Oncotarget

Research Papers:

FBXW7 suppresses epithelial-mesenchymal transition, stemness and metastatic potential of cholangiocarcinoma cells

Hui Yang _, Xiaofei Lu, Ziming Liu, Lili Chen, Yunfei Xu, Yuli Wang, Guangwei Wei and Yuxin Chen

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:6310-6325. https://doi.org/10.18632/oncotarget.3355

Metrics: PDF 3150 views  |   HTML 3107 views  |   ?  


Abstract

Hui Yang1, Xiaofei Lu1,2, Ziming Liu3, Lili Chen4, Yunfei Xu1, Yuli Wang5, Guangwei Wei5 and Yuxin Chen1

1 Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan, China

2 Department of General Surgery, Jinan Central Hospital of Shandong University, Jinan, China

3 Department of Emergency Medicine, Jinan Fifth People’s Hospital, Jinan, China

4 Department of Pathology, Jinan Fourth People’s Hospital, Jinan, China

5 Department of Anatomy and Key Laboratory of Experimental Teratology, Ministry of Education, Shandong University School of Medicine, Jinan, China

Correspondence:

Yuxin Chen, email:

Guangwei Wei, email:

Keywords: ubiquitin ligase, tumor suppressor, mTOR, ZEB1, metastasis

Received: December 11, 2014 Accepted: January 17, 2015 Published: January 31, 2015

Abstract

Epithelial-mesenchymal transition (EMT) plays a fundamental role in cancer metastasis. The ubiquitin ligase FBXW7, a general tumor suppressor in human cancer, has been implicated in diverse cellular processes, however, its role in cholangiocarcinoma (CCA) metastasis has not been identified. Here, we report a crucial role of FBXW7 in CCA metastasis by regulating EMT. Loss of FBXW7 expression was detected in CCA cells and clinical specimens. Clinicopathological analysis revealed a close correlation between FBXW7 deficiency and metastasis, TNM stage and differentiation in intrahepatic CCA and perihilar CCA. Moreover, FBXW7 silencing in CCA cells dramatically promoted EMT, stem-like capacity and metastasis both in vitro and in vivo. Conversely, FBXW7 overexpression attenuated these processes. Mechanistically, treatment with rapamycin, a mTOR inhibitor, inhibited EMT, stem-like capacity and metastasis induced by FBXW7 silencing both in vitro and in vivo. Furthermore, the expression of EMT regulating transcription factors, snail, slug and ZEB1, were also decreased markedly with rapamycin treatment. In addition, silencing ZEB1 inhibited EMT and metastasis of both CCA cells and FBXW7 deficient CCA cells, which implicated the potential role of ZEB1 in FBXW7/mTOR signaling pathway related CCA metastasis. In conclusion, our findings defined a pivotal function of FBXW7 in CCA metastasis by regulating EMT.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 3355