Network


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

Hotspot


Dive into the research topics where Jiucun Wang is active.

Publication


Featured researches published by Jiucun Wang.


Nature Genetics | 2011

A genome-wide association study identifies two new lung cancer susceptibility loci at 13q12.12 and 22q12.2 in Han Chinese

Zhibin Hu; Chen Wu; Yongyong Shi; Huan Guo; Xueying Zhao; Zhihua Yin; Lei Yang; Juncheng Dai; Lingmin Hu; Wen Tan; Zhiqiang Li; Qifei Deng; Jiucun Wang; Wei Wu; Guangfu Jin; Jiang Y; Dianke Yu; Guoquan Zhou; Hongyan Chen; Peng Guan; Yijiang Chen; Yongqian Shu; Lin Xu; Xiangyang Liu; Li Liu; Ping Xu; Baohui Han; Chunxue Bai; Yuxia Zhao; Haibo Zhang

Lung cancer is the leading cause of cancer-related deaths worldwide. To identify genetic factors that modify the risk of lung cancer in individuals of Chinese ancestry, we performed a genome-wide association scan in 5,408 subjects (2,331 individuals with lung cancer (cases) and 3,077 controls) followed by a two-stage validation among 12,722 subjects (6,313 cases and 6,409 controls). The combined analyses identified six well-replicated SNPs with independent effects and significant lung cancer associations (P < 5.0 × 10−8) located in TP63 (rs4488809 at 3q28, P = 7.2 × 10−26), TERT-CLPTM1L (rs465498 and rs2736100 at 5p15.33, P = 1.2 × 10−20 and P = 1.0 × 10−27, respectively), MIPEP-TNFRSF19 (rs753955 at 13q12.12, P = 1.5 × 10−12) and MTMR3-HORMAD2-LIF (rs17728461 and rs36600 at 22q12.2, P = 1.1 × 10−11 and P = 6.2 × 10−13, respectively). Two of these loci (13q12.12 and 22q12.2) were newly identified in the Chinese population. These results suggest that genetic variants in 3q28, 5p15.33, 13q12.12 and 22q12.2 may contribute to the susceptibility of lung cancer in Han Chinese.


Nature Genetics | 2012

Association analyses identify multiple new lung cancer susceptibility loci and their interactions with smoking in the Chinese population

Jing Dong; Zhibin Hu; Chen Wu; Huan Guo; Baosen Zhou; Jiachun Lv; Daru Lu; Kexin Chen; Yongyong Shi; Minjie Chu; Cheng Wang; Ruyang Zhang; Juncheng Dai; Jiang Y; Songyu Cao; Zhenzhen Qin; Dianke Yu; Hongxia Ma; Guangfu Jin; Jianhang Gong; Chongqi Sun; Xueying Zhao; Zhihua Yin; Lei Yang; Zhiqiang Li; Qifei Deng; Jiucun Wang; Wei Wu; Hong Zheng; Guoquan Zhou

To find additional susceptibility loci for lung cancer, we tested promising associations from our previous genome-wide association study (GWAS) of lung cancer in the Chinese population in an extended validation sample size of 7,436 individuals with lung cancer (cases) and 7,483 controls. We found genome-wide significant (P < 5.0 × 10−8) evidence for three additional lung cancer susceptibility loci at 10p14 (rs1663689, close to GATA3, P = 2.84 × 10−10), 5q32 (rs2895680 in PPP2R2B-STK32A-DPYSL3, P = 6.60 × 10−9) and 20q13.2 (rs4809957 in CYP24A1, P = 1.20 × 10−8). We also found consistent associations for rs247008 at 5q31.1 (IL3-CSF2-P4HA2, P = 7.68 × 10−8) and rs9439519 at 1p36.32 (AJAP1-NPHP4, P = 3.65 × 10−6). Four of these loci showed evidence for interactions with smoking dose (P = 1.72 × 10−10, P = 5.07 × 10−3, P = 6.77 × 10−3 and P = 4.49 × 10−2 for rs2895680, rs4809957, rs247008 and rs9439519, respectively). These results advance our understanding of lung cancer susceptibility and highlight potential pathways that integrate genetic variants and smoking in the development of lung cancer.


