Abstract

In the search for novel broad-spectrum therapeutics to fight chronic infections, inflammation, and cancer, host defense peptides (HDPs) have garnered increasing interest. Characterizing their biologically-active conformations and minimum motifs for function represents a requisite step to developing them into efficacious and safe therapeutics. Here, we demonstrate that metallating HDPs with Cu2+ is an effective chemical strategy to improve their cytotoxicity on cancer cells. Mechanistically, we find that prepared as Cu2+-complexes, the peptides not only physically but also chemically damage lipid membranes. Our testing ground features piscidins 1 and 3 (P1/3), two amphipathic, histidine-rich, membrane-interacting, and cell-penetrating HDPs that are α-helical bound to membranes. To investigate their membrane location, permeabilization effects, and lipid-oxidation capability, we employ neutron reflectometry, impedance spectroscopy, neutron diffraction, and UV spectroscopy. While P1-apo is more potent than P3-apo, metallation boosts their cytotoxicities by up to two- and seven-fold, respectively. Remarkably, P3-Cu2+ is particularly effective at inserting in bilayers, causing water crevices in the hydrocarbon region and placing Cu2+ near the double bonds of the acyl chains, as needed to oxidize them. This study points at a new paradigm where complexing HDPs with Cu2+ to expand their mechanistic reach could be explored to design more potent peptide-based anticancer therapeutics.

Highlights

  • In the search for novel broad-spectrum therapeutics to fight chronic infections, inflammation, and cancer, host defense peptides (HDPs) have garnered increasing interest

  • In previous biophysical studies of piscidins 1 and 3 (P1/3), we demonstrated that the stronger membrane permeabilizing effects and pH-resiliency of piscidin 1 (P1) correlate with its higher histidine content and a membrane-inserted state that places its C-terminus in the bilayer c­ ore[56]

  • Free C­ u2+ tested at up to 30 μmol/L was not cytotoxic under the same conditions (Fig. S1B). While it is not known whether the peptides remain bound to ­Cu2+ throughout the process of killing cancer cells, it is essential for C­ u2+ and the peptides to be bound to each other in order to reach low I­C50 values

Read more

Summary

Introduction

In the search for novel broad-spectrum therapeutics to fight chronic infections, inflammation, and cancer, host defense peptides (HDPs) have garnered increasing interest Characterizing their biologically-active conformations and minimum motifs for function represents a requisite step to developing them into efficacious and safe therapeutics. Given the intersection between life-threatening infections and cancers, there is a strong motivation to develop therapeutics with broad-spectrum activity to treat both conditions simultaneously With this regard, several membrane-interacting host-defense peptides (HDPs) with cytotoxic properties against cancer cells have emerged as promising ­templates[7,8,15,16,17,18,19,20,21,22]. We show that these biological effects correlate with the ability of the peptides to physically and chemically impair lipid membranes

Methods
Results
Discussion
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