Anti-JNK2 peptide-siRNA nanostructures improve plaque endothelium and reduce thrombotic risk in atherosclerotic mice

Int J Nanomedicine. 2018 Sep 6:13:5187-5205. doi: 10.2147/IJN.S168556. eCollection 2018.

Abstract

Background: A direct and independent role of inflammation in atherothrombosis was recently highlighted by the Canakinumab Antiinflammatory Thrombosis Outcome Study (CANTOS) trial, showing the benefit of inhibiting signaling molecules, eg, interleukins. Accordingly, we sought to devise a flexible platform for preventing the inflammatory drivers at their source to preserve plaque endothelium and mitigate procoagulant risk.

Methods: p5RHH-siRNA nanoparticles were formulated through self-assembly processes. The therapeutic efficacy of p5RHH-JNK2 siRNA nanoparticles was evaluated both in vitro and in vivo.

Results: Because JNK2 is critical to macrophage uptake of oxidized lipids through scavenger receptors that engender expression of myriad inflammatory molecules, we designed an RNA-silencing approach based on peptide-siRNA nanoparticles (p5RHH-siRNA) that localize to atherosclerotic plaques exhibiting disrupted endothelial barriers to achieve control of JNK2 expression by macrophages. After seven doses of p5RHH-JNK2 siRNA nanoparticles over 3.5 weeks in ApoE-/- mice on a Western diet, both JNK2 mRNA and protein levels were significantly decreased by 26% (P=0.044) and 42% (P=0.042), respectively. Plaque-macrophage populations were markedly depleted and NFκB and STAT3-signaling pathways inhibited by 47% (P<0.001) and 46% (P=0.004), respectively. Endothelial barrier integrity was restored (2.6-fold reduced permeability to circulating 200 nm nanoparticles in vivo, P=0.003) and thrombotic risk attenuated (200% increased clotting times to carotid artery injury, P=0.02), despite blood-cholesterol levels persistently exceeding 1,000 mg/dL. No adaptive or innate immunoresponses toward the nanoparticles were observed, and blood tests after the completion of treatment confirmed the largely nontoxic nature of this approach.

Conclusion: The ability to formulate these nanostructures rapidly and easily interchange or multiplex their oligonucleotide content represents a promising approach for controlling deleterious signaling events locally in advanced atherosclerosis.

Keywords: ApoE−/− mice; JNK2; atherosclerosis; endothelium; erosions; macrophages; peptide nanoparticles; perfluorocarbon nanoparticles; plaque; scavenger receptors; siRNA; thrombosis.

MeSH terms

  • Animals
  • Apolipoproteins E / deficiency
  • Apolipoproteins E / metabolism
  • Atherosclerosis / complications*
  • Atherosclerosis / pathology
  • Atherosclerosis / therapy
  • Disease Models, Animal
  • Endothelium / pathology*
  • Inflammation / metabolism
  • Macrophages / metabolism
  • Male
  • Mice
  • Mice, Knockout
  • Mitogen-Activated Protein Kinase 9 / metabolism*
  • Nanoparticles / chemistry
  • Nanostructures / chemistry*
  • Peptides / metabolism*
  • Plaque, Atherosclerotic / complications*
  • Plaque, Atherosclerotic / pathology
  • Plaque, Atherosclerotic / therapy
  • RAW 264.7 Cells
  • RNA Interference
  • RNA, Small Interfering / metabolism*
  • Risk Factors
  • Signal Transduction / drug effects
  • Thrombosis / complications*
  • Thrombosis / pathology
  • Thrombosis / therapy

Substances

  • Apolipoproteins E
  • Peptides
  • RNA, Small Interfering
  • Mitogen-Activated Protein Kinase 9