Network


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

Hotspot


Dive into the research topics where Zhujun Tan is active.

Publication


Featured researches published by Zhujun Tan.


Nature Genetics | 2014

Whole-exome and targeted gene sequencing of gallbladder carcinoma identifies recurrent mutations in the ErbB pathway

Maolan Li; Zhou Zhang; Xiaoguang Li; Junyi Ye; Xiangsong Wu; Zhujun Tan; Chang Liu; Baiyong Shen; Xu-An Wang; Wen-Guang Wu; Daizhan Zhou; Di Zhang; Ting Wang; Bingya Liu; Kai Qu; Qichen Ding; Hao Weng; Qian Ding; Jiasheng Mu; Yijun Shu; Run-Fa Bao; Yang Cao; Peizhan Chen; Tian-Yu Liu; Lin Jiang; Yunping Hu; Ping Dong; Jun Gu; Wei Lu; Weibin Shi

Individuals with gallbladder carcinoma (GBC), the most aggressive malignancy of the biliary tract, have a poor prognosis. Here we report the identification of somatic mutations for GBC in 57 tumor-normal pairs through a combination of exome sequencing and ultra-deep sequencing of cancer-related genes. The mutation pattern is defined by a dominant prevalence of C>T mutations at TCN sites. Genes with a significant frequency (false discovery rate (FDR) < 0.05) of non-silent mutations include TP53 (47.1%), KRAS (7.8%) and ERBB3 (11.8%). Moreover, ErbB signaling (including EGFR, ERBB2, ERBB3, ERBB4 and their downstream genes) is the most extensively mutated pathway, affecting 36.8% (21/57) of the GBC samples. Multivariate analyses further show that cases with ErbB pathway mutations have a worse outcome (P = 0.001). These findings provide insight into the somatic mutational landscape in GBC and highlight the key role of the ErbB signaling pathway in GBC pathogenesis.


Cancer Biology & Therapy | 2014

MALAT1 promotes the proliferation and metastasis of gallbladder cancer cells by activating the ERK/MAPK pathway.

Xiangsong Wu; Xu-An Wang; Wen-Guang Wu; Yunping Hu; Maolan Li; Qian Ding; Hao Weng; Yijun Shu; Tian-Yu Liu; Lin Jiang; Yang Cao; Run-Fa Bao; Jiasheng Mu; Zhujun Tan; Feng Tao; Yingbin Liu

Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), a long non-coding RNA (lncRNA), is associated with metastasis and is an independent prognostic factor for lung cancer. Recent studies have demonstrated that MALAT1 plays an important role in other malignancies. However, little is known about the role of MALAT1 in gallbladder carcinoma (GBC), which is the most common cancer of the biliary tract and has an extremely poor prognosis. In this study, we focused on the expression, biological functions and mechanism of MALAT1 in GBC and found that MALAT1 was significantly upregulated in GBC tissues compared with corresponding non-cancerous tissues. Knockdown of MALAT1 in GBC cell lines using lentivirus-mediated RNA interference significantly inhibited the proliferation and metastasis of the GBC cells both in vitro and in vivo. Furthermore, ERK/MAPK pathway was found to be inactivated in the GBC cell lines after MALAT1 knockdown. These results indicated that MALAT1 might serve as an oncogenic lncRNA that promotes proliferation and metastasis of GBC and activates the ERK/MAPK pathway


Molecular and Cellular Biochemistry | 2012

NLK is a key regulator of proliferation and migration in gallbladder carcinoma cells

Zhujun Tan; Maolan Li; Wen-Guang Wu; Lin Zhang; Qichen Ding; Xiangsong Wu; Jiasheng Mu; Yingbin Liu

Gallbladder cancer (GBC) is one of the most lethal neoplasm and is the fifth most common malignancy of gastrointestinal tract. The prognosis of gallbladder cancer is extremely terrible partially due to metastasis. Thus, understanding the molecular pathways controlling metastasis of this lethal disease may provide new targets for targeted therapeutic approach. In this study, we investigated the function of nemo-like kinase (NLK) in GBC growth and migration. Lentivirus-mediated siRNA was employed to alleviate the expression level of NLK in GBC cell lines (GBC-SD and SGC-996). Real-time PCR and western-blot analysis demonstrated that both mRNA and protein levels of NLK in GBC-SD and SGC-996 cells were decreased after infection with NLK-siRNA-expressing lentivirus (Lv-shNLK). The proliferation and in vitro tumorigenesis (colony formation) ability as well as migration of GBC-SD and SGC-996 cells with low NLK expression decreased significantly. Our results suggested that NLK is a key regulator involved in proliferation and migration of GBC, and it could be used as a potential therapeutic target for GBC.


Cancer Cell International | 2013

Curcumin induces apoptosis in gallbladder carcinoma cell line GBC-SD cells

Tian-Yu Liu; Zhujun Tan; Lin Jiang; Jianfeng Gu; Xiangsong Wu; Yang Cao; Maolan Li; Ke-Jin Wu; Yingbin Liu

BackgroundGallbladder carcinoma is a malignant tumor with a very low 5-year survival rate because of the difficulty with its early diagnosis and the very poor prognosis of the advanced cancer state. The aims of this study were to determine whether curcumin could induce the apoptosis of a gallbladder carcinoma cell line, GBC-SD, and to clarify its related mechanism.MethodsFirst, the anti-proliferative activities of curcumin-treated and untreated GBC-SD cells were determined using the MTT and colony formation assays. Then, the early apoptosis of cells was detected by the annexin V/propidium iodide double-staining assay and Hoechst 33342 staining assay. Detection of mitochondrial membrane potential was used to validate the ability of curcumin on inducing apoptosis in GBC-SD cells. Cell cycle changes were detected by flow cytometric analysis. Finally, the expressions of the apoptosis-related proteins or genes caspase-3, PARP, Bcl-2, and Bax were analyzed by western blot and quantitative real time PCR assay. Statistical analyses were performed using the Student’s t-test for comparison of the results obtained from cells with or without curcumin treatment.ResultsThe MTT assay revealed that curcumin had induced a dose- and a time-dependent decrease in cell viability. Colony counting indicated that curcumin had induced a dose-dependent decrease in the colony formation ability in GBC-SD cells. Cells treated with curcumin were arrested at the S phase, according to the flow cytometric analysis. A significant induction of both the early and late phases of apoptosis was shown by the annexin V-FITC and PI staining. Morphological changes in apoptotic cells were also found by the Hoechst 33342 staining. After treatment with curcumin fluorescence shifted from red to green as ΔΨm decreased. Furthermore, western blot and quantitative real time PCR assays demonstrated that the curcumin induced apoptosis in GBC-SD cells by regulating the ratio of Bcl-2/Bax and activating the expression of cleaved caspase-3.ConclusionsTaken together, the results indicate that curcumin may be a potential agent for the treatment of gallbladder cancer.


Cancer Letters | 2014

Yes-associated protein 1 (YAP1) promotes human gallbladder tumor growth via activation of the AXL/MAPK pathway.

Maolan Li; Jianhua Lu; Fei Zhang; Huai-Feng Li; Bingtai Zhang; Xiangsong Wu; Zhujun Tan; Lin Zhang; Guofeng Gao; Jiasheng Mu; Yijun Shu; Run-Fa Bao; Qichen Ding; Wen-Guang Wu; Ping Dong; Jun Gu; Yingbin Liu

The transcriptional coactivator Yes-associated protein 1 (YAP1), a key regulator of cell proliferation and organ size in vertebrates, has been implicated in various malignancies. However, little is known about the expression and biological function of YAP1 in human gallbladder cancer (GBC). In this study we examined the clinical significance and biological functions of YAP1 in GBC and found that nuclear YAP1 and its target gene AXL were overexpressed in GBC tissues. We also observed a significant correlation between high YAP1 and AXL expression levels and worse prognosis. The depletion of YAP1 using lentivirus shRNAs significantly inhibited cell proliferation by inducing cell cycle arrest in S phase in concordance with the decrease of CDK2, CDC25A, and cyclin A, and resulted in increased cell apoptosis and invasive repression in GBC cell lines in vitro. Furthermore, knockdown of YAP1 also inhibited tumor growth in vivo. Additionally, we demonstrated that the activation of the AXL/MAPK pathway was involved in the oncogenic functions of YAP1 in GBC. These results demonstrated that YAP1 is a putative oncogene and represents a prognostic marker and potentially a novel therapeutic target for GBC.


Tumor Biology | 2013

Prognostic significance of nemo-like kinase (NLK) expression in patients with gallbladder cancer.

