Abstract

BackgroundAdequate vascularization is crucial for supplying nutrition and discharging metabolic waste in freshly transplanted tissue-engineered constructs. Obtaining the appropriate building blocks for vascular tissue engineering (i.e. endothelial and mural cells) is a challenging task for tissue neovascularization. Hence, we investigated whether stem cells from human exfoliated deciduous teeth (SHED) could be induced to differentiate into functional vascular smooth muscle cells (vSMCs).MethodsWe utilized two cytokines of the TGF-β family, transforming growth factor beta 1 (TGF-β1) and bone morphogenetic protein 4 (BMP4), to induce SHED differentiation into SMCs. Quantitative real-time polymerase chain reaction (RT-qPCR) was used to assess mRNA expression, and protein expression was analyzed using flow cytometry, western blot and immunostaining. Additionally, to examine whether these SHED-derived SMCs possess the same function as primary SMCs, in vitro Matrigel angiogenesis assay, fibrin gel bead assay, and functional contraction study were used here.ResultsBy analyzing the expression of specific markers of SMCs (α-SMA, SM22α, Calponin, and SM-MHC), we confirmed that TGF-β1, and not BMP4, could induce SHED differentiation into SMCs. The differentiation efficiency was relatively high (α-SMA+ 86.1%, SM22α+ 93.9%, Calponin+ 56.8%, and SM-MHC+ 88.2%) as assessed by flow cytometry. In vitro Matrigel angiogenesis assay showed that the vascular structures generated by SHED-derived SMCs and human umbilical vein endothelial cells (HUVECs) were comparable to primary SMCs and HUVECs in terms of vessel stability. Fibrin gel bead assay showed that SHED-derived SMCs had a stronger capacity for promoting vessel formation compared with primary SMCs. Further analyses of protein expression in fibrin gel showed that cultures containing SHED-derived SMCs exhibited higher expression levels of Fibronectin than the primary SMCs group. Additionally, it was also confirmed that SHED-derived SMCs exhibited functional contractility. When SB-431542, a specific inhibitor of ALK5 was administered, TGF-β1 stimulation could not induce SHED into SMCs, indicating that the differentiation of SHED into SMCs is somehow related to the TGF-β1-ALK5 signaling pathway.ConclusionsSHED could be successfully induced into functional SMCs for vascular tissue engineering, and this course could be regulated through the ALK5 signaling pathway. Hence, SHED appear to be a promising candidate cell type for vascular tissue engineering.

Highlights

  • Adequate vascularization is crucial for supplying nutrition and discharging metabolic waste in freshly transplanted tissue-engineered constructs

  • TGF-β1 can induce the differentiation of stem cells from human exfoliated deciduous teeth (SHED) into SMCs As two distinct cytokines of the TGF-β family, TGF-β1 and bone morphogenetic protein 4 (BMP4) were arbitrarily selected to induce SMCs differentiation

  • We demonstrated that under the stimulatory effects of TGF-β1, SHED could differentiate into functional SMCs. alpha-smooth muscle actin (α-SMA) is the most widely recognized marker of SMCs [28], but it is still necessary to combine this with other markers to positively identify SMCs

Read more

Summary

Introduction

Adequate vascularization is crucial for supplying nutrition and discharging metabolic waste in freshly transplanted tissue-engineered constructs. We investigated whether stem cells from human exfoliated deciduous teeth (SHED) could be induced to differentiate into functional vascular smooth muscle cells (vSMCs). Generating functional perivascular cells is crucial for vascular tissue engineering. Most studies on vascular tissue engineering utilize commercially available primary cells, such as human umbilical vein endothelial cells (HUVECs), human umbilical artery endothelial cells (HUAECs), vascular smooth muscle cells (vSMCs), and pericytes (PCs) [3, 4]. The scarce availability of human tissue sources for primary cell isolation, limited proliferative capacity, slow expansion rate, and variability of tissue-specific phenotype would hinder the clinical translation of mature ECs and vSMCs [4]. Inducing human embryonic, induced pluripotent, or adult stem cells, and endothelial progenitor cells to generate ECs and vSMCs have been explored [5, 6]. Tumorigenicity, immunocompatibility and lack of precise control of differentiation are challenges that need to be addressed

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

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