Abstract

Background: Target identification is a critical step in elucidating the mechanism of action (MoA) for bioactive compounds. For phenotypic drug discovery pipelines, current unbiased, label-free chemoproteomics-based methods rely predominantly on the modulation of target thermal stability upon drug binding. We developed an automated drug target deconvolution workflow combining limited proteolysis with mass spectrometry (LiP-Quant) that exploits protein structural alterations, as well as steric effects driven by drug binding. A major advantage of LiP-Quant is its unique focus on the detection of signature peptides that discern ligand binding, peptides that are generated by a limited digestion and identified by proteomic analysis. Here we demonstrate the performance of LiP-Quant using two well-characterized kinase inhibitors (KIs), Selumetinib (SE) and Staurosporine (ST), as well as two natural product-derived phosphatase inhibitors (PIs) Calyculin A and Fostriecin in human cell lysate. Furthermore, LiP-Quant can be deployed to estimate in-lysate EC50 value of compound binding.

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