miR-342-5p Is a Notch Downstream Molecule and Regulates Multiple Angiogenic Pathways Including Notch, Vascular Endothelial Growth Factor and Transforming Growth Factor β Signaling

J Am Heart Assoc. 2016 Feb 8;5(2):e003042. doi: 10.1161/JAHA.115.003042.

Abstract

Background: Endothelial cells (ECs) form blood vessels through angiogenesis that is regulated by coordination of vascular endothelial growth factor (VEGF), Notch, transforming growth factor β, and other signals, but the detailed molecular mechanisms remain unclear.

Methods and results: Small RNA sequencing initially identified miR-342-5p as a novel downstream molecule of Notch signaling in ECs. Reporter assay, quantitative reverse transcription polymerase chain reaction and Western blot analysis indicated that miR-342-5p targeted endoglin and modulated transforming growth factor β signaling by repressing SMAD1/5 phosphorylation in ECs. Transfection of miR-342-5p inhibited EC proliferation and lumen formation and reduced angiogenesis in vitro and in vivo, as assayed by using a fibrin beads-based sprouting assay, mouse aortic ring culture, and intravitreal injection of miR-342-5p agomir in P3 pups. Moreover, miR-342-5p promoted the migration of ECs, accompanied by reduced endothelial markers and increased mesenchymal markers, indicative of increased endothelial-mesenchymal transition. Transfection of endoglin at least partially reversed endothelial-mesenchymal transition induced by miR-342-5p. The expression of miR-342-5p was upregulated by transforming growth factor β, and inhibition of miR-342-5p attenuated the inhibitory effects of transforming growth factor β on lumen formation and sprouting by ECs. In addition, VEGF repressed miR-342-5p expression, and transfection of miR-342-5p repressed VEGFR2 and VEGFR3 expression and VEGF-triggered Akt phosphorylation in ECs. miR-342-5p repressed angiogenesis in a laser-induced choroidal neovascularization model in mice, highlighting its clinical potential.

Conclusions: miR-342-5p acts as a multifunctional angiogenic repressor mediating the effects and interaction among angiogenic pathways.

Keywords: Notch; angiogenesis; endothelial cell; endothelial cell differentiation; microRNA; vascular endothelial growth factor.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • 3' Untranslated Regions
  • Animals
  • Binding Sites
  • Cell Adhesion Molecules / genetics
  • Cell Adhesion Molecules / metabolism
  • Cell Movement / drug effects
  • Cell Proliferation / drug effects
  • Choroidal Neovascularization / genetics
  • Choroidal Neovascularization / metabolism*
  • Choroidal Neovascularization / pathology
  • Choroidal Neovascularization / prevention & control
  • Disease Models, Animal
  • Endoglin / genetics
  • Endoglin / metabolism
  • Epithelial-Mesenchymal Transition / drug effects
  • HeLa Cells
  • Human Umbilical Vein Endothelial Cells / drug effects*
  • Human Umbilical Vein Endothelial Cells / metabolism
  • Humans
  • Mice, Inbred BALB C
  • Mice, Transgenic
  • MicroRNAs / genetics
  • MicroRNAs / metabolism*
  • Neovascularization, Physiologic / drug effects*
  • Phosphorylation
  • Proto-Oncogene Proteins c-akt
  • Receptor, Notch1 / genetics
  • Receptor, Notch1 / metabolism*
  • Signal Transduction / drug effects
  • Time Factors
  • Transfection
  • Transforming Growth Factor beta / metabolism
  • Transforming Growth Factor beta / pharmacology*
  • Vascular Endothelial Growth Factor A / metabolism
  • Vascular Endothelial Growth Factor A / pharmacology*

Substances

  • 3' Untranslated Regions
  • Cell Adhesion Molecules
  • ENG protein, human
  • EVL protein, human
  • Endoglin
  • MIRN342 microRNA, human
  • MicroRNAs
  • Mirn342 microRNA, mouse
  • NOTCH1 protein, human
  • Notch1 protein, mouse
  • Receptor, Notch1
  • Transforming Growth Factor beta
  • Vascular Endothelial Growth Factor A
  • Proto-Oncogene Proteins c-akt