The FASEB Journal | 2009

Functional characterization of a promoter polymorphism in APE1/Ref-1 that contributes to reduced lung cancer susceptibility

Juan Lu; Shuyu Zhang; Dan Chen; Huibo Wang; Wenting Wu; Xiaotian Wang; Yunping Lei; Jiucun Wang; Ji Qian; Weiwei Fan; Zhibin Hu; Li Jin; Hongbing Shen; Wei Huang; Qingyi Wei; Daru Lu

Apurinic/apyrimidinic endonuclease 1/redox effector factor‐1 (APE1/Ref‐1) is a ubiquitous multifunctional protein that possesses both DNA‐repair and redox regulatory activities. Although it was originally identified as a DNA‐repair enzyme, accumulating evidence supports a role of APE1/Ref‐1 in tumor development. To investigate association between APE1/ Ref‐1 polymorphisms and lung cancer risk in Chinese populations, we first genotyped three variants of APE1/ Ref‐1 and found a ‐141 T‐to‐G variant (rs1760944) in the promoter associated with decreased risk of lung cancer [odds ratio (OR) = 0.62 for GG;P=0.043]. Similar results were obtained in a follow‐up replication study. Combined data from the two studies comprising a total of 1072 lung cancer patients and 1064 cancer‐free control participants generated a more significant association (P= 0.002). We observed lower APE1/Ref‐1 mRNA levels in the presence of the protective G allele in human peripheral blood mononuclear cells and normal lung tissues. The ‐ 141G‐allele‐promoter construct exhibited decreased luciferase reporter gene expression. Electrophoretic mobility shift assays and surface plasmon resonance analysis showed that the — 141G allele impaired the binding affinity of some transcription factor, accounting for lower APE1/Ref‐1‐promoter activity. Supershift assays further revealed that the protein of interest was octamer‐binding transcription factor‐1 (Oct‐1). Chromatin immunoprecipitation reconfirmed binding of Oct‐1 to the APE1/Ref‐1 — 141‐promoter region. We also found that Oct‐1 conferred attenuated transactivation capacity toward the — 141G variant by exogenously introducing Oct‐1. These data indicate that genetic variations in APE1/Ref‐1 may modify susceptibility to lung cancer and provide new insights into an unexpected effect of APE1/Ref‐1 on lung carcinogenesis.—Lu, J., Zhang, S., Chen, D., Wang, H., Wu, W., Wang, X., Lei, Y., Wang, J., Qian, J., Fan, W., Hu, Z., Jin, L., Shen, H., Huang, W., Wei, Q., Lu, D. Functional characterization of a promoter polymorphism in APE1/Ref‐1 that contributes to reduced lung cancer susceptibility. FASEB J. 23, 3459–3469 (2009). www.fasebj.org


Clinical Cancer Research | 2004

Defective Expression of Transforming Growth Factor β Receptor Type II Is Associated with CpG Methylated Promoter in Primary Non-Small Cell Lung Cancer

Hongtao Zhang; Xiaofeng Chen; Minghua Wang; Jiucun Wang; Qing-Yuan Qi; Rongmei Zhang; Weiqing Xu; Qing-Yan Fei; Fei Wang; Qi-Qun Cheng; Feng Chen; Cheng-Song Zhu; Shiheng Tao; Zewei Luo

Purpose: Reduced expression of the transforming growth factor β receptor type II (TGFβRII), a key inhibitor of epithelial cell growth and tumor suppressor gene, was reported frequently in many types of tumors including non-small cell lung cancer (NSCLC). This study explored the significance of the TGFβRII gene in NSCLC carcinogenesis. Experimental Design: With 43 independent pairs of tumor and paracarcinoma tissue samples from patients with primary NSCLC, we carried out PCR-denaturing gradient gel electrophoresis screening for DNA variants over the coding sequence of the TGFβRII gene, immunohistochemical assay of TGFβRII expression, methylation-specific PCR analysis, and semiquantitative reverse transcription-PCR. Results: The PCR-denaturing gradient gel electrophoresis did not detect variation in the whole coding sequence of the TGFβRII gene, but the immunohistochemistry experiment revealed reduced or lost expression of the gene in 44% (19 of 43) of the tumor samples. The methylation analysis on the 19 pairs detected the frequent occurrence of methylated TGFβRII promoter in tumor tissues, whereas most of the paracarcinoma tissues were free of methylation. The reduced TGFβRII expression was highly significantly associated with the methylation event (P < 10−4). The reverse transcription-PCR analysis demonstrated a clear agreement between reduced TGFβRII expression and decreased mRNA level of the gene in the tumor tissue samples. Conclusions: TGFβRII plays an important role as a tumor suppressor in NSCLC carcinogenesis. The defective expression may serve as one of most important molecular mechanisms in explaining progression of the disease. In particular, aberrant 5′ CpG methylation of the gene has explained the down-regulation of the gene at a transcriptional level.


