Abstract

Chemical biology studies, exemplified by metabolic glycoengineering experiments that employ short chain fatty acid (SCFA)-hexosamine monosaccharide hybrid molecules, often suffer from off-target effects. Here we demonstrate that systematic structure-activity relationship (SAR) studies can deconvolute multiple biological activities of SCFA-hexosamine analogues by demonstrating that triacylated monosaccharides, including both n-butyrate- and acetate-modified ManNAc analogues, had dramatically different activities depending on whether the free hydroxyl group was at the C1 or C6 position. The C1-OH (hemiacetal) analogues enhanced growth inhibition in MDA-MB-231 human breast cancer cells and suppressed expression of MUC1, which are attractive properties for an anticancer agent. By contrast, C6-OH analogues supported high metabolic flux into the sialic acid pathway with negligible growth inhibition or toxicity, which are desirable properties for glycan labeling in healthy cells. Importantly, these SAR were general, applying to other hexosamines ( e.g., GlcNAc) and non-natural sugar "scaffolds" ( e.g., ManNLev). From a practical standpoint, the ability to separate toxicity from flux will facilitate the use of MOE analogues for cancer treatment and glycomics applications, respectively. Mechanistically, these findings overturn the premise that the bioactivities of SCFA-monosaccharide hybrid molecules result from their hydrolysis products ( e.g., n-butyrate, which acts as a histone deacetylase inhibitor, and ManNAc, which activates sialic acid biosynthesis); instead the SAR establish that inherent properties of partially acylated hexosamines supersede the cellular responses supported by either the acyl or monosaccharide moieties.

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