Oncotarget

Research Papers:

Overexpression of N-terminal kinase like gene promotes tumorigenicity of hepatocellular carcinoma by regulating cell cycle progression and cell motility

Jian Wang _, Ming Liu, Leilei Chen, Tim Hon Man Chan, Lingxi Jiang, Yun-Fei Yuan and Xin-Yuan Guan

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:1618-1630. https://doi.org/10.18632/oncotarget.2730

Metrics: PDF 2219 views  |   HTML 2328 views  |   ?  


Abstract

Jian Wang1,2,3, Ming Liu1,2,3, Leilei Chen1,2, Tim Hon Man Chan1,2, Lingxi Jiang1,2, Yun-Fei Yuan4, Xin-Yuan Guan1,2,3,4

1Departments of Clinical Oncology, The University of Hong Kong, Hong Kong, China

2State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China

3Center for Cancer Research, The University of Hong Kong, Hong Kong, China

4State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China

Correspondence to:

Xin-Yuan Guan, e-mail: [email protected]

Keywords: HCC, NTKL, CHD1L, dynamin2, metastasis

Received: September 02, 2014     Accepted: November 08, 2014     Published: January 10, 2015

ABSTRACT

Amplification and overexpression of CHD1L is one of the most frequent genetic alterations in hepatocellular carcinoma (HCC). Here we found that one of CHD1L downstream targets, NTKL, was frequently upregulated in HCC, which was significantly correlated with vascular invasion (P = 0.012) and poor prognosis (P = 0.050) of HCC. ChIP assay demonstrated the binding of CHD1L to the promoter region of NTKL. QRT-PCR study showed that the expression of NTKL positively correlated with CHD1L expression in both clinical samples and cell lines. Functional study found that NTKL had strong oncogenic roles, including increased cell growth, colony formation in soft agar, and tumor formation in nude mice. Further study found that NTKL could promote G1/S transition by decreasing P53 and increasing CyclinD1 expressions. NTKL overexpression could accelerate the mitotic exit and chromosome segregation, which led to the cytokinesis failure and subsequently induced apoptosis. NTKL also regulated cell motility by facilitating philopodia and lamellipodia formation through regulating F-actin reorganization and the phosphorylation of small GTPase Rac1/cdc42. Using co-IP and mass spectrometry approach, we identified the large GTPase dynamin2 as an interacting protein of NTKL, which might be responsible for the phenotype alterations caused by NTKL overexpression, such as cytokinesis failure, increased cell motility and abnormal of cell division.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 2730