Oncotarget

Research Papers:

Missing-in-metastasis B (MIM-B) combined with caveolin-1 promotes metastasis of hepatocellular carcinoma

Xiu-Yan Huang _, Zi-Li Huang, Tao Niu, Zhen-Qian Wu, Bin Xu, Yong-Hua Xu, Xin-Yu Huang, Qi Zheng, Jian Zhou, Zi Chen and Zhao-You Tang

PDF  |  HTML  |  How to cite

Oncotarget. 2017; 8:95450-95465. https://doi.org/10.18632/oncotarget.20735

Metrics: PDF 1494 views  |   HTML 2930 views  |   ?  


Abstract

Xiu-Yan Huang1,*, Zi-Li Huang2,*, Tao Niu3,*, Zhen-Qian Wu1,*, Bin Xu4,*, Yong-Hua Xu2, Xin-Yu Huang1, Qi Zheng1, Jian Zhou5, Zi Chen6 and Zhao-You Tang5

1Department of General Surgery, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China

2Department of Radiology, Xuhui Central Hospital, Shanghai, P.R. China

3Department of General Surgery, People’s Hospital of Menghai County, Yunnan Province, P.R. China

4Department of General Surgery, The Tenth People’s Hospital of Tongji University, Shanghai, P.R. China

5Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, P.R. China

6Thayer School of Engineering, Norris Cotton Cancer Center, Dartmouth College, Hanover, NH, USA

*These authors have contributed equally to this work

Correspondence to:

Xiu-Yan Huang, email: [email protected]

Keywords: hepatocellular carcinoma; missing in metastasis B; caveolin-1; epidermal growth factor receptor; metastasis

Received: December 14, 2016     Accepted: August 04, 2017     Published: September 08, 2017

ABSTRACT

Background: Increasing amounts of evidence indicate that Missing in metastasis B (MIM-B) promotes cancer metastasis. Here, we sought to better understand the mechanism through which MIM-B promotes tumor metastasis in hepatocellular carcinoma (HCC).

Methods: We performed confocal microscopy analysis to determine the distributions of MIM-B and caveolin-1 and conducted co-immunoprecipitation assays to detect the interactions between MIM-B and caveolin-1 in vitro. We performed transwell assays to analyze the invasive ability of HCC cells. Changes in the expression levels of key genes and some molecular makers were detected by immunohistochemistry and western blotting in HCC tissue samples.

Results: We found that MIM-B co-localizes with caveolin-1 and demonstrated that MIM-B and caveolin-1 interact in vitro. Repressing MIM-B and caveolin-1 expression inhibited the epidermal growth factor receptor signaling pathway. We overexpressed MIM-B and caveolin-1 in Hep3B cells, which enhanced Hep3B cell invasiveness. Furthermore, MHCC97H cell invasiveness was significantly decreased in cells in which MIM-B and caveolin-1 expression was inhibited. Additionally, we found that MIM-B and caveolin-1 were expressed at higher levels in HCC tissues than in paired normal tissues. Moreover, HCC patients with MIM-B and caveolin-1 up-regulation experienced significantly worse outcomes than controls (P < 0.001), and HCC patients with high MIM-B and caveolin-1 expression levels often developed pulmonary metastasis (P < 0.001).

Conclusions: MIM-B combined with caveolin-1 promotes metastasis of HCC, and elevated MIM-B and caveolin-1 expression levels are associated with a poor prognosis in HCC patients; therefore, MIM-B and caveolin-1 may represent novel targets for the diagnosis and treatment of HCC.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 20735