Correlation Engine 2.0
Clear Search sequence regions


Sizes of these terms reflect their relevance to your search.

In immunoglobulin light-chain (LC) amyloidosis, transient unfolding or unfolding and proteolysis enable aggregation of LC proteins, causing potentially fatal organ damage. A drug that kinetically stabilizes LCs could suppress aggregation; however, LC sequences are variable and have no natural ligands, hindering drug development efforts. We previously identified high-throughput screening hits that bind to a site at the interface between the two variable domains of the LC homodimer. We hypothesized that extending the stabilizers beyond this initially characterized binding site would improve affinity. Here, using protease sensitivity assays, we identified stabilizers that can be divided into four substructures. Some stabilizers exhibit nanomolar EC50 values, a 3000-fold enhancement over the screening hits. Crystal structures reveal a key π-π stacking interaction with a conserved tyrosine residue that was not utilized by the screening hits. These data provide a foundation for developing LC stabilizers with improved binding selectivity and enhanced physicochemical properties.

Citation

Nicholas L Yan, Diogo Santos-Martins, Reji Nair, Alan Chu, Ian A Wilson, Kristen A Johnson, Stefano Forli, Gareth J Morgan, H Michael Petrassi, Jeffery W Kelly. Discovery of Potent Coumarin-Based Kinetic Stabilizers of Amyloidogenic Immunoglobulin Light Chains Using Structure-Based Design. Journal of medicinal chemistry. 2021 May 13;64(9):6273-6299

Expand section icon Mesh Tags

Expand section icon Substances


PMID: 33939422

View Full Text