Arthritis Research & Therapy | 2010

Attenuation of fibrosis in vitro and in vivo with SPARC siRNA

Jiucun Wang; Syeling Lai; Xinjian Guo; Xuefeng Zhang; Benoit de Crombrugghe; Sonali Sonnylal; Frank C. Arnett; Xiaodong Zhou

IntroductionSPARC is a matricellular protein, which, along with other extracellular matrix components including collagens, is commonly over-expressed in fibrotic diseases. The purpose of this study was to examine whether inhibition of SPARC can regulate collagen expression in vitro and in vivo, and subsequently attenuate fibrotic stimulation by bleomycin in mouse skin and lungs.MethodsIn in vitro studies, skin fibroblasts obtained from a Tgfbr1 knock-in mouse (TBR1CA; Cre-ER) were transfected with SPARC siRNA. Gene and protein expressions of the Col1a2 and the Ctgf were examined by real-time RT-PCR and Western blotting, respectively. In in vivo studies, C57BL/6 mice were induced for skin and lung fibrosis by bleomycin and followed by SPARC siRNA treatment through subcutaneous injection and intratracheal instillation, respectively. The pathological changes of skin and lungs were assessed by hematoxylin and eosin and Massons trichrome stains. The expression changes of collagen in the tissues were assessed by real-time RT-PCR and non-crosslinked fibrillar collagen content assays.ResultsSPARC siRNA significantly reduced gene and protein expression of collagen type 1 in fibroblasts obtained from the TBR1CA; Cre-ER mouse that was induced for constitutively active TGF-β receptor I. Skin and lung fibrosis induced by bleomycin was markedly reduced by treatment with SPARC siRNA. The anti-fibrotic effect of SPARC siRNA in vivo was accompanied by an inhibition of Ctgf expression in these same tissues.ConclusionsSpecific inhibition of SPARC effectively reduced fibrotic changes in vitro and in vivo. SPARC inhibition may represent a potential therapeutic approach to fibrotic diseases.


Molecular Carcinogenesis | 2013

Genetic variations of mTORC1 genes and risk of gastric cancer in an eastern chinese population

Jing He; Meng Yun Wang; Li Xin Qiu; Mei Ling Zhu; Ting Yan Shi; Xiao Yan Zhou; Meng Hong Sun; Ya Jun Yang; Jiucun Wang; Li Jin; Ya Nong Wang; Jin Li; Hong Ping Yu; Qingyi Wei

Mammalian target of rapamycin complex 1 (mTORC1) plays an important role in maintaining proper cellular functions, and genetic variations in this complex may affect cancer risk. In this study, we examined the associations between eight potential functional single nucleotide polymorphisms in the mTORC1 genes (rs2536T>C and rs1883965G>A for mTOR, rs3160T>C, and rs26865A>G for mLST8, rs3751934C>A, rs1062935T>C, rs3751932T>C, and rs12602885G>A for Raptor, not included in published gastric cancer genome‐wide association studies) and gastric cancer risk in 1125 gastric cancer cases and 1196 cancer‐free controls. We performed conditional logistic regression and multifactor dimensionality reduction (MDR) analyses to assess their associations with gastric cancer risk. We also used false‐positive report probabilities (FPRP) for assessing significant findings. We found that only the rs1883965A variant genotypes were associated with an increased risk of gastric cancer (AG vs. GG: adjusted odds ratio (OR) = 1.26, 95% confidence interval (CI) = 1.00–1.59; AA vs. GG: adjusted OR = 1.85, 95% CI = 0.67–5.16 and dominant model: adjusted OR = 1.28, 95% CI = 1.03–1.61). Patients with ≥1 risk genotypes of mTOR had significant increased risk (adjusted OR = 1.25, 95% CI = 1.04–1.49), compared with those having zero risk genotypes. In the stratified analysis, the risk effect of the rs1883965 AG/AA genotypes was evident in subgroups of ever‐smokers, non‐gastric cardia adenocarcinoma and clinical stage I + II, which were noteworthy findings as evaluated by FPRP. The MDR analysis identified smoking status and rs1883965 as the strongest two‐factors for gastric cancer risk. These data support the hypothesis that functional polymorphisms of mTOR may contribute to gastric cancer risk. Clearly, our results require validation in larger studies with different ethnic populations.


