Network


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

Hotspot


Dive into the research topics where Xiefeng Wang is active.

Publication


Featured researches published by Xiefeng Wang.


Journal of Cancer Research and Clinical Oncology | 2012

MiR-181d acts as a tumor suppressor in glioma by targeting K-ras and Bcl-2

Xiefeng Wang; Zhumei Shi; Xirui Wang; Lei Cao; Yingyi Wang; Junxia Zhang; Yu Yin; Hui Luo; Chunsheng Kang; Ning Liu; Tao Jiang; Yongping You

PurposeRecently, several microRNAs (miRNAs) were reported to be involved in the modulation of glioma development. The aim of our study was to determine the effect of miR-181d on the growth of glioma and to investigate whether this growth is modulated by targeting K-ras and Bcl-2.MethodsReal-time PCR was used to analyze the expression of miR-181d in human glioma samples and glioma cell lines. Apoptosis, cell cycle, and proliferation (MTT) assays were performed to assess the phenotypic changes in glioma cells. Immunohistochemistry was used to determine the expression of K-ras and Bcl-2 in glioma tissues, and a luciferase reporter assay was carried out to confirm whether K-ras and Bcl-2 are direct targets of miR-181d. Western blotting was used to identify the potential signaling pathways affected glioma cell growth by miR-181d. In vivo, xenograft tumors were examined for an anti-glioma effect of miR-181d.ResultsMiR-181d was down-regulated in human glioma samples and up-regulated in transfected glioma cells. Ectopic expression of miR-181d suppressed proliferation and triggered cell cycle arrest and apoptosis in glioma cell lines. K-ras and Bcl-2 were identified as direct targets of miR-181d and were up-regulated in glioma samples. The results showed evidence linking the tumor suppressor activity of miR-181d in glioma cells with the K-ras-related PI3K/AKT and MAPK/ERK pathways. Furthermore, xenograft tumors from miR-181d-treated U251 cells were suppressed in vivo.ConclusionMiR-181d may act as a glioma suppressor by targeting K-ras and Bcl-2.


Neuro-oncology | 2014

MiR-124 governs glioma growth and angiogenesis and enhances chemosensitivity by targeting R-Ras and N-Ras.

Zhumei Shi; Qiudan Chen; Chongyong Li; Lin Wang; Xu Qian; Chengfei Jiang; Xue Liu; Xiefeng Wang; Hai Li; Chunsheng Kang; Tao Jiang; Ling-Zhi Liu; Yongping You; Ning Liu; Bing-Hua Jiang

BACKGROUND Glioma is one of the most aggressive and lethal human brain tumors. Accumulating evidence shows that microRNAs play important roles in cancers, including glioma. Previous studies reported that miR-124 levels were downregulated in glioma specimens. Here, we further investigate the potential role of miR-124 in glioma. METHODS The expression levels of miR-124 were detected in glioma specimens by quantitative reverse transcriptase PCR. The direct targets of miR-124 were identified by bioinformatics analysis and were further validated by immunoblotting and luciferase reporter assay. The effects of miR-124 on glioma cell proliferation and chemosensitivity to temozolomide were analyzed by Cell-Counting Kit 8 assay. Apoptosis was evaluated by fluorescence activated cell sorting analysis. A xenograft model was used to study the effect of miR-124 on tumor growth and angiogenesis. RESULTS Expression levels of miR-124 were greatly downregulated in glioma specimens. related Ras viral oncogene homolog (R-Ras) and neuroblastoma Ras viral oncogene homolog (N-Ras) were identified as direct targets of miR-124. MiR-124 inhibited glioma cell growth, invasion, angiogenesis, and tumor growth and increased chemosensitivity to temozolomide treatment by negatively regulating the Ras family and its downstream signaling pathways: phosphatidylinositol-3 kinase/Akt and Raf/extracellular signal-regulated kinase 1/2. Furthermore, overexpression of R-Ras rescued the inhibitory effects of miR-124. Meanwhile, overexpression of R-Ras and N-Ras restored miR-124-inhibited vascular endothelial growth factor (VEGF) transcription activation. In clinical glioma specimens, protein levels of R-Ras and N-Ras were upregulated and inversely correlated with miR-124 expression levels. CONCLUSIONS Taken together, these results revealed that miR-124 levels in tumor tissues are associated with glioma occurrence, angiogenesis, and chemoresistance and that miR-124 may be used as a new diagnostic marker and therapeutic target for glioma in the future.


