[HTML][HTML] PERK inhibition promotes post-angioplasty re-endothelialization via modulating SMC phenotype changes

B Wang, M Zhang, G Urabe, T Shirasu, LW Guo… - Journal of Surgical …, 2021 - Elsevier
B Wang, M Zhang, G Urabe, T Shirasu, LW Guo, KC Kent
Journal of Surgical Research, 2021Elsevier
Background Drug-eluting stents impair post-angioplasty re-endothelialization thus
compromising restenosis prevention while heightening thrombotic risks. We recently found
that inhibition of protein kinase RNA-like endoplasmic reticulum kinase (PERK) effectively
mitigated both restenosis and thrombosis in rodent models. This motivated us to determine
how PERK inhibition impacts re-endothelialization. Methods Re-endothelialization was
evaluated in endothelial-denuded rat carotid arteries after balloon angioplasty and …
Background
Drug-eluting stents impair post-angioplasty re-endothelialization thus compromising restenosis prevention while heightening thrombotic risks. We recently found that inhibition of protein kinase RNA-like endoplasmic reticulum kinase (PERK) effectively mitigated both restenosis and thrombosis in rodent models. This motivated us to determine how PERK inhibition impacts re-endothelialization.
Methods
Re-endothelialization was evaluated in endothelial-denuded rat carotid arteries after balloon angioplasty and periadventitial administration of PERK inhibitor in a hydrogel. To study whether PERK in smooth muscle cells (SMCs) regulates re-endothelialization by paracrinally influencing endothelial cells (ECs), denuded arteries exposing SMCs were lentiviral-infected to silence PERK; in vitro, the extracellular vesicles isolated from the medium of PDGF-activated, PERK-upregulating human primary SMCs were transferred to human primary ECs.
Results
Treatment with PERK inhibitor versus vehicle control accelerated re-endothelialization in denuded arteries. PERK-specific silencing in the denuded arterial wall (mainly SMCs) also enhanced re-endothelialization compared to scrambled shRNA control. In vitro, while medium transfer from PDGF-activated SMCs impaired EC viability and increased the mRNA levels of dysfunctional EC markers, either PERK inhibition or silencing in donor SMCs mitigated these EC changes. Furthermore, CXCL10, a paracrine cytokine detrimental to ECs, was increased by PDGF activation and decreased after PERK inhibition or silencing in SMCs.
Conclusions
Attenuating PERK activity pharmacologically or genetically provides an approach to accelerating post-angioplasty re-endothelialization in rats. The mechanism may involve paracrine factors regulated by PERK in SMCs that impact neighboring ECs. This study rationalizes future development of PERK-targeted endothelium-friendly vascular interventions.
Elsevier