Oncotarget

Research Papers:

Metabolic flux-driven sialylation alters internalization, recycling, and drug sensitivity of the epidermal growth factor receptor (EGFR) in SW1990 pancreatic cancer cells

Mohit P. Mathew, Elaine Tan, Christopher T. Saeui, Patawut Bovonratwet, Samuel Sklar, Rahul Bhattacharya and Kevin J. Yarema _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2016; 7:66491-66511. https://doi.org/10.18632/oncotarget.11582

Metrics: PDF 2009 views  |   HTML 3263 views  |   ?  


Abstract

Mohit P. Mathew1, Elaine Tan1, Christopher T. Saeui1, Patawut Bovonratwet2, Samuel Sklar1, Rahul Bhattacharya1 and Kevin J. Yarema1

1 Department of Biomedical Engineering and the Translational Tissue Engineering Center, The Johns Hopkins University, Baltimore, Maryland, USA

2 Current address: Yale School of Medicine, New Haven, Connecticut, USA

Correspondence to:

Kevin J. Yarema, email:

Keywords: pancreatic cancer, sialic acid, galectin, epidermal growth factor receptor (EGFR), glycobiology

Received: June 22, 2016 Accepted: August 01, 2016 Published: August 24, 2016

Abstract

In prior work we reported that advanced stage, drug-resistant pancreatic cancer cells (the SW1990 line) can be sensitized to the EGFR-targeting tyrosine kinase inhibitors (TKIs) erlotinib and gefitinib by treatment with 1,3,4-O-Bu3ManNAc (Bioorg. Med. Chem. Lett. (2015) 25(6):1223-7). Here we provide mechanistic insights into how this compound inhibits EGFR activity and provides synergy with TKI drugs. First, we showed that the sialylation of the EGFR receptor was at most only modestly enhanced (by ~20 to 30%) compared to overall ~2-fold increase in cell surface levels of this sugar. Second, flux-driven sialylation did not alter EGFR dimerization as has been reported for cancer cell lines that experience increased sialylation due to spontaneous mutations. Instead, we present evidence that 1,3,4-O-Bu3ManNAc treatment weakens the galectin lattice, increases the internalization of EGFR, and shifts endosomal trafficking towards non-clathrin mediated (NCM) endocytosis. Finally, by evaluating downstream targets of EGFR signaling, we linked synergy between 1,3,4-O-Bu3ManNAc and existing TKI drugs to a shift from clathrin-coated endocytosis (which allows EGFR signaling to continue after internalization) towards NCM endocytosis, which targets internalized moieties for degradation and thereby rapidly diminishes signaling.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 11582