CNS Neuroscience & Therapeutics | 2012

MiR‐21 Modulates hTERT Through a STAT3‐Dependent Manner on Glioblastoma Cell Growth

Yingyi Wang; Guan Sun; Hui Luo; Xiefeng Wang; Fengming Lan; Xiao Yue; Linshan Fu; Peiyu Pu; Chunsheng Kang; Ning Liu; Yongping You

As an important oncogenic miRNA, miR‐21 has been reported to play crucial roles in glioblastoma (GBM) carcinogenesis. However, the precise biological function and molecular mechanism of miR‐21 in GBM remain elusive. This study is designed to explore the mechanism of miR‐21 involved in the control of GBM cell growth.


Neuro-oncology | 2013

Overexpressed let-7a inhibits glioma cell malignancy by directly targeting K-ras, independently of PTEN

Xirui Wang; Hui Luo; Hailin Li; Lei Cao; Xiefeng Wang; Wei Yan; Yingyi Wang; Junxia Zhang; Tao Jiang; Chunsheng Kang; Ning Liu; Yongping You

BACKGROUND Altered expression of micro(mi)RNAs has been shown to be associated with tumorigenesis and tumor progression. The expression of phosphatase and tensin homolog (PTEN) plays an important role in glioma and is regarded as a prognostic marker of glioma patients. The goal of this study was to investigate the function of lethal (let)-7a miRNA in glioma cell lines with different PTEN phenotypes. METHODS One hundred ninety-eight glioma tissues were used to profile miRNA expression. RESULTS Let-7a was shown to have lower expression in high-grade glioma than in low-grade glioma. Low expression of let-7a was correlated with poor prognosis of primary glioblastoma patients. We demonstrated that K-ras was a functional target for let-7a to induce cell cycle arrest, apoptosis, and inhibition of cell migration and invasion in vitro. Our further results showed no difference in malignancy inhibition induced by let-7a in 4 glioma cells, including U87 (PTEN null), U251 (PTEN mutant), LN229 (PTEN wild type), and LN229 (PTEN small interfering RNA). The phosphatidylinositol-3 kinase/Akt and mitogen-activated protein kinase/extracellular signal-regulated kinase pathways were inhibited by let-7a, and the inhibition effects had no difference in 4 glioma cells. We demonstrated that let-7a could induce suppression of glioma in vivo by generating a glioma xenograft model. CONCLUSION Our results indicated that let-7a suppresses its target transcript K-ras and inhibits glioma malignancy independent of PTEN expression.


Journal of Neuro-oncology | 2012

MicroRNAs involved in the EGFR/PTEN/AKT pathway in gliomas

Yingyi Wang; Xiefeng Wang; Junxia Zhang; Guan Sun; Hui Luo; Chunsheng Kang; Peiyu Pu; Tao Jiang; Ning Liu; Yongping You

Gliomas are the most common type of malignant primary brain tumor. Despite advances in surgery, radiation therapy, and chemotherapy, the prognosis of patients with gliomas has not significantly improved. MicroRNAs (miRNAs), a class of non-coding RNAs, 21–25 nucleotides long, negatively regulate the expression of target genes by interacting with specific sites in mRNAs, and play a critical role in the development of gliomas. The EGFR/PTEN/AKT pathway is a promising target for anti-glioma therapy. Recent studies have showed that regulation of the EGFR/PTEN/AKT pathway by miRNAs plays a major role in glioma progression, indicating a novel way to investigate the tumorigenesis, diagnosis, and therapy of gliomas. Here, we focus on recent findings of miRNAs with respect to the EGFR/PTEN/AKT pathway in gliomas.


Journal of Experimental & Clinical Cancer Research | 2014

Identification of intrinsic subtype-specific prognostic microRNAs in primary glioblastoma.

