Abstract

Galectin-3 binding to cell surface glycoproteins, including branched N-glycans generated by N-acetylglucosaminyltransferase V (Mgat5) activity, forms a multivalent, heterogeneous, and dynamic lattice. This lattice has been shown to regulate integrin and receptor tyrosine kinase signaling promoting tumor cell migration. N-cadherin is a homotypic cell-cell adhesion receptor commonly overexpressed in tumor cells that contributes to cell motility. Here we show that galectin-3 and N-cadherin interact and colocalize with the lipid raft marker GM1 ganglioside in cell-cell junctions of mammary epithelial cancer cells. Disruption of the lattice by deletion of Mgat5, siRNA depletion of galectin-3, or competitive inhibition with lactose stabilizes cell-cell junctions. It also reduces, in a p120-catenin-dependent manner, the dynamic pool of junctional N-cadherin. Proteomic analysis of detergent-resistant membranes (DRMs) revealed that the galectin lattice opposes entry of many proteins into DRM rafts. N-cadherin and catenins are present in DRMs; however, their DRM distribution is not significantly affected by lattice disruption. Galectin lattice integrity increases the mobile fraction of the raft marker, GM1 ganglioside binding cholera toxin B subunit Ctb, at cell-cell contacts in a p120-catenin-independent manner, but does not affect the mobility of either Ctb-labeled GM1 or GFP-coupled N-cadherin in nonjunctional regions. Our results suggest that the galectin lattice independently enhances lateral molecular diffusion by direct interaction with specific glycoconjugates within the adherens junction. By promoting exchange between raft and non-raft microdomains as well as molecular dynamics within junction-specific raft microdomains, the lattice may enhance turnover of N-cadherin and other glycoconjugates that determine junctional stability and rates of cell migration.

Highlights

  • Galectin-3-N-glycan binding forms a lattice that regulates cancer cell adhesion, migration, and signaling

  • Interaction of Gal-3 and N-cadherin at Cell-Cell Junctions—Using cell lines derived from epithelial mammary tumors from Mgat5ϩ/ϩ and Mgat5Ϫ/Ϫ transgenic mice on a polyoma middle T (PyMT) background, we have previously shown that Mgat5 and the galectin lattice regulate EGFR signaling and focal adhesion dynamics (8 –10, 16)

  • N-cadherin has been localized in rafts [33], and we observed the localization of cholera toxin B subunit (Ctb)-FITC to cell-cell junctions, indicating an enrichment of GM1 ganglioside in these domains

Read more

Summary

Introduction

Galectin-3-N-glycan binding forms a lattice that regulates cancer cell adhesion, migration, and signaling. Galectin-3 binding to cell surface glycoproteins, including branched N-glycans generated by N-acetylglucosaminyltransferase V (Mgat5) activity, forms a multivalent, heterogeneous, and dynamic lattice. This lattice has been shown to regulate integrin and receptor tyrosine kinase signaling promoting tumor cell migration. Disruption of the lattice by deletion of Mgat, siRNA depletion of galectin-3, or competitive inhibition with lactose stabilizes cell-cell junctions. It reduces, in a p120catenin-dependent manner, the dynamic pool of junctional N-cadherin. Our results suggest that the galectin lattice independently enhances lateral molecular diffusion by

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call