Oncotarget

Research Papers:

Role of the polypeptide N-acetylgalactosaminyltransferase 3 in ovarian cancer progression: possible implications in abnormal mucin O-glycosylation

Zhi-Qiang Wang, Magdalena Bachvarova, Chantale Morin, Marie Plante, Jean Gregoire, Marie-Claude Renaud, Alexandra Sebastianelli and Dimcho Bachvarov _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2014; 5:544-560. https://doi.org/10.18632/oncotarget.1652

Metrics: PDF 3487 views  |   HTML 5853 views  |   ?  


Abstract

Zhi-Qiang Wang1,2, Magdalena Bachvarova2, Chantale Morin2, Marie Plante2,3, Jean Gregoire2,3, Marie-Claude Renaud2,3, Alexandra Sebastianelli2,3, and Dimcho Bachvarov1,2

1 Department of Molecular Medicine, Laval University, Québec (Québec), Canada

2 Centre de recherche du CHU de Québec, L’Hôtel-Dieu de Québec, Québec (Québec), Canada

3 Department of Obstetrics and Gynecology, Laval University, Québec (Québec), Canada

Correspondence:

Dimcho Bachvarov, email:

Keywords: GALNT3, epithelial ovarian cancer; DNA hypomethylation, progression-free survival, microarrays, mucin O-glycosylation

Received: December 3, 2013 Accepted: January 25, 2014 Published: January 27, 2014

Abstract

Previously, we have identified the polypeptide N-acetylgalactosaminyltransferase 3 (GALNT3) gene as notably hypomethylated in low-malignant potential (LMP) and high-grade (HG) serous epithelial ovarian tumors, compared to normal ovarian tissues. Here we show that GALNT3 is strongly overexpressed in HG serous EOC tumors as compared to normal ovarian tissue. Moreover, the GALNT3 expression significantly correlated with shorter progression-free survival (PFS) intervals in epithelial ovarian cancer (EOC) patients with advanced disease.

Knockdown of the GALNT3 expression in EOC cells led to sharp decrease of cell proliferation and induced S-phase cell cycle arrest. Additionally, GALNT3 suppression significantly inhibited EOC cell migration and invasion. Gene expression profiling and consecutive network and pathway analyses confirmed these findings, as numerous genes and pathways known previously to be implicated in ovarian tumorigenesis, including EOC tumor invasion and metastasis, were found to be downregulated upon GALNT3 suppression, while some tumor suppressor genes were induced. Moreover, GALNT3 downregulation was associated with reduced MUC1 protein expression in EOC cells, probably related to destabilization of the MUC1 protein due to lack of GALNT3 glycosylation activity. GALNT3 knockdown was also accompanied with increase of the cell adhesion molecules β-catenin and E-cadherin, which are normally suppressed by MUC1 in cancer, thus supporting the role of the GALNT3-MUC1 axis in EOC invasion.

Taken together, our data are indicative for a strong oncogenic potential of the GALNT3 gene in advanced EOC and identify this transferase as a novel EOC biomarker and putative EOC therapeutic target. Our findings also suggest that GALNT3 overexpression might contribute to EOC progression through aberrant mucin O-glycosylation


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 1652