Abstract

Cell penetrating peptides constitute a potent approach to overcome the limitations of in vivo siRNA delivery. We recently proposed a peptide-based nanoparticle system, CADY, for efficient delivery of siRNA into numerous cell lines. CADY is a secondary amphipathic peptide that forms stable complexes with siRNA thereby improving both their cellular uptake and biological response. With the aim of understanding the cellular uptake mechanism of CADY:siRNA complexes, we have combined biochemical, confocal and electron microscopy approaches. In the present work, we provide evidence that the major route for CADY:siRNA cellular uptake involves direct translocation through the membrane but not the endosomal pathway. We have demonstrated that CADY:siRNA complexes do not colocalize with most endosomal markers and remain fully active in the presence of inhibitors of the endosomal pathway. Moreover, neither electrostatic interactions with cell surface heparan sulphates nor membrane potential are essential for CADY:siRNA cell entry. In contrast, we have shown that CADY:siRNA complexes clearly induce a transient cell membrane permeabilization, which is rapidly restored by cell membrane fluidity. Therefore, we propose that direct translocation is the major gate for cell entry of CADY:siRNA complexes. Membrane perturbation and uptake are driven mainly by the ability of CADY to interact with phospholipids within the cell membrane, followed by rapid localization of the complex in the cytoplasm, without affecting cell integrity or viability.

Highlights

  • Small interfering RNA has great potential as a therapeutic molecule, due to its high target specificity, efficiency for gene silencing and its simple design [1,2]

  • This result confirms our previous results, showing that in vitro, CADY:Small interfering RNA (siRNA) complexes can interact with heparan sulfate proteoglycans (HSPGs), implying a possible initial electrostatic contact at the cell surface [28]

  • Due to it positive net charge CADY:siRNA complexes can interact with HSPG, an interaction which is not required for cellular uptake, but which partially dissociate the CADY:siRNA complex

Read more

Summary

Introduction

Small interfering RNA (siRNA) has great potential as a therapeutic molecule, due to its high target specificity, efficiency for gene silencing and its simple design [1,2]. The success of siRNA is dependent on carrier molecules and numerous non-viral strategies have been proposed to improve the delivery of synthetic small oligonucleotides [6,7,8,9]. Cell-penetrating peptides (CPPs) have been widely used for the delivery of therapeutic molecules and have been reported to favour the delivery of a large panel of cargos (plasmid DNA, oligonucleotide, siRNA, PNA, protein, peptide, liposome, nanoparticle...) into a wide variety of cell types and in vivo models [10,11,12]. CPPs can be subdivided into two main classes, the first requiring chemical linkage with the cargo and the second involving the formation of stable, non-covalent complexes [10,11,12]

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