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

Mahmoud, Amira D. and Ballantyne, Margaret D. and Miscianinov, Vladislav and Pinel, Karine and Hung, John and Scanlon, Jessica P. and Iyinikkel, Jean and Kaczynski, Jakub and Tavares, Adriana S. and Bradshaw, Angela C. and Mills, Nicholas L. and Newby, David E. and Caporali, Andrea and Gould, Gwyn W. and George, Sarah J. and Ulitsky, Igor and Sluimer, Judith C. and Rodor, Julie and Baker, Andrew H. (2019) 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. Circulation Research, 125 (5). pp. 535-551. ISSN 0009-7330 (https://doi.org/10.1161/CIRCRESAHA.119.314876)

[thumbnail of Mahmoud-etal-CR-2019-The-human-specific-and-smooth-muscle-cell-enriched-LncRNA-SMILR-promotes-proliferation]
Preview
Text. Filename: Mahmoud_etal_CR_2019_The_human_specific_and_smooth_muscle_cell_enriched_LncRNA_SMILR_promotes_proliferation.pdf
Final Published Version
License: Creative Commons Attribution 4.0 logo

Download (8MB)| Preview

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)-1α 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 ≈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 ≈29% in controls 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.