Maolan Li; Shenglai Zhang; Zhiqiang Wang; Bingtai Zhang; Xiangsong Wu; Hao Weng; Qian Ding; Zhujun Tan; Ning Zhang; Jiasheng Mu; Jiahua Yang; Yijun Shu; Run-Fa Bao; Qichen Ding; Wen-Guang Wu; Yang Cao; Yingbin Liu

Nemo-like kinase (NLK), a serine/threonine protein kinase, has been implicated in tumor development and progression, and plays an important role in diverse signaling pathways by phosphorylating a variety of transcription factors. Recent studies demonstrated that altered expression of NLK was observed in various types of human cancers. However, the clinical significance of NLK expression in gallbladder cancer (GBC) remains largely unknown. In this study, we focused on the clinical significance of NLK in GBC, and found that nuclear NLK protein overexpression was frequently detected in GBC tissues. The overexpression of NLK was significantly correlated with histological grade, TNM stage, and perineural invasion. The results of Kaplan–Meier analysis indicated that a high expression level of NLK resulted in a significantly poorer prognosis of GBC patients (P = 0.002). Furthermore, multivariate Cox regression analysis showed that high NLK expression was an independent prognostic factor for GBC patients (HR = 3.077). In conclusion, overexpression of NLK is closely related to progression of GBC, and NLK could be used as a potential prognostic marker for GBC patients.


Oncology Reports | 2014

Thioridazine, an antipsychotic drug, elicits potent antitumor effects in gastric cancer.

Jiasheng Mu; Haineng Xu; Yu Yang; Weidan Huang; Jing Xiao; Maolan Li; Zhujun Tan; Qichen Ding; Lin Zhang; Jianhua Lu; Xiangsong Wu; Yingbin Liu

Thioridazine, an antipsychotic drug, has been reported to induce apoptosis in various types of cancer cells, with specificity on targeting cancer stem cells (CSCs). However, whether it elicits anticancer effects in gastric cancer has never been reported. In the present study, we examined the ability of thioridazine to induce cell death in the gastric cancer cell lines NCI-N87 and AGS, and detected its in vivo tumor inhibition capacity. Thioridazine elicited cytotoxic effects on NCI-N87 and AGS cells in a dose-dependent manner, and inhibited the colony formation abilitiy of the NCI-N87 and AGS cells. Thioridazine treatment induced nuclear fragmentation, increased the proportion of sub-G1 phase cells, and elevated the percentage of Annexin V-positive cells, suggesting the occurrence of apoptosis. Moreover, thioridazine induced gastric cancer cell apoptosis in a caspase-dependent manner, as shown by a decrease in the precursors of casapse-9, caspase-8 and caspase-3, and by the ability of the caspase inhibitor Z-VAD-FMK to reverse the cytotoxic effect of thioridazine. JC-1 staining further revealed that thioridazine induced gastric cancer cell apoptosis via the mitochondrial pathway. In addition, thioridazine pretreatment inhibited the growth of NCI-N87 cell-derived tumors. The present study demonstrated that the antipsychotic drug thioridazine possesses anti-gastric cancer ability through in vitro and in vivo experiments, suggesting thioridazine as a potential drug in gastric cancer therapy.


Medical Oncology | 2013

Downregulated expression of hepatoma-derived growth factor (HDGF) reduces gallbladder cancer cell proliferation and invasion.

Maolan Li; Jun Shen; Xiangsong Wu; Bingtai Zhang; Rui Zhang; Hao Weng; Qian Ding; Zhujun Tan; Guofeng Gao; Jiasheng Mu; Jiahua Yang; Yijun Shu; Run-Fa Bao; Qichen Ding; Wen-Guang Wu; Yang Cao; Yingbin Liu

Hepatoma-derived growth factor (HDGF), a heparin-binding growth factor, has a wide range of biological functions, including mitogenic activity and vascular development. Recent studies demonstrated that HDGF also acted as an oncogene with aberrantly increased activity in multiple human cancers; however, little is known about the biological function of HDGF in gallbladder cancer (GBC). In this study, we focused on the clinical significance and biological functions of HDGF in GBC and found that Nuclear HDGF protein overexpression was frequently detected in GBC tissues. Patients with nuclear HDGF-positive tumors had worse overall survival than patients with HDGF-negative tumors. Furthermore, treatment of GBC lines with HDGF-targeting siRNA oligonucleotides (HDGF-siRNA) significantly reduced the proliferation of GBC-SD and SGC-996 cell lines and diminished both anchorage-independent growth on soft agar and cell migration. These data indicate that HDGF acts as a putative oncogene in GBC and could be a novel diagnostic and therapeutic target for GBC.


Cancer Cell International | 2014

Ursolic acid induces cell cycle arrest and apoptosis of gallbladder carcinoma cells

Hao Weng; Zhujun Tan; Yunping Hu; Yijun Shu; Run-Fa Bao; Lin Jiang; Xiangsong Wu; Maolan Li; Qian Ding; Xu-An Wang; Shan-Shan Xiang; Huai-Feng Li; Yang Cao; Feng Tao; Yingbin Liu

BackgroundUrsolic acid (UA), a plant extract used in traditional Chinese medicine, exhibits potential anticancer effects in various human cancer cell lines in vitro. In the present study, we evaluated the anti-tumoral properties of UA against gallbladder carcinoma and investigated the potential mechanisms responsible for its effects on proliferation, cell cycle arrest and apoptosis in vitro.MethodsThe anti-tumor activity of UA against GBC-SD and SGC-996 cells was assessed using MTT and colony formation assays. An annexin V/PI double-staining assay was used to detect cell apoptosis. Cell cycle changes were detected using flow cytometry. Rhodamine 123 staining was used to assess the mitochondrial membrane potential (ΔΨm) and validate UA’s ability to induce apoptosis in both cell lines. The effectiveness of UA in gallbladder cancer was further verified in vivo by establishing a xenograft GBC model in nude mice. Finally, the expression levels of cell cycle- and apoptosis-related proteins were analyzed by western blotting.ResultsOur results suggest that UA can significantly inhibit the growth of gallbladder cancer cells. MTT and colony formation assays indicated dose-dependent decreases in cell proliferation. S-phase arrest was observed in both cell lines after treatment with UA. Annexin V/PI staining suggested that UA induced both early and late phases of apoptosis. UA also decreased ΔΨm and altered the expression of molecules regulating the cell cycle and apoptosis. In vivo study showed intraperitoneally injection of UA can significantly inhibited the growth of xenograft tumor in nude mice and the inhibition efficiency is dose related. Activation of caspase-3,-9 and PARP indicated that mitochondrial pathways may be involved in UA-induced apoptosis.ConclusionsTaken together, these results suggest that UA exhibits significant anti-tumor effects by suppressing cell proliferation, promoting apoptosis and inducing 7cell cycle arrest both in vitro and in vivo. It may be a potential agent for treating gallbladder cancer.


Molecules | 2014

Cordycepin Induces S Phase Arrest and Apoptosis in Human Gallbladder Cancer Cells

Xu-An Wang; Shan-Shan Xiang; Huai-Feng Li; Xiangsong Wu; Maolan Li; Yijun Shu; Fei Zhang; Yang Cao; Yuan-Yuan Ye; Run-Fa Bao; Hao Weng; Wen-Guang Wu; Jiasheng Mu; Yunping Hu; Lin Jiang; Zhujun Tan; Wei Lu; Ping Wang; Yingbin Liu

Gallbladder cancer is the most common malignant tumor of the biliary tract, and this condition has a rather dismal prognosis, with an extremely low five-year survival rate. To improve the outcome of unresectable and recurrent gallbladder cancer, it is necessary to develop new effective treatments and drugs. The purpose of the present study was to evaluate the effects of cordycepin on human gallbladder cells and uncover the molecular mechanisms responsible for these effects. The Cell Counting Kit-8 (CCK-8) and colony formation assays revealed that cordycepin affected the viability and proliferation of human gallbladder cancer cells in a dose- and time-dependent manner. Flow cytometric analysis showed that cordycepin induced S phase arrest in human gallbladder cancer cell lines(NOZ and GBC-SD cells). Cordycepin-induced apoptosis was observed using an Annexin V/propidium iodide (PI) double-staining assay, and the mitochondrial membrane potential (ΔΨm) decreased in a dose-dependent manner. Additionally, western blot analysis revealed the upregulation of cleaved-caspase-3, cleaved-caspase-9, cleaved-PARP and Bax and the downregulation of Bcl-2, cyclin A and Cdk-2 in cordycepin-treated cells. Moreover, cordycepin inhibited tumor growth in nude mice bearing NOZ tumors. Our results indicate that this drug may represent an effective treatment for gallbladder carcinoma.

Collaboration


Dive into the Zhujun Tan's collaboration.

Top Co-Authors

Avatar

Maolan Li

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Xiangsong Wu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Yingbin Liu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Jiasheng Mu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Wen-Guang Wu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Hao Weng

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Run-Fa Bao

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Yijun Shu

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Qichen Ding

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Qian Ding

Shanghai Jiao Tong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge