Oncotarget

Research Papers:

ZNF350 promoter methylation accelerates colon cancer cell migration

Hiroki Tanaka, Yuki Kuwano, Tatsuya Nishikawa, Kazuhito Rokutan and Kensei Nishida _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2018; 9:36750-36769. https://doi.org/10.18632/oncotarget.26353

Metrics: PDF 1383 views  |   HTML 2265 views  |   ?  


Abstract

Hiroki Tanaka1, Yuki Kuwano1, Tatsuya Nishikawa1, Kazuhito Rokutan1 and Kensei Nishida1

1Department of Pathophysiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770-8503, Japan

Correspondence to:

Kensei Nishida, email: [email protected]

Keywords: DNA methylation; EMT; cell migration; ZNF350; colon cancer cells

Received: May 25, 2018    Accepted: October 24, 2018    Published: December 04, 2018

ABSTRACT

Diversification of transcriptomic and epigenomic states may occur during the expansion of colorectal cancers. Certain cancer cells lose their epithelial characters and gain mesenchymal properties, known as epithelial-mesenchymal transition (EMT), and they aggressively migrate into the non-tumorigenic extracellular matrix. In this study, we isolated a subpopulation with accelerated baseline motility (MG cells) and an immotile one (non-MG cells) from a colon cancer cell line (HCT116). Gene expression signatures of the MG cells indicated that this subpopulation was likely an EMT hybrid. The MG cells substantially lost their migratory properties after treatment with a methyltransferase inhibitor, 5-azacytidine, suggesting a role of DNA methylation in this process. Global transcriptome assays of both types of cells with or without 5-azacytidine treatment identified 640 genes, whose expression might be methylation-dependently down-regulated in the MG cells. Global methylation analysis revealed that 35 out of the 640 genes were hyper-methylated in the MG cells. Among them, we focused on the anti-oncogene ZNF350, which encodes a zinc-finger and BRCA1-interacting protein. Notably, ZNF350 knockdown accelerated migration of the non-MG cells, while overexpression of ZNF350 in the MG cells significantly impaired their migration. Finally, pyrosequence analysis together with dual luciferase assays of serially truncated fragments of the ZNF350 promoter (-268 to +49 bp) indicated that three hyper-methylated sites were possibly responsible for the basal promoter activity of ZNF350. Taken together, our results suggest that hyper-methylation of the ZNF350 proximal promoter may be one of the crucial determinants for acquiring increased migratory capabilities in colon cancer cells.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 26353