Targeted Therapy of Atherosclerosis by a Broad-Spectrum Reactive Oxygen Species Scavenging Nanoparticle with Intrinsic Anti-inflammatory Activity

ACS Nano. 2018 Sep 25;12(9):8943-8960. doi: 10.1021/acsnano.8b02037. Epub 2018 Aug 21.

Abstract

Atherosclerosis is a leading cause of vascular diseases worldwide. Whereas antioxidative therapy has been considered promising for the treatment of atherosclerosis in view of a critical role of reactive oxygen species (ROS) in the pathogenesis of atherosclerosis, currently available antioxidants showed considerably limited clinical outcomes. Herein, we hypothesize that a broad-spectrum ROS-scavenging nanoparticle can serve as an effective therapy for atherosclerosis, taking advantage of its antioxidative stress activity and targeting effects. As a proof of concept, a broad-spectrum ROS-eliminating material was synthesized by covalently conjugating a superoxide dismutase mimetic agent Tempol and a hydrogen-peroxide-eliminating compound of phenylboronic acid pinacol ester onto a cyclic polysaccharide β-cyclodextrin (abbreviated as TPCD). TPCD could be easily processed into a nanoparticle (TPCD NP). The obtained nanotherapy TPCD NP could be efficiently and rapidly internalized by macrophages and vascular smooth muscle cells (VSMCs). TPCD NPs significantly attenuated ROS-induced inflammation and cell apoptosis in macrophages, by eliminating overproduced intracellular ROS. Also, TPCD NPs effectively inhibited foam cell formation in macrophages and VSMCs by decreasing internalization of oxidized low-density lipoprotein. After intravenous (i.v.) administration, TPCD NPs accumulated in atherosclerotic lesions of apolipoprotein E-deficient (ApoE-/-) mice by passive targeting through the dysfunctional endothelium and translocation via inflammatory cells. TPCD NPs significantly inhibited the development of atherosclerosis in ApoE-/- mice after i.v. delivery. More importantly, therapy with TPCD NPs afforded stabilized plaques with less cholesterol crystals, a smaller necrotic core, thicker fibrous cap, and lower macrophages and matrix metalloproteinase-9, compared with those treated with control drugs previously developed for antiatherosclerosis. The therapeutic benefits of TPCD NPs mainly resulted from reduced systemic and local oxidative stress and inflammation as well as decreased inflammatory cell infiltration in atherosclerotic plaques. Preliminary in vivo tests implied that TPCD NPs were safe after long-term treatment via i.v. injection. Consequently, TPCD NPs can be developed as a potential antiatherosclerotic nanotherapy.

Keywords: anti-inflammation; antioxidative stress; atherosclerosis; nanoparticle; reactive oxygen species.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Anti-Inflammatory Agents, Non-Steroidal / chemistry
  • Anti-Inflammatory Agents, Non-Steroidal / pharmacology*
  • Apolipoproteins E / deficiency
  • Apolipoproteins E / metabolism
  • Apoptosis / drug effects
  • Atherosclerosis / drug therapy*
  • Atherosclerosis / metabolism
  • Boronic Acids / chemistry
  • Boronic Acids / pharmacology
  • Cyclic N-Oxides / chemistry
  • Cyclic N-Oxides / pharmacology
  • Hydrogen Peroxide / chemistry
  • Hydrogen Peroxide / pharmacology
  • Inflammation / drug therapy
  • Inflammation / metabolism
  • Macrophages / drug effects
  • Macrophages / metabolism
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Nanoparticles / chemistry
  • Nanoparticles / metabolism*
  • Polysaccharides / chemistry
  • Polysaccharides / pharmacology
  • Reactive Oxygen Species / metabolism*
  • Spin Labels
  • beta-Cyclodextrins / chemistry
  • beta-Cyclodextrins / pharmacology

Substances

  • Anti-Inflammatory Agents, Non-Steroidal
  • Apolipoproteins E
  • Boronic Acids
  • Cyclic N-Oxides
  • Polysaccharides
  • Reactive Oxygen Species
  • Spin Labels
  • beta-Cyclodextrins
  • Hydrogen Peroxide
  • betadex
  • benzeneboronic acid
  • tempol