Abstract

Hyperglycaemia causes endothelial dysfunction, which is the initial process in the development of diabetic vascular complications. Upon injury, endothelial cells undergo an endothelial-to-mesenchymal transition (EndMT), lose their specific marker, and gain mesenchymal phenotypes. This study investigated the effect of liraglutide, a glucagon-like peptide 1 (GLP-1) receptor agonist, on EndMT inhibition and neointima formation in diabetic mice induced by streptozotocin. The diabetic mice with a wire-induced vascular injury in the right carotid artery were treated with or without liraglutide for four weeks. The degree of neointima formation and re-endothelialisation was evaluated by histological assessments. Endothelial fate tracing revealed that endothelium-derived cells contribute to neointima formation through EndMT in vivo. In the diabetic mouse model, liraglutide attenuated wire injury-induced neointima formation and accelerated re-endothelialisation. In vitro, a high glucose condition (30 mmol/L) triggered morphological changes and mesenchymal marker expression in human umbilical vein endothelial cells (HUVECs), which were attenuated by liraglutide or Activin receptor-like 5 (ALK5) inhibitor SB431542. The inhibition of AMP-activated protein kinase (AMPK) signaling by Compound C diminished the liraglutide-mediated inhibitory effect on EndMT. Collectively, liraglutide was found to attenuate neointima formation in diabetic mice partially through EndMT inhibition, extending the potential therapeutic role of liraglutide.

Highlights

  • Diabetes mellitus (DM) causes hyperglycaemia and systemic inflammation, leading to endothelial dysfunction [1,2,3]

  • The inhibition of AMPK by Compound C attenuated the liraglutide-mediated effects on signal transduction (p-AMPK and p-Smad2) and endothelial-to-mesenchymal transition (EndMT) marker expression (CD31, SM22α, Snail, and vimentin) (Figure 6b,c). These results indicate that liraglutide reverses high glucose (HG)+IL-1β-induced EndMT via the AMPK pathway

  • Our results show that HG condition-treated human umbilical vein endothelial cells (HUVECs) acquired spindle morphology and with previous reports that HG induced a loss of endothelial phenotype in HUVECs [16,31]

Read more

Summary

Introduction

Diabetes mellitus (DM) causes hyperglycaemia and systemic inflammation, leading to endothelial dysfunction [1,2,3]. Endothelial damage is the initial process in the progression of diabetic vascular complications [4,5]. The endothelial origin of mesenchymal or fibroblast-like cells can be characterized by the loss of. Cells 2019, 8, 589 cell-cell junctions, acquisition of invasive and migratory capacities, loss of endothelial markers (e.g., CD31, Tie, Tie, Vascular endothelial (VE)-Cadherin, and von Willebrand factor (vWF)), and gain of mesenchymal markers (e.g., fibroblast-specific protein 1(FSP-1), alpha-smooth muscle actin (a-SMA), smooth muscle-22alpha actin (SM22α), N-cadherin, and vimentin) [7,8,9,10]. Recent studies have revealed that inflammation and diabetic conditions, such as high glucose, trigger the trans-differentiation of endothelial cells into mesenchymal-like cells [13,14,15,16]. The EndMT process may be the initial stage of neointima hyperplasia under diabetic conditions

Methods
Results
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.