Clinical Cancer Research | 2012

Predictive Value of XRCC1 Gene Polymorphisms on Platinum-Based Chemotherapy in Advanced Non–Small Cell Lung Cancer Patients: A Systematic Review and Meta-analysis

Junjie Wu; Jie Liu; Yuhao Zhou; Jun Ying; Houdong Zou; Shicheng Guo; Lei Wang; Naiqing Zhao; Jianjun Hu; Daru Lu; Li Jin; Qiang Li; Jiucun Wang

Purpose: Published data have shown conflicting results about the relationship between X-ray repair cross-complementing group 1 (XRCC1) gene polymorphisms (Arg399Gln and Arg194Trp) and clinical outcome of platinum-based chemotherapy in patients with advanced non–small cell lung cancer (NSCLC). A meta-analysis is needed to provide a systematic review of the published findings. Experimental Design: We conducted a systematic review and meta-analysis to evaluate the predictive value of XRCC1 gene polymorphisms on clinical outcome up to October 1, 2010. The quality of each study was scored on the basis of predefined criteria. Results: A total of 13 eligible follow-up studies met all the inclusion criteria. The XRCC1194Trp allele was found to be significantly associated with a favorable response rate relative to 194Arg [Trp vs. Arg: OR, 1.88; 95% confidence interval (CI), 1.48–2.38]. XRCC1399Gln was less favorably associated with both response rate (Gln vs. Arg: OR, 0.67; 95% CI, 0.52–0.87) and overall survival (Gln vs. Arg: HR, 1.30; 95% CI, 1.04–1.63) than 399Arg in analyses using all available studies; but these associations became insignificant when only high-quality studies were used. Conclusion: These findings suggest a predictive role for XRCC1 gene polymorphisms in clinical outcome. However, the role of 399Gln could be considered controversial because its impact on clinical outcome was insignificant in high-quality studies. These findings show the importance of establishing suitable criteria, including genetic epidemiologic, phenotypic, and clinical criteria, to improve quality control of study design and methods in pharmacogenomic studies related to XRCC1 gene polymorphism. Clin Cancer Res; 18(14); 3972–81. ©2012 AACR.


Journal of Medical Genetics | 2013

Confirmation of papillary thyroid cancer susceptibility loci identified by genome-wide association studies of chromosomes 14q13, 9q22, 2q35 and 8p12 in a Chinese population

Yu Long Wang; Shou Hao Feng; Shicheng Guo; Wen Jun Wei; Duan Shu Li; Yu Wang; Xiaofeng Wang; Zhuo Ying Wang; Yan Yun Ma; Li Jin; Qing Hai Ji; Jiucun Wang

Background Five single nucleotide polymorphisms (SNPs) were previously reported to be associated with thyroid cancer in European populations in two genome-wide association studies (GWAS): rs965513 (9q22.33), rs944289 (14q13.3), rs116909374 (14q13.3), rs966423 (2q35) and rs2439302 (8p12). Only the first two SNPs have been validated in independent populations and none were replicated in Chinese populations. Methods The above five SNPs were genotyped in 845 papillary thyroid cancer (PTC) and 503 benign thyroid tumour (BN) patients and 1005 controls in a Chinese population using the SNaPshot multiplex single nucleotide extension system. Results Significant associations were detected among PTC and rs944289 (p=8.007e-11), rs965513 (p=1.013e-4), rs966423 (p=1.688e-3) and rs2439302 (p=1.096e-4) in a dominant model, while the rs116909374 SNP was not detected in the Chinese population. The PTC risk increased with rise in accumulative numbers of risk alleles carried by individuals (p=5.929e-13). The PTC OR of carriers of six risk alleles (1.4% of the control population) was 23.587 compared with non-risk homozygotes (1.0% of the control population, with zero risk alleles). No individuals were homozygous for all the four SNPs (carriers of eight risk alleles) and only three PTC cases were carriers of seven risk alleles. A significant association between 14q13.3 SNP rs944289T and BN was also found (p=0.0014). Conclusions Four candidate loci, rs965513 (9q22.33), rs944289 (14q13.3), rs966423 (2q35) and rs2439302 (8p12), identified by GWAS for PTC risk were confirmed in a Chinese population. The PTC risk of accumulative risk allele carriers increased with the number of risk alleles.


