Oncotarget

Research Papers:

Colon cancer cell invasion is promoted by protein kinase CK2 through increase of endothelin-converting enzyme-1c protein stability

Ignacio Niechi _, Eduardo Silva, Pablo Cabello, Hernan Huerta, Valentina Carrasco, Paulina Villar, Luis Rodrigo Cataldo, Katherine Marcelain, Ricardo Armisen, Manuel Varas-Godoy, Cristina Fernandez and Julio C. Tapia

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2015; 6:42749-42760. https://doi.org/10.18632/oncotarget.5722

Metrics: PDF 2362 views  |   HTML 2396 views  |   ?  


Abstract

Ignacio Niechi1, Eduardo Silva1, Pablo Cabello1, Hernan Huerta1, Valentina Carrasco1, Paulina Villar1, Luis Rodrigo Cataldo1, Katherine Marcelain2, Ricardo Armisen2, Manuel Varas-Godoy3, Cristina Fernandez4, Julio C. Tapia1,2

1Cell Transformation Laboratory, Program of Cellular and Molecular Biology, ICBM, Faculty of Medicine, University of Chile, Santiago, Chile

2ICBM, Faculty of Medicine, University of Chile, Santiago, Chile

3Fundacion Ciencia y Vida, Santiago, Chile

4Department of Anatomopathology, HCUCH, Faculty of Medicine, University of Chile, Santiago, Chile

Correspondence to:

Julio C. Tapia, e-mail: [email protected]

Keywords: CK2, ECE-1, endothelin, colon cancer, metastasis

Received: February 08, 2015     Accepted: October 06, 2015     Published: October 16, 2015

ABSTRACT

Endothelin-converting enzyme-1c (ECE-1c) is a membrane metalloprotease involved in endothelin-1 synthesis, which has been shown in vitro to have a role in breast, ovary and prostate cancer cell invasion. N-terminal end of ECE-1c displays three putative phosphorylation sites for the protein kinase CK2. We studied whether CK2 phosphorylates N-terminal end of ECE-1c as well as whether this has a role in migration and invasion of colon cancer cells. CK2 phosphorylated the N-terminal end of ECE-1c and this was precluded upon inhibition of CK2. Inhibition also led to diminished protein levels of both endogen ECE-1 or GFP-fused N-terminal end of ECE-1c in 293T embryonic and DLD-1 colon cancer cells, which highlighted the importance of this motif on UPS-dependent ECE-1c degradation. Full-length ECE-1c mutants designed either to mimic or abrogate CK2-phosphorylation displayed increased or decreased migration/invasion of colon cancer cells, respectively. Moreover, ECE-1c overexpression or its silencing with a siRNA led to increased or diminished cell migration/invasion, respectively. Altogether, these data show that CK2-increased ECE-1c protein stability is related to augmented migration and invasion of colon cancer cells, shedding light on a novel mechanism by which CK2 may promote malignant progression of this disease.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 5722