In addition, TRAF2 knockdown can enhance apoptosis and downregulate SIRT1 expression in TRAIL-treated DU-145 cells. In vitro experiments further showed that SIRT1 knockdown can inhibit growth, and promote apoptosis in TRAIL-treated DU-145 cells. Overall, TRAF2 can influence in vitro growth of TRAIL-treated DU-145 cells at least partially via regulating SIRT1 expression, and may be a potentially valuable biomarker predicting recurrence-free survival in prostate cancer patients.


Molecular Biology Reports | 2012

MTHFR 677C>T and 1298A>C polymorphisms and male infertility risk: a meta-analysis

Bingbing Wei; Zhuoqun Xu; Jun Ruan; Ming Zhu; Ke Jin; Deqi Zhou; Zeqiao Xu; Qiang Hu; Qiang Wang; Zhirong Wang


Urology | 2011

Re: Cheng et al.: shRNA Targeting PLC Inhibits Bladder Cancer Cell Growth In Vitro and In Vivo (Urology 2011;78:474e.7-474e.11)

Zhuoqun Xu; Bingbing Wei; Jun Ruan; Ming Zhu; Qiang Hu; Qiang Wang; Zhirong Wang


Urology | 2011

Re: Geng et al.: XRCC1 genetic polymorphism Arg399Gln and prostate cancer risk: a meta-analysis (Urology 2009;74:648-653).

Bingbing Wei; Zhuoqun Xu; Jun Ruan; Ming Zhu; Qiang Hu; Qiang Wang; Zhirong Wang; Zhiqiang Yan


/data/revues/00904295/v79i5/S0090429512000283/ | 2012

Re: Isen et al.: Experience With the Diagnosis and Management of Symptomatic Ureteric Stones During Pregnancy (Urology 2012;79:508-512)

Bingbing Wei; Zhuoqun Xu; Jun Ruan; Ming Zhu; Qiang Hu; Qiang Wang; Zhirong Wang; Zhiqiang Yan

Collaboration


Dive into the Zhirong Wang's collaboration.

Top Co-Authors

Avatar

Jun Ruan

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Qiang Hu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Bingbing Wei

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Ming Zhu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Qiang Wang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhuoqun Xu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhiqiang Yan

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Deqi Zhou

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Jian Zhang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Ke Jin

Nanjing Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge