The Human-Specific and Smooth Muscle Cell-Enriched LncRNA SMILR Promotes Proliferation by Regulating Mitotic CENPF mRNA and Drives Cell-Cycle Progression Which Can Be Targeted to Limit Vascular Remodeling

Amira D. Mahmoud, Margaret D. Ballantyne, Vladislav Miscianinov, Karine Pinel, John Hung, Jessica P. Scanlon, Jean Iyinikkel, Jakub Kaczynski, Adriana S. Tavares, Angela C. Bradshaw, Nicholas L. Mills, David E. Newby, Andrea Caporali, Gwyn W. Gould, Sarah J. George, Igor Ulitsky, Judith C. Sluimer, Julie Rodor, Andrew H. Baker*

*Corresponding author for this work

Research output: Contribution to journalArticleAcademicpeer-review

Abstract

Rationale: In response to blood vessel wall injury, aberrant proliferation of vascular smooth muscle cells (SMCs) causes pathological remodeling. However, the controlling mechanisms are not completely understood. Objective: We recently showed that the human long noncoding RNA, SMILR, promotes vascular SMCs proliferation by a hitherto unknown mechanism. Here, we assess the therapeutic potential of SMILR inhibition and detail the molecular mechanism of action. Methods and Results: We used deep RNA-sequencing of human saphenous vein SMCs stimulated with IL (interleukin)-1 alpha and PDGF (platelet-derived growth factor)-BB with SMILR knockdown (siRNA) or overexpression (lentivirus), to identify SMILR-regulated genes. This revealed a SMILR-dependent network essential for cell cycle progression. In particular, we found using the fluorescent ubiquitination-based cell cycle indicator viral system that SMILR regulates the late mitotic phase of the cell cycle and cytokinesis with SMILR knockdown resulting in approximate to 10% increase in binucleated cells. SMILR pulldowns further revealed its potential molecular mechanism, which involves an interaction with the mRNA of the late mitotic protein CENPF (centromere protein F) and the regulatory Staufen1 RNA-binding protein. SMILR and this downstream axis were also found to be activated in the human ex vivo vein graft pathological model and in primary human coronary artery SMCs and atherosclerotic plaques obtained at carotid endarterectomy. Finally, to assess the therapeutic potential of SMILR, we used a novel siRNA approach in the ex vivo vein graft model (within the 30 minutes clinical time frame that would occur between harvest and implant) to assess the reduction of proliferation by EdU incorporation. SMILR knockdown led to a marked decrease in proliferation from approximate to 29% in controls to approximate to 5% with SMILR depletion. Conclusions: Collectively, we demonstrate that SMILR is a critical mediator of vascular SMC proliferation via direct regulation of mitotic progression. Our data further reveal a potential SMILR-targeting intervention to limit atherogenesis and adverse vascular remodeling.

Original languageEnglish
Pages (from-to)535-551
Number of pages17
JournalCirculation Research
Volume125
Issue number5
DOIs
Publication statusPublished - 16 Aug 2019

Keywords

  • blood vessels
  • cell cycle
  • growth factors
  • interleukins
  • muscle cells
  • noncoding RNA
  • saphenous vein
  • LONG NONCODING RNA
  • EXPRESSION
  • MIGRATION
  • STENTS
  • DIFFERENTIATION
  • PROGNOSIS
  • NETWORKS
  • BIOLOGY

Cite this