Network


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

Hotspot


Dive into the research topics where Wenjuan Li is active.

Publication


Featured researches published by Wenjuan Li.


The Journal of Pathology | 2009

FoxM1 is up‐regulated in gastric cancer and its inhibition leads to cellular senescence, partially dependent on p27kip1

Jiping Zeng; Lixiang Wang; Qiao Li; Wenjuan Li; Magnus Björkholm; Jihui Jia; Dawei Xu

The FoxM1 transcription factor, a master regulator of mitotic gene expression, promotes the pathogenesis of several malignancies. However, little is known about its expression and function in gastric cancer. In the present study we determined whether FoxM1 is over‐expressed in gastric cancer, and whether it is required to maintain an immortal phenotype of gastric cancer cells. The over‐expression of FoxM1 was observed in 37/42 tumour specimens from patients with gastric cancer. When FoxM1 in gastric cancer cells was knocked‐down, impaired clonogenicity and cellular senescence occurred independently of p53 and p16 status. FoxM1 depletion led to the down‐regulation of its target genes c‐MYC and Skp2, coupled with the accumulation of the CDK inhibitor p27kip1. Importantly, the FoxM1 inhibition‐mediated cellular senescence and clonogenic defect was attenuated by the abolition of p27kip1 induction. Telomerase reverse transcriptase, the key component of telomerase essential for cellular immortalization, was also inhibited in the FoxM1‐depleted cells. Taken together, the FoxM1 gene is aberrantly activated in gastric cancer and its inhibition triggers p53‐ and p16‐independent senescence of cancer cells by regulating the expression of p27kip1 and other targets. These findings provide mechanistic insights into the role of FoxM1 in the pathogenesis of gastric cancer, which may have diagnostic and therapeutic implications in gastric cancer. Copyright


Molecular Cancer Research | 2013

FoxM1 is Overexpressed in Helicobacter pylori–Induced Gastric Carcinogenesis and Is Negatively Regulated by miR-370

Yimin Feng; Lixiang Wang; Jiping Zeng; Li Shen; Xiuming Liang; Han Yu; Shili Liu; Zhifang Liu; Yundong Sun; Wenjuan Li; Chunyan Chen; Jihui Jia

Helicobacter pylori (H. pylori) infections are strongly implicated in human gastric mucosa–associated diseases. Forkhead box M1 (FoxM1), a key positive regulator of cell proliferation, is overexpressed in gastric cancer. MicroRNAs are important post-transcriptional regulators of gene expression. In this study, the effects of H. pylori infection on FoxM1 expression and possible mechanisms of carcinogenesis were explored. The expression of FoxM1 was gradually increased in human gastric specimens from inflammation to cancer. FoxM1 upregulation was time- and concentration-dependent in gastric epithelial-derived cell lines infected with H. pylori. CagA, a key virulence factor of H. pylori, was associated with increased FoxM1 expression. H. pylori and CagA inhibited the expression of p27Kip1 (CDKN1B) and promoted cell proliferation by upregulating FoxM1. The expression of miR-370 was decreased in human gastritis and gastric cancer. FoxM1 was directly downregulated by miR-370 in gastric cell lines. H. pylori and CagA inhibited miR-370 expression, which led to overexpression of FoxM1 and cell proliferation. Furthermore, the overexpression of FoxM1 and reduced expression of miR-370 was confirmed in H. pylori–infected C57BL/6J mice. H. pylori infection and CagA upregulated FoxM1 expression, dependent on miR-370, altered the expression of p27Kip1, and promoted proliferation in gastric cells. Implications: These findings delineate the mechanisms governing FoxM1 regulation and the role of H. pylori in the process of gastric carcinogenesis. Mol Cancer Res; 11(8); 834–44. ©2013 AACR.


Journal of Cellular and Molecular Medicine | 2014

RUNX3 regulates vimentin expression via miR‐30a during epithelial–mesenchymal transition in gastric cancer cells

Zhifang Liu; Long Chen; Xinchao Zhang; Xia Xu; Huaixin Xing; Yingjie Zhang; Wenjuan Li; Han Yu; Jiping Zeng; Jihui Jia

Runt‐related transcription factor 3 (RUNX3) is a putative tumour suppressor via regulating the expression of a series of target genes. Clinical studies demonstrated that loss of RUNX3 expression is associated with gastric cancer progression and poor prognosis, but the underlying mechanism is not entirely clear. Accumulating evidence shows that the epithelial–mesenchymal transition (EMT) plays an important role in cancer relapse and metastasis. Therefore, we addressed whether RUNX3 has a role in the EMT in gastric cancer. Knockdown of RUNX3 promoted cell invasion and increased the protein expression of the mesenchymal marker vimentin in human gastric cancer cells. Overexpression of RUNX3 suppressed cell invasion and decreased the protein expression of vimentin in the cells and inhibited gastric cancer cells colonization in nude mice. Furthermore, overexpression of RUNX3 increased the expression of microRNA‐30a (miR‐30a), and miR‐30a directly targeted the 3′ untranslated region of vimentin and decreased its protein level. miR‐30a inhibitor abrogated RUNX3‐mediated inhibition of cell invasion and downregulation of vimentin. Thus, RUNX3 suppressed gastric cancer cell invasion and vimentin expression by activating miR‐30a. In gastric cancer patients, levels of RUNX3 were positively correlated with miR‐30a and negatively associated with the levels of vimentin. Collectively, our data suggest a novel molecular mechanism for the tumour suppressor activity of RUNX3. Effective therapy targeting the RUNX3 pathway may help control gastric cancer cell invasion and metastasis by inhibiting the EMT.


PLOS ONE | 2013

Histone demethylase retinoblastoma binding protein 2 is overexpressed in hepatocellular carcinoma and negatively regulated by hsa-miR-212.

Xiuming Liang; Jiping Zeng; Lixiang Wang; Ming Fang; Qing Wang; Min Zhao; Xia Xu; Zhifang Liu; Wenjuan Li; Shili Liu; Han Yu; Jihui Jia; Chunyan Chen

Background The H3K4 demethylase retinoblastoma binding protein 2 (RBP2) is involved in the pathogenesis of gastric cancer, but its role and regulation in hepatocellular carcinoma (HCC) is unknown. We determined the function of RBP2 and its regulation in HCC in vitro and in human tissues. Methods We analyzed gene expression in 20 specimens each of human HCC and normal liver tissue by quantitative real-time PCR and immunohistochemistry. Proliferation was analyzed by foci formation and senescence by β-galactosidase staining. Promoter activity was detected by luciferase reporter assay. Results The expression of RBP2 was stronger in cancerous than non-cancerous tissues, but that of its binding microRNA, Homo sapiens miR-212 (hsa-miR-212), showed an opposite pattern. SiRNA knockdown of RBP2 significantly upregulated cyclin-dependent kinase inhibitors (CDKIs), with suppression of HCC cell proliferation and induction of senescence. Overexpression of hsa-miR-212 suppressed RBP2 expression, with inhibited cell proliferation and induced cellular senescence, which coincided with upregulated CDKIs; with low hsa-miR-212 expression, CDKIs were downregulated in HCC tissue. Inhibition of hsa-miR-212 expression upregulated RBP2 expression. Luciferase reporter assay detected the direct binding of hsa-miR-212 to the RBP2 3′ UTR. Conclusions RBP2 is overexpressed in HCC and negatively regulated by hsa-miR-212. The hsa-miR-212–RBP2–CDKI pathway may be important in the pathogenesis of HCC.


Biochemical and Biophysical Research Communications | 2011

Histone demethylase JMJD2B is required for tumor cell proliferation and survival and is overexpressed in gastric cancer

Wenjuan Li; Li Zhao; Wen Zang; Zhifang Liu; Long Chen; Tiantian Liu; Dawei Xu; Jihui Jia

Epigenetic alterations such as aberrant expression of histone-modifying enzymes have been implicated in tumorigenesis. Jumonji domain containing 2B (JMJD2B) is a newly identified histone demethylase that regulates chromatin structure or gene expression by removing methyl residues from trimethylated lysine 9 on histone H3. Recent observations have shown oncogenic activity of JMJD2B. We explored the functional role of JMJD2B in cancer cell proliferation, survival and tumorigenesis, and determined its expression profile in gastric cancer. Knocking down JMJD2B expression by small interfering RNA (siRNA) in gastric and other cancer cells inhibited cell proliferation and/or induced apoptosis and elevated the expression of p53 and p21(CIP1) proteins. The enhanced p53 expression resulted from activation of the DNA damage response pathway. JMJD2B knockdown markedly suppressed xenograft tumor growth in vivo in mice. Moreover, JMJD2B expression was increased in primary gastric-cancer tissues of humans. Thus, JMJD2B is required for sustained proliferation and survival of tumor cells in vitro and in vivo, and its aberrant expression may contribute to the pathogenesis of gastric cancer.


Journal of Medical Microbiology | 2010

Helicobacter pylori CagA upregulation of CIP2A is dependent on the Src and MEK/ERK pathways

Dapeng Zhao; Zhifang Liu; Jian Ding; Wenjuan Li; Yundong Sun; Han Yu; Yabin Zhou; Jiping Zeng; Chunyan Chen; Jihui Jia

Helicobacter pylori is classified as a class I carcinogenic factor and its persistent colonization in the stomach induces gastric cancer. Cancerous Inhibitor of PP2A (CIP2A) is a newly identified oncoprotein overexpressed in gastric cancer. Serving as a key oncoprotein, CIP2A also participates in regulation of senescence and proliferation of gastric cells. The combination of aberrant CIP2A expression inducing unlimited cell proliferation, and H. pylori infection eliciting aberrant expression of some key proteins, results in the onset of gastric tumorigenesis. However, the relationship between H. pylori infection and CIP2A expression still remains undefined. The aim of our study was to verify the effect of H. pylori infection on CIP2A expression levels and identify H. pylori signalling molecules and corresponding pathways influencing CIP2A expression. Following plasmid-mediated expression of CagA in human gastric cell lines, the cells were infected with H. pylori and CIP2A expression levels were examined by immunoblotting. Signal inhibitors were used to verify which signal pathways were involved. We also performed CIP2A depletion and H. pylori infection after depletion in AGS cells. H. pylori infection-induced CIP2A expression was dependent on cagA gene expression and CagA phosphorylation. Bacterial oncoprotein CagA upregulated CIP2A expression and this upregulation effect was dependent on Src and Ras/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase pathways. H. pylori infection-induced Myc stabilization was partially attenuated by CIP2A depletion. The results of our study provide further information for understanding the mechanism of H. pylori carcinogenesis.


Fems Immunology and Medical Microbiology | 2011

Upregulation of progranulin by Helicobacter pylori in human gastric epithelial cells via p38MAPK and MEK1/2 signaling pathway: role in epithelial cell proliferation and migration.

Hongyan Wang; Yundong Sun; Shili Liu; Han Yu; Wenjuan Li; Jiping Zeng; Chunyan Chen; Jihui Jia

Helicobacter pylori is a major human pathogen associated with gastric diseases such as chronic active gastritis, peptic ulcer, and gastric carcinoma. The growth factor progranulin (PGRN) is a secreted glycoprotein that functions as an important regulator of cell growth, migration, and transformation. We aimed to determine the molecular mechanisms by which H. pylori upregulates the expression of PGRN and the relationship between H. pylori infection and production of PGRN in controlling cell proliferation and migration. Levels of PGRN were examined in gastric tissues from patients and in vitro in gastric epithelial cells. Cell proliferation was measured by colony formation assay. Cell migration was monitored by wound healing migration assay. PGRN protein levels were increased in patients with gastritis and gastric cancer tissue. Infection of gastric epithelial cells with H. pylori significantly increased PGRN expression in a time-dependent manner. Blockade of the p38 and MEK1/2 pathway by inhibitor inhibited H. pylori-mediated PGRN upregulation. Activation of p38 and MEK1/2 pathway by H. pylori was also identified. Knockdown of PGRN attenuated the H. pylori-induced proliferative activity and migration of cancer cells. These findings suggest that the upregulation of PGRN in H. pylori-infected gastric epithelial cells may contribute to the carcinogenic process.


PLOS ONE | 2014

Helicobacter pylori promotes epithelial-mesenchymal transition in gastric cancer by downregulating programmed cell death protein 4 (PDCD4).

Han Yu; Jiping Zeng; Xiuming Liang; Wenfu Wang; Yabin Zhou; Yundong Sun; Shili Liu; Wenjuan Li; Chunyan Chen; Jihui Jia

Helicobacter pylori, a Gram-negative, microaerophilic bacterium found in the stomach, is assumed to be associated with carcinogenesis, invasion and metastasis in digestive diseases. Cytotoxin-associated gene A (CagA) is an oncogenic protein of H. pylori that is encoded by a Cag pathogenicity island related to the development of gastric cancer. The epithelial–mesenchymal transition (EMT) is the main biological event in invasion or metastasis of epithelial cells. H. pylori may promote EMT in human gastric cancer cell lines, but the specific mechanisms are still obscure. We explored the underlying molecular mechanism of EMT induced by H. pylori CagA in gastric cancer. In our article, we detected gastric cancer specimens and adjacent non-cancerous specimens by immunohistochemistry and found increased expression of the EMT-related regulatory protein TWIST1 and the mesenchymal marker vimentin in cancer tissues, while programmed cell death factor 4 (PDCD4) and the epithelial marker E-cadherin expression decreased in cancer specimens. These changes were associated with degree of tissue malignancy. In addition, PDCD4 and TWIST1 levels were related. In gastric cancer cells cocultured with CagA expression plasmid, CagA activated TWIST1 and vimentin expression, and inhibited E-cadherin expression by downregulating PDCD4. CagA also promoted mobility of gastric cancer cells by regulating PDCD4. Thus, H. pylori CagA induced EMT in gastric cancer cells, which reveals a new signaling pathway of EMT in gastric cancer cell lines.


Journal of Cellular Biochemistry | 2012

Helicobacter pylori CagA inhibits the expression of Runx3 via Src/MEK/ERK and p38 MAPK pathways in gastric epithelial cell

Zhifang Liu; Xia Xu; Long Chen; Wenjuan Li; Yundong Sun; Jiping Zeng; Han Yu; Chunyan Chen; Jihui Jia

Infection with CagA‐positive Helicobacter pylori is the strongest risk factor for gastric carcinoma. Upon delivery into gastric epithelial cells, CagA disturbs cellular functions by physically interacting with and deregulating intracellular signaling molecules via both tyrosine phosphorylation‐dependent and ‐independent mechanisms. Runx3 was suggested to be a tumor suppressor and closely associated with tumorigenesis and progression of gastric cancer. The aim of our study is to verify the effect of H. pylori virulence factor CagA on Runx3 expression level and investigate the corresponding molecular mechanisms and signaling pathways influencing Runx3 expression. Human gastric epithelial immortalized GES‐1 cells were transfected with CagA‐expression vector or control vector with FuGENE HD transfection reagent. Runx3 expression levels were determined by QRT‐PCR and immunoblotting. Then we constructed a 1,150u2009bp Runx3 promoter luciferase reporter plasmid, pGL3‐1150u2009bp, which was co‐transfected into GES‐1 cell with CagA‐expression vector or control vector. Luciferase reporter assay was used to determine the effects of CagA on the 1,150u2009bp promoter activity of Runx3. Signal inhibitors were used to detect the signal pathway(s) through which CagA affects Runx3. Our results showed that CagA can reduce the expression level of Runx3 at both mRNA and protein levels significantly. Importantly, the 1,150u2009bp Runx3 promoter activity was decreased in cells transfected with CagA‐expression vector comparing with cells transfected with control vector. And this inhibition is dependent on the phosphorylation of CagA. Signal pathways Src/MEK/ERK and p38 MAPK are involved in this regulation. Our findings provide new insights for understanding the mechanism of H. pylori carcinogenesis. J. Cell. Biochem. 113: 1080–1086, 2012.


Fems Microbiology Letters | 2010

The antibacterial mode of action of allitridi for its potential use as a therapeutic agent against Helicobacter pylori infection

Shuang Liu; Yundong Sun; Wenjuan Li; Han Yu; Xi Li; Zhifang Liu; Jiping Zeng; Yabin Zhou; Chunyan Chen; Jihui Jia

Eradication of Helicobacter pylori with traditional therapy often fails in clinical treatment. As a result, a novel efficacious therapeutic agent is strongly needed. Allitridi, a proprietary garlic derivative, has been successfully used to treat both systemic fungal and bacterial infections in China. Our previous study has shown a dose-dependent inhibitory effect of allitridi on H. pylori growth. However, the antibacterial mode of action of allitridi is still unclear. Proteomic analysis was used to study the global protein alterations induced by allitridi. A total of 21 protein spots were identified to be differentially expressed. Our results indicated that the bacteriostatic mechanism of allitridi in H. pylori can be attributed to its multitarget inhibitory effects in energy metabolism and biosynthesis including amino acid biosynthesis, protein synthesis, mRNA synthesis and fatty acid biosynthesis. Allitridi can also disturb the expression of antioxidant proteins and decrease the production of virulence factors. Western blot analysis showed that allitridi at subinhibitory concentrations can potently suppress the production of CagA and VacA. Our investigations on the antibacterial mode of action of allitridi provide an insight into the potential use of allitridi as a therapeutic agent against H. pylori infection.

Collaboration


Dive into the Wenjuan Li's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Han Yu

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yundong Sun

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Shili Liu

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lixiang Wang

Chinese Ministry of Education

View shared research outputs
Top Co-Authors

Avatar

Li Shen

Chinese Ministry of Education

View shared research outputs
Researchain Logo
Decentralizing Knowledge