Abstract

BackgroundProliferation and migration of endothelial progenitor cells (EPCs) play important roles in restoring vascular injuries. β2 adrenergic receptors (β2ARs) are widely expressed in many tissues and have a beneficial impact on EPCs regulating neoangiogenesis. The aim of the present study was to determine the effect of overexpressing β2ARs in infused peripheral blood (PB)-derived EPCs on the re-endothelialization in injured vessels.MethodsInduction of endothelial injury was performed in male nude mice that were subjected to wire-mediated injury to the carotid artery. Human PB-derived EPCs were transfected with an adenovirus serotype 5 vector expressing β2AR (Ad5/β2AR-EPCs) and were examined 48 h later. β2AR gene expression in EPCs was detected by real-time polymerase chain reaction and Western blot analysis. In vitro, the proliferation, migration, adhesion, and nitric oxide production of Ad5/β2AR-EPCs were measured. Meanwhile, phosphorylated Akt and endothelial nitric oxide synthase (eNOS), which are downstream of β2AR signaling, were also elevated. In an in vivo study, CM-DiI-labeled EPCs were injected intravenously into mice subjected to carotid injury. After 3 days, cells recruited to the injury sites were detected by fluorescent microscopy, and the re-endothelialization was assessed by Evans blue dye.ResultsIn vitro, β2AR overexpression augmented EPC proliferation, migration, and nitric oxide production and enhanced EPC adhesion to endothelial cell monolayers. In vivo, when cell tracking was used, the number of recruited CM-DiI-labeled EPCs was significantly higher in the injured zone in mice transfused with Ad5/β2AR-EPCs compared with non-transfected EPCs. The degree of re-endothelialization was also higher in the mice transfused with Ad5/β2AR-EPCs compared with non-transfected EPCs. We also found that the phosphorylation of Akt and eNOS was increased in Ad5/β2AR-EPCs. Preincubation with β2AR inhibitor (ICI118,551), Akt inhibitor (ly294002), or eNOS inhibitor (L-NAME) significantly attenuated the enhanced in vitro function and in vivo re-endothelialization capacity of EPCs induced by β2AR overexpression.ConclusionsThe present study demonstrates that β2AR overexpression enhances EPC functions in vitro and enhances the vascular repair abilities of EPCs in vivo via the β2AR/Akt/eNOS pathway. Upregulation of β2AR gene expression through gene transfer may be a novel therapeutic target for endothelial repair.

Highlights

  • Proliferation and migration of endothelial progenitor cells (EPCs) play important roles in restoring vascular injuries. β2 adrenergic receptors (β2ARs) are widely expressed in many tissues and have a beneficial impact on EPCs regulating neoangiogenesis

  • Cellar immunostaining showed that most of the adherent cells have doublepositive staining for the uptake of DiI-acetylated low-density lipoprotein (ac-LDL) and for the binding of fluorescein isothiocyanate (FITC)-lectin, indicating that these cells possess the functional properties of endothelial cell (EC) (Fig. 1a)

  • Fluorescence microscopy revealed that β2ARs were found to localize to the cell membrane of EPCs, and these results were confirmed by using a positive marker, endothelial nitric oxide synthase (eNOS), which is expressed in EPCs (Fig. 1c)

Read more

Summary

Introduction

Proliferation and migration of endothelial progenitor cells (EPCs) play important roles in restoring vascular injuries. β2 adrenergic receptors (β2ARs) are widely expressed in many tissues and have a beneficial impact on EPCs regulating neoangiogenesis. Proliferation and migration of endothelial progenitor cells (EPCs) play important roles in restoring vascular injuries. The aim of the present study was to determine the effect of overexpressing β2ARs in infused peripheral blood (PB)-derived EPCs on the re-endothelialization in injured vessels. Accumulating evidence indicates that the balance between endothelial injury and repair is a key component of atherosclerosis [2], and maintaining endothelial integrity is crucially important to preventing the initiation and development of atherosclerosis, coronary heart disease, and postangioplasty restenosis [3]. Endothelial progenitor cells (EPCs) mobilized from bone marrow into the peripheral blood (PB) have been shown to play an important role for vascular regeneration, endothelial repair, and replacement of dysfunctional endothelium by incorporating into the site of vessel injury, differentiating into endothelial cells (ECs), and releasing paracrine factors [4, 5]. Attempts to improve the function of transplanted EPCs with gene modifications may facilitate the repair of damaged endothelia and accelerate re-endothelialization

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.