Rui Li; Kaiming Gao; Hui Luo; Xiefeng Wang; Yan Shi; Qingsheng Dong; Wenkang Luan; Yongping You

BackgroundGlioblastoma multiforme (GBM) is the most malignant type of glioma. Integrated classification based on mRNA expression microarrays and whole–genome methylation subdivides GBM into five subtypes: Classical, Mesenchymal, Neural, Proneural-CpG island methylator phenotype (G-CIMP) and Proneural-non G-CIMP. Biomarkers that can be used to predict prognosis in each subtype have not been systematically investigated.MethodsIn the present study, we used Cox regression and risk-score analysis to construct respective prognostic microRNA (miRNA) signatures in the five intrinsic subtypes of primary glioblastoma in The Cancer Genome Atlas (TCGA) dataset.ResultsPatients who had high-risk scores had poor overall survival compared with patients who had low-risk scores. The prognostic miRNA signature for the Mesenchymal subtype (four risky miRNAs: miR-373, miR-296, miR-191, miR-602; one protective miRNA: miR-223) was further validated in an independent cohort containing 41 samples.ConclusionWe report novel diagnostic tools for deeper prognostic sub-stratification in GBM intrinsic subtypes based upon miRNA expression profiles and believe that such signature could lead to more individualized therapies to improve survival rates and provide a potential platform for future studies on gene treatment for GBM.


European Journal of Human Genetics | 2012

Association between survivin −31G>C promoter polymorphism and cancer risk: a meta-analysis

Xiefeng Wang; Lili Huang; Yanjie Xu; Zhumei Shi; Yingyi Wang; Junxia Zhang; Xirui Wang; Lei Cao; Hui Luo; Jiawei Chen; Ning Liu; Yongmei Yin; Yongping You

Survivin is an inhibitor of apoptosis protein and has a crucial role in the development of cancer. The survivin −31G>C (rs9904341) promoter polymorphism influences survivin expression and has been implicated in cancer risk. However, conflicting results have been published from studies on the association between survivin −31G>C polymorphism and the risk of cancer. To clarify the role of this polymorphism in cancer, we performed a meta-analysis of all available and relevant published studies, involving a total of 3485 cancer patients and 3964 control subjects. Odds ratios (ORs) and 95% confidence intervals (CIs) were used to assess the strength of the associations. The overall results indicated that the variant genotypes were associated with a significantly increased cancer risk (CC vs GG: OR=1.58, 95% CI=1.20–2.10; CC/GC vs GG: OR=1.23, 95% CI=1.00–1.51; CC vs GG/GC: OR=1.51, 95% CI=1.23–1.85). In the stratified analyses, significantly increased risk was associated with the Asian populations (CC vs GG: OR=1.67, 95% CI=1.16–2.40; CC vs GG/GC: OR=1.50, 95% CI=1.17–1.91). We also performed the analyses by cancer type, and no statistical association was observed. The results suggest that the survivin −31G>C promoter polymorphism might be associated with an increased risk of cancer, especially in the Asian populations.


Oncotarget | 2015

NF-κB/RelA-PKM2 mediates inhibition of glycolysis by fenofibrate in glioblastoma cells

Dongfeng Han; Wenjin Wei; Xincheng Chen; Yaxuan Zhang; Yingyi Wang; Junxia Zhang; Xiefeng Wang; Tianfu Yu; Qi Hu; Ning Liu; Yongping You

Aerobic glycolysis (production of lactate from glucose in the presence of oxygen) is a hallmark of cancer. Fenofibrate is a lipid-lowering drug and an agonist of the peroxisome proliferator-activated receptor alpha (PPARα). We found that FF inhibited glycolysis in a PPARα-dependent manner in glioblastoma cells. Fenofibrate inhibited the transcriptional activity of NF-κB/RelA and also disrupted its association with hypoxia inducible factor1 alpha (HIF1α), which is required for the binding of NF-κB/RelA to the PKM promoter and PKM2 expression. High ratios of PKM2/PKM1 promote glycolysis and inhibit oxidative phosphorylation, thus favoring aerobic glycolysis. Fenofibrate decreased the PKM2/PKM1 ratio and caused mitochondrial damage. Given that fenofibrate is a widely used non-toxic drug, we suggest its use in patients with glioblastoma multiforme (GBM).


Cancer Epidemiology | 2013

Association between polymorphisms in interleukin-4Rα and interleukin-13 and glioma risk: A meta-analysis

Guan Sun; Xiefeng Wang; Lei Shi; Xiao Yue; Linshan Fu; Chen Chen; Zhengyi Li; Tianhong Pan; Zhengqiang Wan

INTRODUCTION It has been suggested that allergies are inversely associated with glioma risk. Single nucleotide polymorphisms in two allergy-related genes [interleukin (IL)-4Rα, IL-13] have been implicated in susceptibility to glioma; however, results from the published studies remained inconclusive. METHODS To derive a more precise relationship, we conducted a meta-analysis including seven case-control studies that investigated the influence of IL-4Rα rs1801275 and IL13 rs20541 polymorphisms on glioma risk. Data were extracted from these studies and pooled odds ratios (OR) with 95% confidence intervals (CI) were used to investigate the strength of the association. RESULTS Overall, the pooled analysis showed that there was no significant association between the IL-4Rα rs1801275 polymorphism and glioma risk (OR = 0.99, 95%CI: 0.79-1.25, AG/GG vs. AA). However, we found that the IL13 rs20541 variant genotypes (GA/AA) were significantly associated with reduced risk for glioma (OR = 0.85, 95%CI: 0.75-0.97, GA/AA vs. GG). In the stratified analyses by ethnicity, marginally significant association between the IL13 rs20541 polymorphism and decreased glioma risk was found among Asian populations in dominant models (OR = 0.84, 95%CI: 0.70-1.00, GA/AA vs. GG). CONCLUSIONS This meta-analysis suggests that the IL13 rs20541 but not the IL-4Rα rs1801275 polymorphism may be a genetic predictor for glioma. More studies with larger sample size are warranted to further elucidate the impact of the IL13 rs20541 polymorphism on glioma risk.


Tumor Biology | 2015

Fenofibrate induces G0/G1 phase arrest by modulating the PPARα/FoxO1/p27kip pathway in human glioblastoma cells

Dongfeng Han; Junxia Zhang; Wenjin Wei; Tao Tao; Qi Hu; Yingyi Wang; Xiefeng Wang; Ning Liu; Yongping You

Fenofibrate, a fibric acid derivative, is known to possess lipid-lowering effects. Although fenofibrate-induced peroxisome proliferator-activated receptor alpha (PPARα) transcriptional activity has been reported to exhibit anticancer effects, the underlying mechanisms are poorly understood. In this study, we investigated the mechanisms behind the antiproliferative effects of fenofibrate in U87MG cells (human glioma cell line) using the WST-8 Cell Proliferation Assay Kit. Furthermore, we examined genome-wide gene expression profiles and molecular networks using the DAVID online software. Fenofibrate reduced the expression of 405 genes and increased the expression of 2280 genes. DAVID analysis suggested that fenofibrate significantly affected cell cycle progression and pathways involved in cancer, including the mTOR signaling pathway and insulin signaling pathway. Results of flow cytometry analysis indicated that fenofibrate induced cell cycle G0/G1 arrest in U87MG cells. Furthermore, we identified the FoxO1–p27kip signaling axis to be involved in fenofibrate-induced cell cycle arrest. Our findings suggest that in addition to its known lipid-lowering effects, fenofibrate may be used as an antitumor agent in glioma therapy.

Collaboration


Dive into the Xiefeng Wang's collaboration.

Top Co-Authors

Avatar

Yongping You

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Ning Liu

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Yingyi Wang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Junxia Zhang

Tianjin Medical University General Hospital

View shared research outputs
Top Co-Authors

Avatar

Hui Luo

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei Yan

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Chunsheng Kang

Tianjin Medical University General Hospital

View shared research outputs
Top Co-Authors

Avatar

Xirui Wang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Guan Sun

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Qi Hu

Nanjing Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge