Network


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

Hotspot


Dive into the research topics where Yuanyuan Chen is active.

Publication


Featured researches published by Yuanyuan Chen.


American Journal of Epidemiology | 2011

Association of Sequence Variants on Chromosomes 20, 11, and 5 (20q13.33, 11q23.3, and 5p15.33) With Glioma Susceptibility in a Chinese Population

Hongyan Chen; Yuanyuan Chen; Yao Zhao; Weiwei Fan; Keke Zhou; Yanhong Liu; Liangfu Zhou; Ying Mao; Qingyi Wei; Jianfeng Xu; Daru Lu

Two genome-wide association studies of glioma in European populations identified 14 genetic variants strongly associated with risk of glioma, but it is unknown whether these variants are associated with glioma risk in Asian populations. The authors genotyped these 14 variants in 976 glioma patients and 1,057 control subjects to evaluate their associations with risk of glioma, particularly high-grade glioma (glioblastoma; n = 312), in a Chinese population (2004-2009). Overall, the authors identified 3 susceptibility loci for glioma risk at 20q13.33 (RTEL1 rs6010620 (P = 2.79 × 10(-6))), 11q23.3 (PHLDB1 rs498872 (P = 3.8 × 10(-6))), and 5p15.33 (TERT rs2736100 (P = 3.69 × 10(-4))) in this study population; these loci were also associated with glioblastoma risk (20q13.33: RTEL1 rs6010620 (P = 3.57 × 10(-7)); 11q23.3: PHLDB1 rs498872 (P = 7.24 × 10(-3)); 5p15.33: TERT rs2736100 and TERT rs2736098 (P = 1.21 × 10(-4) and P = 2.84 × 10(-4), respectively)). This study provides further evidence for 3 glioma susceptibility regions at 20q13.33, 11q23.3, and 5p15.33 in Chinese populations.


Neuro-oncology | 2009

REV3L confers chemoresistance to cisplatin in human gliomas: The potential of its RNAi for synergistic therapy

Huibo Wang; Shu-Yu Zhang; Juan Lu; Wenting Wu; Lin Weng; Dan Chen; Yu Zhang; Zhipeng Lu; Jingmin Yang; Yuanyuan Chen; Xu Zhang; Xiaofeng Chen; Caihua Xi; Daru Lu; Shiguang Zhao

The REV3L gene, encoding the catalytic subunit of human polymerase zeta, plays a significant role in the cytotoxicity, mutagenicity, and chemoresistance of certain tumors. However, the role of REV3L in regulating the sensitivity of glioma cells to chemotherapy remains unknown. In this study, we investigated the expression of the REV3L gene in 10 normal brain specimens and 30 human glioma specimens and examined the value of REV3L as a potential modulator of cellular response to various DNA-damaging agents. Reverse transcriptase PCR/real-time PCR analysis revealed that REV3L was overexpressed in human gliomas compared with normal brain tissues. A glioma cell model with stable overexpression of REV3L was used to probe the role of REV3L in cisplatin treatment; upregulation of REV3L markedly attenuated cisplatin-induced apoptosis of the mitochondrial apoptotic pathway. We therefore assessed the REV3L-targeted treatment modality that combines suppression of REV3L expression using RNA interference (RNAi) with the cytotoxic effects of DNA-damaging agents. Downregulation of REV3L expression significantly enhanced the sensitivity of glioma cells to cisplatin, as evidenced by the increased apoptosis rate and marked alterations in the anti-apoptotic proteins B-cell lymphoma 2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-xl) and proapoptotic Bcl-2-associated x protein (Bax) expression levels, and reduced mutation frequencies in surviving glioma cells. These results suggest that REV3L may potentially contribute to gliomagenesis and play a crucial role in regulating cellular response to the DNA cross-linking agent cisplatin. Our findings indicate that RNAi targeting REV3L combined with chemotherapy has synergistic therapeutic effects on glioma cells, which warrants further investigation as an effective novel therapeutic regimen for patients with this malignancy.


Neuro-oncology | 2010

Analysis of specialized DNA polymerases expression in human gliomas: association with prognostic significance

Huibo Wang; Wenting Wu; Hongwei Wang; Yuanyuan Chen; Xiaotian Zhang; Jingmin Yang; Shiguang Zhao; Han Fei Ding; Daru Lu

Aberrant activation of the translesion DNA synthesis (TLS) pathway has been suggested to play a role in tumorigenesis by promoting genetic mutations. We therefore examined glioma specimens for the expression of specialized DNA polymerases involved in TLS and assessed their prognostic significance. The expression levels of DNA polymerase κ (Pol κ), Pol ι, and Pol η were assessed in 40 primary glioma samples and 10 normal brain samples using quantitative real-time PCR and Western blot analysis. Their prognostic significance was evaluated using a population-based tissue microarray derived from a cohort of 104 glioma patients. Overexpression of Pol κ and Pol ι was observed in 57.5% (23-40) and 27.5% (11-40) of patients, respectively, whereas no significant expression of Pol η was seen in the specimens. Immunohistochemical studies revealed positive Pol κ and Pol ι staining in 72 (69.2%) and 33 (31.7%) of the 104 glioma specimens, respectively. Pol κ expression was associated with advanced stages of the disease. Both Pol κ- and Pol ι-positive staining were associated with shorter survival in glioma patients (P < .001 and P = .014, respectively). A multivariate survival analysis identified Pol κ as an independent prognostic factor for glioma patients (P < .001). These findings demonstrate, for the first time, that the expression of Pol κ and Pol ι is deregulated in gliomas, and upregulation of Pol κ is associated with poorer prognosis in glioma patients.


Journal of Neuro-oncology | 2012

Overexpression of Golgi phosphoprotein-3 (GOLPH3) in glioblastoma multiforme is associated with worse prognosis

Jinxu Zhou; Tao Xu; Rong Qin; Yong Yan; Chao Chen; Yuanyuan Chen; Hong-Yu Yu; Chunyan Xia; Yicheng Lu; Xuehua Ding; Yuhai Wang; Xuejian Cai; Juxiang Chen

Golgi phosphoprotein-3 (GOLPH3), an important protein in mammalian target of rapamycin (mTOR) signaling, is overexpressed in and correlates with the pathological grade of glioma. However, the potential correlation between GOLPH3 and clinical outcome in patients with glioblastoma multiforme (GBM) remains unknown. In this study, we examined GOLPH3 expression in GBM by tissue microarray and correlated this measure to patient outcome. GOLPH3 expression in tumor tissue from 97 primary GBM patients was examined by tissue microarray and immunohistochemistry. Potential effects of GOLPH3 on tumor growth were also examined in representative cell lines (U251 and U87) by downregulating GOLPH3 with RNA interference. For this cohort, the median overall survival (OS) was 12xa0months [95xa0% confidence interval (CI): 10.31–13.69xa0months], and the median progression-free survival (PFS) was 10xa0months (95xa0%xa0CI: 7.33–12.67xa0months). Tissue microarray analysis revealed high GOLPH3 expression in 40 patients (40/97, 41.2xa0%) and low GOLPH3 expression in the remaining 57 patients (57/97, 58.8xa0%). Log-rank test showed that patients with low GOLPH3 expression had significantly longer median OS (15 versus 10xa0months in patients with high GOLPH3 expression, pxa0=xa00.009) and median PFS (12 versus 7xa0months, pxa0=xa00.015). Univariate and Cox analysis indicated that GOLPH3 was an independent prognostic factor for OS and PFS. In inxa0vitro experiments, GOLPH3 downregulation by small interfering RNA (siRNA) suppressed proliferation and clonogenic growth in cultured cell lines. These findings demonstrate that high GOLPH3 expression is associated with poor outcome of GBM patients.


Neuro-oncology | 2014

MicroRNA-377 inhibited proliferation and invasion of human glioblastoma cells by directly targeting specificity protein 1

Rui Zhang; Hui Luo; Wanghao Chen; Zhengxin Chen; Hongwei Wang; Yuanyuan Chen; Jingmin Yang; Xiaotian Zhang; Wenting Wu; Shu-Yu Zhang; Shuying Shen; Qingsheng Dong; Yaxuan Zhang; Tao Jiang; Daru Lu; Shiguang Zhao; Yongping You; Ning Liu; Huibo Wang

BACKGROUNDnIncreasing evidence has indicated that microRNAs (miRNAs) are strongly implicated in the initiation and progression of glioblastoma multiforme (GBM). Here, we identified a novel tumor suppressive miRNA, miR-377, and investigated its role and therapeutic effect for GBM.nnnMETHODSnMiRNA global screening was performed on GBM patient samples and adjacent nontumor brain tissues. The expression of miR-377 was detected by real-time reverse-transcription PCR. The effects of miR-377 on GBM cell proliferation, cell cycle progression, invasion, and orthotopic tumorigenicity were investigated The therapeutic effect of miR-377 mimic was explored in a subcutaneous GBM model. Western blot and luciferase reporter assay were used to identify the direct and functional target of miR-377.nnnRESULTSnMiR-377 was markedly downregulated in human GBM tissues and cell lines. Overexpression of miR-377 dramatically inhibited cell growth both in culture and in orthotopic xenograft tumor models, blocked G1/S transition, and suppressed cell invasion in GBM cells. Importantly, introduction of miR-377 could strongly inhibit tumor growth in a subcutaneous GBM model. Subsequent investigation revealed that specificity protein 1 (Sp1) was a direct and functional target of miR-377 in GBM cells. Silencing of Sp1 recapitulated the antiproliferative and anti-invasive effects of miR-377, whereas restoring the Sp1 expression antagonized the tumor-suppressive function of miR-377. Finally, analysis of miR-377 and Sp1 levels in human GBM tissues revealed that miR-377 is inversely correlated with Sp1 expression.nnnCONCLUSIONnThese findings reveal that miR-377/Sp1 signaling that may be required for GBM development and may consequently serve as a therapeutic target for the treatment of GBM.


Life Sciences | 2010

Chk2 down-regulation by promoter hypermethylation in human bulk gliomas

Hongwei Wang; Liqin Shen; Yuanyuan Chen; Xiaotian Zhang; Jundong Zhou; Zhi Wang; Chengyi Hu; Wu Yue; Huibo Wang

AIMSnGliomas account for 80% of malignant brain tumors. DNA damage response and subsequent checkpoint control pathways could maintain the integrity of the genome and thus defend tumorigenesis. Four kinases, ATM, ATR, ChK1 and Chk2 are the damage sensors and the early effectors in DNA damage responses. Given their importance, we investigated the transcriptional regulation of these four genes.nnnMAIN METHODSnTissues from ten normal brains and thirty human gliomas were utilized for mRNA analysis via real-time PCR. Another twelve normal brain tissues and forty gliomas were used for confirmation. Methylation-specific PCR (MSP) was used to determine the methylation status of the Chk2 promoter. Quantitative chromatin immunoprecipitation (ChIP) was used to measure the influence of methylation on Sp1 binding.nnnKEY FINDINGSnWe found that the expression of ATR, ChK1 and Chk2 in gliomas was significantly down-regulated relative to the normal brain tissues. The most significant reduction of expression was of the Chk2 gene, whose expression was approximately 10-fold decreased in gliomas (P<0.0001). Down-regulation of Chk2 was validated in the second real-time PCR analysis. This reduction in expression was partially due to promoter methylation. The Chk2 proximal promoter recruited Sp1 for transcriptional activation. We found that hypermethylation of the Chk2 promoter undermined the binding of the transcriptional factor Sp1.nnnSIGNIFICANCEnOur results indicate that Chk2 methylation could be involved in glioma carcinogenesis and Chk2 expression may potentially be used for the diagnosis of glioma.


International Journal of Cancer | 2011

Possible association between polymorphisms of human vascular endothelial growth factor A gene and susceptibility to glioma in a Chinese population

Rui Li; Yao Zhao; Weiwei Fan; Hongyan Chen; Yuanyuan Chen; Yanhong Liu; Gong Chen; Keke Zhou; Fengping Huang; Ying Mao; Liangfu Zhou; Daru Lu; Yin Yao Shugart

Vascular endothelial growth factor A (VEGFA), one of the most predominant mediators of pathologic angiogenesis, plays a critical role in glioma carcinogenesis and development via promoting tumor growth. We hypothesized that VEGFA polymorphisms may influence glioma risk. We recently genotyped 9 VEGFA single‐nucleotide polymorphisms (SNPs) in 766 glioma patients and 824 cancer‐free controls selected from a Chinese population. We evaluated the glioma risk conferred by individual SNPs, haplotypes as well as cumulative SNP effect. In the single‐locus analysis, we found that rs2010963 (G+405C, G‐634C) [odds ratio (OR) = 1.29; 95% confidence interval (CI) = 1.04–1.58; GC/CC vs. GG] and rs3025030 (OR = 2.21; 95% CI = 1.18–4.14; CC vs. GG/GC) were associated with increased risk for glioma, and rs3024994 (OR = 0.66; 95% CI = 0.47–0.94; CT/TT vs. CC) was associated with reduced glioma risk, albeit insignificant after Bonferroni correction for multiple comparisons. The haplotype‐based analysis revealed that AGG in block 1 and ATT, ACT in block 2 were associated with 20–40% reductions in glioma risk. The inverse association of haplotype AGG containing rs2010963G remained significant after correction for multiple testing (p = 0.002, pcorrected = 0.022). The aforementioned 3 SNPs revealed a significant cumulative risk effect; the increased risk for glioma was 1.38‐fold for each additional adverse genotype he or she carries (ptrend = 8.4 × 10−5). Our findings suggested that VEGFA variants may be involved in glioma risk. Larger studies with ethnically diverse populations are warranted to confirm the results reported in this investigation.


Neuro-oncology | 2015

VAMP8 facilitates cellular proliferation and temozolomide resistance in human glioma cells

Yuanyuan Chen; Delong Meng; Huibo Wang; Ruochuan Sun; Dongrui Wang; Jiajun Fan; Yingjie Zhao; Jingkun Wang; Cong Huai; Xiao Song; Rong Qin; Tao Xu; Dapeng Yun; Lingna Hu; Jingmin Yang; Xiaotian Zhang; Haoming Chen; Juxiang Chen; Hongyan Chen; Daru Lu

BACKGROUNDnMalignant glioma is a common and lethal primary brain tumor in adults. Here we identified a novel oncoprotein, vesicle-associated membrane protein 8 (VAMP8), and investigated its roles in tumorigenisis and chemoresistance in glioma.nnnMETHODSnThe expression of gene and protein were determined by quantitative PCR and Western blot, respectively. Histological analysis of 282 glioma samples and 12 normal controls was performed by Pearsons chi-squared test. Survival analysis was performed using the log-rank test and Cox proportional hazards regression. Cell proliferation and cytotoxicity assay were conducted using Cell Counting Kit-8. Autophagy was detected by confocal microscopy and Western blot.nnnRESULTSnVAMP8 was significantly overexpressed in human glioma specimens and could become a potential novel prognostic and treatment-predictive marker for glioma patients. Overexpression of VAMP8 promoted cell proliferation in vitro and in vivo, whereas knockdown of VAMP8 attenuated glioma growth by arresting cell cycle in the G0/G1 phase. Moreover, VAMP8 contributed to temozolomide (TMZ) resistance by elevating the expression levels of autophagy proteins and the number of autophagosomes. Further inhibition of autophagy via siRNA-mediated knockdown of autophagy-related gene 5 (ATG5) or syntaxin 17 (STX17) reversed TMZ resistance in VAMP8-overexpressing cells, while silencing of VAMP8 impaired the autophagic flux and alleviated TMZ resistance in glioma cells.nnnCONCLUSIONnOur findings identified VAMP8 as a novel oncogene by promoting cell proliferation and therapeutic resistance in glioma. Targeting VAMP8 may serve as a potential therapeutic regimen for the treatment of glioma.


Molecular Cancer | 2014

Down regulation of Thrombospondin2 predicts poor prognosis in patients with gastric cancer

Ruochuan Sun; Jifeng Wu; Yuanyuan Chen; Mingdian Lu; Shangxin Zhang; Daru Lu; Yongxiang Li

BackgroundThrombospondins (THBSs) are a family of multidomain and secreted matricellular Ca2+-binding glycoproteins which has at least five members encoded by independent genes. As a THBSs family member, Thrombospondin2 (THBS2) has been reported to regulate angiogenesis. Nevertheless, the functions and clinical significance of THBS2 still remains unclear in gastric cancer.MethodsThe mRNA and protein expression levels of THBS2 were assessed in 14 paired of gastric cancer specimens and corresponding normal mucosas using quantitative real-time PCR and western blot analysis. Immunohistochemistry of THBS2 and CD34 on population-based tissue microarrays consisting of 129 gastric cancer cases were used to evaluate the prognostic significance of THBS2 and microvessel density (MVD) of each sample. Survival analyses were performed by Kaplan–Meier method and Cox’s proportional hazards model. Colony formation assay, endothelial cell tube formation assay, cell migration assay and apoptosis analysis in MKN-45 and SGC-7901 cell lines were carried out to evaluate the effects of THBS2 on gastric cancer in vitro.Results85.71% (12 of 14) gastric cancer tissues expressed remarkably lower THBS2 in both mRNA and protein levels than the corresponding normal controls. Consistently, tissue microarray (TMA) results showed THBS2 levels were also inhibited in gastric cancer tissues compared with the normal controls. Moreover, we observed that patients with higher levels of THBS2 were significantly correlated with more favourable prognosis while decreased THBS2 expression were associated with poorer histological grades of gastric cancer. Additionally, our in vitro experiments further demonstrated that overexpression of THBS2 could impede both the proliferation rate and the tube formation of Human umbilical vein endothelial cells (HUVECs) in MKN-45 and SGC-7901 cell lines.ConclusionOur study suggests THBS2 is aberrantly expressed in gastric cancer and plays a critical role in cancer progression, which can be a potential prognosis predictor of gastric cancer.


Brain | 2015

c-Myc-miR-29c-REV3L signalling pathway drives the acquisition of temozolomide resistance in glioblastoma.

Hui Luo; Zhengxin Chen; Shuai Wang; Rui Zhang; Wenjin Qiu; Lin Zhao; Chenghao Peng; Ran Xu; Wanghao Chen; Hongwei Wang; Yuanyuan Chen; Jingmin Yang; Xiaotian Zhang; Shu-Yu Zhang; Dan Chen; Wenting Wu; Chunsheng Zhao; Gang Cheng; Tao Jiang; Daru Lu; Yongping You; Ning Liu; Huibo Wang

Resistance to temozolomide poses a major clinical challenge in glioblastoma multiforme treatment, and the mechanisms underlying the development of temozolomide resistance remain poorly understood. Enhanced DNA repair and mutagenesis can allow tumour cells to survive, contributing to resistance and tumour recurrence. Here, using recurrent temozolomide-refractory glioblastoma specimens, temozolomide-resistant cells, and resistant-xenograft models, we report that loss of miR-29c via c-Myc drives the acquisition of temozolomide resistance through enhancement of REV3L-mediated DNA repair and mutagenesis in glioblastoma. Importantly, disruption of c-Myc/miR-29c/REV3L signalling may have dual anticancer effects, sensitizing the resistant tumours to therapy as well as preventing the emergence of acquired temozolomide resistance. Our findings suggest a rationale for targeting the c-Myc/miR-29c/REV3L signalling pathway as a promising therapeutic approach for glioblastoma, even in recurrent, treatment-refractory settings.

Collaboration


Dive into the Yuanyuan Chen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Huibo Wang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hongwei Wang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Juxiang Chen

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge