Elsevier

Biotechnology Advances

Volume 33, Issue 6, Part 1, 1 November 2015, Pages 931-940
Biotechnology Advances

Research review paper
A role for peptides in overcoming endosomal entrapment in siRNA delivery — A focus on melittin

https://doi.org/10.1016/j.biotechadv.2015.05.005Get rights and content

Abstract

siRNA has the possibility to revolutionize medicine by enabling highly specific and efficient silencing of proteins involved in disease pathogenesis. Despite nearly 20 years of research dedicated to translating siRNA from a research tool into a clinically relevant therapeutic, minimal success has been had to date. Access to RNA interference machinery located in the cytoplasm is often overlooked, but must be considered when designing the next generation of siRNA delivery strategies. Peptide transduction domains (PTDs) have demonstrated moderate siRNA transfection, which is primarily limited by endosomal entrapment. Strategies aimed at overcoming endosomal entrapment associated with peptide vectors are reviewed here, including osmotic methods, lipid conjugation, and fusogenic peptides. As an alternative to traditional PTD, the hemolytic peptide melittin exhibits the native capacity for endosomal disruption but causes cytotoxicity. However, appropriate packaging and protection of melittin with activation and release in the endosomal compartment has allowed melittin-based strategies to demonstrate both in vitro and in vivo safety and efficacy. These data suggest that melittin's membrane disruptive properties can enable safe and effective endosomolysis, building a case for melittin as a key component in a new generation of siRNA therapeutics.

Introduction

RNA interference (RNAi) refers to an evolutionarily conserved mechanism for post-transcriptional control of protein expression in which short double-stranded RNAs target specific messenger RNA (mRNA) for degradation, thus decreasing protein translation (Eccleston and Eggleston, 2004). Tuschl et al. were the first to demonstrate that RNAi can be artificially induced by the delivery of exogenous small interfering RNA (siRNA) (Elbashir et al., 2001). siRNAs are short 21–23 base pair duplex RNA oligonucleotides with 5′-phosphorylated ends and 2-nucleotide 3′ overhangs. The “antisense” strand shares sequence complementarity to a target mRNA, while the “sense” strand serves as a bystander. When delivered into the cytoplasm of a cell, siRNA can co-opt the native RNAi machinery and induce assembly of the RNA induced silencing complex (RISC). The RISC unwinds siRNA, binds the antisense strand, and cleaves the sense strand, allowing the RISC to bind and cleave targeted mRNA based on the sequence of the antisense strand (Sakurai et al., 2011) (Fig. 1). This selective degradation of mRNA provides an avenue to decrease the expression of proteins involved in disease pathogenesis. Indeed, initial experimental siRNA therapeutics appeared just three years after the discovery of siRNA (McCaffrey et al., 2002). Recently, clinical trials have been initiated for the use of siRNA in ocular, renal, and hepatic diseases with limited success (Haussecker, 2012).

On a cellular level, efficient and non-toxic siRNA delivery to the cytoplasm remains a major impediment to the use of siRNA as a therapeutic. siRNAs are large (~ 21 kDa), highly charged macromolecules, which cannot directly translocate across the hydrophobic core of the cell membrane (Overhoff and Sczakiel, 2005). Moreover, the barrier provided by impermeable membrane bilayers not only applies to direct translocation from the extracellular milieu into the cytoplasm, but also applies to cytosolic access of siRNA enclosed in endocytic vesicles (Detzer and Sczakiel, 2009, Gilleron et al., 2013). Entrapment of siRNA in endocytic compartments not only prevents siRNA from reaching the cytosol, but also accelerates siRNA degradation due to the harsh acidic environment encountered during endosome–lysosome trafficking (Fig. 2) (Wang and Thanou, 2010).

This barrier represents a major impediment in the delivery of biological therapeutics to the cytoplasm, especially when packaged with peptide transduction domains (El-Sayed et al., 2009). Strategies to achieve endosomolysis have traditionally been based on osmotic agents, fusogenic lipids, and fusogenic peptides. The application of these methodologies to peptide-mediated siRNA delivery will be reviewed here. In addition, novel strategies enabled by the membrane-disruptive properties of melittin appear to negate the need for secondary endosomolytic methodologies and will be presented as an alternative to traditional peptide-based siRNA delivery strategies.

Section snippets

Cationic peptides for siRNA transfection

Given the challenges associated with traditional siRNA delivery methodology, there is clearly a need for siRNA delivery technology to enable endosomal escape with minimal cytotoxicity. With the observation that the Trans-Activator of Transcription (TAT) peptide from HIV could directly translocate across cell membranes to trans-activate the viral promoter in tissue culture, cell penetrating peptides (CPPs) have become a widely utilized tool for delivery of therapeutics (Frankel and Pabo, 1988).

Melittin as a basis for endosomal escape

It is apparent that peptide-mediated transfection is hampered by poor efficiency as a consequence of endosomal entrapment. Methods to overcome this limitation have focused on peptide modification to include membrane active peptides, lipids as well as chemical conjugation to proton buffering moieties. Other strategies focus on leveraging the membrane lytic properties of melittin to enable endosomal release of siRNA. Melittin is a 26 amino acid alpha helical peptide first purified from the

Conclusions

Successful application of siRNA as a therapeutic requires the development of vectors that can promote cytoplasmic delivery of siRNA without inducing cytotoxicity, but providing adequate endosomal escape. Peptides are a relatively new strategy for siRNA transfection, which appear to be limited by endosomal entrapment. Traditional methods to overcome the barrier of endosomal entrapment rely on augmenting existing vectors with osmotic agents, lipid moieties, or fusogenic peptides. These strategies

Acknowledgments

Research described here was supported by grants from the National Institutes of Health (U01 CA141541 to Dr. Robert Schreiber and RO1 HL073646-08 to SAW) and the Sigma Aldrich predoctoral fellowship to Dr. Kirk Hou.

References (108)

  • A.D. Frankel et al.

    Cellular uptake of the TAT protein from human immunodeficiency virus

    Cell

    (1988)
  • J.C. Geoghegan et al.

    Gene silencing mediated by siRNA-binding fusion proteins is attenuated by double-stranded RNA-binding domain structure

    Mol. Ther. Nucleic Acids

    (2012)
  • D. Haussecker

    The business of RNAi therapeutics in 2012

    Mol. Ther. Nucleic Acids

    (2012)
  • K.K. Hou et al.

    Melittin derived peptides for nanoparticle based siRNA transfection

    Biomaterials

    (2013)
  • J. Hoyer et al.

    Knockdown of a G protein-coupled receptor through efficient peptide-mediated siRNA delivery

    J. Control. Release

    (2012)
  • W.J. Kim et al.

    Cholesteryl oligoarginine delivering vascular endothelial growth factor siRNA effectively inhibits tumor growth in colon adenocarcinoma

    Mol. Ther.

    (2006)
  • S.W. Kim et al.

    RNA interference in vitro and in vivo using an arginine peptide/siRNA complex system

    J. Control. Release

    (2010)
  • A.S. Ladokhin et al.

    ‘Detergent-like’ permeabilization of anionic lipid vesicles by melittin

    BBA — Biomembranes

    (2001)
  • T. Lehto et al.

    Delivery of nucleic acids with a stearylated (RXR)4 peptide using a non-covalent co-incubation strategy

    J. Control. Release

    (2010)
  • T. Lehto et al.

    A peptide-based vector for efficient gene transfer in vitro and in vivo

    Mol. Ther.

    (2011)
  • S. Lindberg et al.

    Pepfect15, a novel endosomolytic cell-penetrating peptide for oligonucleotide delivery via scavenger receptors

    Int. J. Pharm.

    (2013)
  • S.L. Lo et al.

    An endosomolytic tat peptide produced by incorporation of histidine and cysteine residues as a nonviral vector for DNA transfection

    Biomaterials

    (2008)
  • M. Mäe et al.

    A stearylated cpp for delivery of splice correcting oligonucleotides using a non-covalent co-incubation strategy

    J. Control. Release

    (2009)
  • B.R. Meade et al.

    Exogenous siRNA delivery using peptide transduction domains/cell penetrating peptides

    Adv. Drug Deliv. Rev.

    (2007)
  • B.R. Meade et al.

    Enhancing the cellular uptake of siRNA duplexes following noncovalent packaging with protein transduction domain peptides

    Adv. Drug Deliv. Rev.

    (2008)
  • A. Muratovska et al.

    Conjugate for efficient delivery of short interfering RNA (siRNA) into mammalian cells

    FEBS Lett.

    (2004)
  • J. Regberg et al.

    Rational design of a series of novel amphipathic cell-penetrating peptides

    Int. J. Pharm.

    (2014)
  • G. Sessa et al.

    Interaction of a lytic polypeptide, melittin, with lipid membrane systems

    J. Biol. Chem.

    (1969)
  • K. Tanaka et al.

    Disulfide crosslinked stearoyl carrier peptides containing arginine and histidine enhance siRNA uptake and gene silencing

    Int. J. Pharm.

    (2010)
  • J.J. Turner et al.

    RNA targeting with peptide conjugates of oligonucleotides, siRNA and PNA

    Blood Cells Mol. Dis.

    (2007)
  • A.K. Varkouhi et al.

    Endosomal escape pathways for delivery of biologicals

    J. Control. Release

    (2011)
  • S.E.L. Andaloussi et al.

    Design of a peptide-based vector, pepfect6, for efficient delivery of siRNA in cell culture and systemically in vivo

    Nucleic Acids Res.

    (2011)
  • arrowheadresearch.com [Internet]

    Pasadena: Arrowhead Pharmaceuticals; [updated 2012 Oct 29; cited 2015 Mar 12]. Arrowhead presents data showing 99% target knockdown in monkeys without toxicity using subcutaneous formulation of dynamic polyconjugate siRNA delivery system

  • arrowheadresearch.com [Internet]

    Pasadena: Arrowhead Pharmaceuticals; [updated 2014 Nov 10; cited 2015 Mar 12]. “Arrowhead presents data on ARC-520 and ARC-AAT at AASLD the liver meeting 2014”

  • L. Babiss

    Driving creativity & innovation in research [Internet]

  • G.V.D. Bogaart et al.

    On the mechanism of pore formation by melittin

    J. Biol. Chem.

    (2008)
  • C.P. Chen et al.

    Gene transfer with poly-melittin peptides

    Bioconjug. Chem.

    (2006)
  • S.T. Chou et al.

    Selective modification of HK peptides enhances siRNA silencing of tumor targets in vivo

    Cancer Gene Ther.

    (2011)
  • L. Crombez et al.

    A non-covalent peptide-based strategy for siRNA delivery

  • L. Crombez et al.

    A new potent secondary amphipathic cell-penetrating peptide for siRNA delivery into mammalian cells

    Mol. Ther.

    (2008)
  • L. Crombez et al.

    Targeting cyclin B1 through peptide-based delivery of siRNA prevents tumour growth

    Nucleic Acids Res.

    (2009)
  • T.J. Davidson et al.

    Highly efficient small interfering RNA delivery to primary mammalian neurons induces microRNA-like effects before mRNA degradation

    J. Neurosci.

    (2004)
  • S. Deshayes et al.

    Self-assembling peptide-based nanoparticles for siRNA delivery in primary cell lines

    Small

    (2012)
  • A. Detzer et al.

    Phosphorothioate-stimulated uptake of siRNA by mammalian cells: a novel route for delivery

    Curr. Top. Med. Chem.

    (2009)
  • A. Eccleston et al.

    RNA interference

    Nature

    (2004)
  • A. Eguchi et al.

    Efficient siRNA delivery into primary cells by a peptide transduction domain–dsRNA binding domain fusion protein

    Nat. Biotechnol.

    (2009)
  • S.M. Elbashir et al.

    Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells

    Nature

    (2001)
  • A. El-Sayed et al.

    Delivery of macromolecules using arginine-rich cell-penetrating peptides: ways to overcome endosomal entrapment

    AAPS J.

    (2009)
  • T. Endoh et al.

    Cellular siRNA delivery mediated by a cell-permeant RNA-binding protein and photoinduced RNA interference

    Bioconjug. Chem.

    (2008)
  • A. Erazo-Oliveras et al.

    Improving the endosomal escape of cell-penetrating peptides and their cargos: strategies and challenges

    Pharmaceuticals

    (2012)
  • Cited by (63)

    • Three ‘E’ challenges for siRNA drug development

      2024, Trends in Molecular Medicine
    • Therapeutic targeting non-coding RNAs

      2023, Navigating Non-coding RNA: From Biogenesis to Therapeutic Application
    View all citing articles on Scopus
    View full text