PLOS ONE | 2012

Potentially functional variants of PLCE1 identified by GWASs contribute to gastric adenocarcinoma susceptibility in an Eastern Chinese population

Mengyun Wang; Ruoxin Zhang; Jing He; Li-Xin Qiu; Jin Li; Yanong Wang; Menghong Sun; Yajun Yang; Jiucun Wang; Yang J; Ji Qian; Li Jin; Hongxia Ma; Qingyi Wei; Xiaoyan Zhou

Background Recent genome-wide association studies (GWAS) have found a single nucleotide polymorphism (SNP, rs2274223 A>G) in PLCE1 to be associated with risk of gastric adenocarcinoma. In the present study, we validated this finding and also explored the risk associated with another unreported potentially functional SNP (rs11187870 G>C) of PLCE1 in a hospital-based case-control study of 1059 patients with pathologically confirmed gastric adenocarcinoma and 1240 frequency-matched healthy controls. Methodology/Principal Findings We determined genotypes of these two SNPs by the Taqman assay and used logistic regression models to estimate odds ratios (ORs) and 95% confidence intervals (95% CI). We found that a significant higher gastric adenocarcinoma risk was associated with rs2274223 variant G allele (adjusted OR = 1.35, 95% CI = 1.14–1.60 for AG+GG vs. AA) and rs11187870 variant C allele (adjusted OR = 1.26, 95% CI = 1.05–1.50 for CG+CC vs. GG). We also found that the number of combined risk alleles (i.e., rs2274223G and rs11187870C) was associated with risk of gastric adenocarcinoma in an allele-dose effect manner (P trend = 0.0002). Stratification analysis indicated that the combined effect of rs2274223G and rs11187870C variant alleles was more evident in subgroups of males, non-smokers, non-drinkers and patients with gastric cardia adenocarcinoma. Further real-time PCR results showed that expression levels of PLCE1 mRNA were significantly lower in tumors than in adjacent noncancerous tissues (0.019±0.002 vs. 0.008±0.001, P<0.05). Conclusions/Significances Our results further confirmed that genetic variations in PLCE1 may contribute to gastric adenocarcinoma risk in an eastern Chinese population.


PLOS ONE | 2014

Pri-miR-124 rs531564 and pri-miR-34b/c rs4938723 polymorphisms are associated with decreased risk of esophageal squamous cell carcinoma in Chinese populations.

Junjie Zhang; Xuewen Huang; Juanjuan Xiao; Yajun Yang; Yinghui Zhou; Xiaofeng Wang; Qingmei Liu; Yang J; Mengyun Wang; Li-Xin Qiu; Yabiao Zheng; Ping Zhang; Jin Li; Ya Nong Wang; Qingyi Wei; Li Jin; Jiucun Wang; Minghua Wang

MicroRNAs are a new class of small non-protein-coding RNAs that sometimes function as tumor suppressors or oncogenes. Aberrant expression and structural alteration of microRNAs have been reported to be involved in tumorigenesis and cancer development. Recently, rs531564/pri-miR-124-1, rs4938723/pri-miR-34b/c, rs7372209/pri-miR-26a-1, rs895819/pre-miR-27a, and rs11134527/pri-miR-218 were reported to be associated with risks of various cancers. In order to evaluate the relationship of these SNPs and esophageal squamous cell carcinoma (ESCC) risk, we conducted a case-control study with 1109 ESCC patients and 1275 control subjects to examine the potential association of these pri/pre-miRNA polymorphisms with ESCC susceptibility. As a result, two SNPs were associated with a significant risk of ESCC. We found that the GG genotype of pri-miR-124-1 rs531564 was associated to a significantly decreased risk of ESCC comparing with the CC/CG genotypes (p = 0.005; OR = 0.61, 95% CI = 0.43–0.86). In addition, the CC genotype of pri-miR-34b/c rs4938723 was associated with a significant decreased risk of ESCC (CC VS. TT/TC: p = 0.007, OR = 0.82, 95% CI = 0.71–0.95) in Chinese population. The present study provides the first evidence that pri-miR-124-1 rs531564 and pri-miR-34 rs4938723 were associated with the risk of ESCC in Chinese population.

Collaboration


Dive into the Jiucun Wang'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

Xiaodong Zhou

University of Texas Health Science Center at Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge