Abstract

IntroductionEndothelial dysfunction is found in different pathologies such as diabetes and renal and heart diseases, representing one of the major health problems. The reduced vasodilation of impaired endothelium starts a prothrombotic state associated with irregular blood flow. We aimed to explore the potential of amniotic fluid stem (AFS) cells as a source for regenerative medicine in this field; for the first time, we focused on third-trimester amniotic fluid AFS cells and compared them with the already-described AFS cells from the second trimester.MethodsCells from the two trimesters were cultured, selected and expanded in normoxia (20 % oxygen) and hypoxia (5 % oxygen). Cells were analysed to compare markers, proliferation rate and differentiation abilities. Endothelial potential was assessed not only in vitro—Matrigel tube formation assay, acetylated human low-density lipoprotein (AcLDL) uptake—but also in vivo (Matrigel plug with cell injection and two animal models). Specifically, for the latter, we used established protocols to assess the involvement of AFS cells in two different mouse models of endothelial dysfunction: (1) a chronic ischemia model with local injection of cells and (2) an electric carotid damage where cells were systemically injected.ResultsWe isolated and expanded AFS cells from third-trimester amniotic fluid samples by using CD117 as a selection marker. Hypoxia enhanced the proliferation rate, the surface protein pattern was conserved between the trimesters and comparable differentiation was achieved after culture in both normoxia and hypoxia. Notably, the expression of early endothelial transcription factors and AngiomiRs was detected before and after induction. When in vivo, AFS cells from both trimesters expanded in hypoxia were able to rescue the surface blood flow when locally injected in mice after chronic ischemia damage, and importantly AFS cells at term of gestation possessed enhanced ability to fix carotid artery electric damage compared with AFS cells from the second trimester.ConclusionsTo the best of our knowledge, this is the first research work that fully characterizes AFS cells from the third trimester for regenerative medicine purposes. The results highlight how AFS cells, in particular at term of gestation and cultured in hypoxia, can be considered a promising source of stem cells possessing significant endothelial regenerative potential.Electronic supplementary materialThe online version of this article (doi:10.1186/s13287-015-0204-0) contains supplementary material, which is available to authorized users.

Highlights

  • Endothelial dysfunction is found in different pathologies such as diabetes and renal and heart diseases, representing one of the major health problems

  • Endothelial progenitor cells derived from induced pluripotent stem and embryonic stem cells [2, 3], Wharton Jelly and umbilical cord blood (CB) cells [2] have been recently employed to recover the endothelial function after ischemia, but restricted therapeutic use due to safety issues and weak differentiation efficiency is limiting the applications of these cells

  • A specific subset of amniotic fluid stem (AFS) cells presenting the surface antigen CD117 has been isolated, demonstrated to be broadly multipotent [4, 5], and can be reprogrammed without viral transduction [6]. Because of their fetal but non-embryonic origin, AFS cells overcome many ethical concerns; they are easy to obtain from routinely scheduled procedures during the second trimester of pregnancy [7], retain immunoprivileged properties [8] and do not form teratoma once injected in vivo [4]

Read more

Summary

Introduction

Endothelial dysfunction is found in different pathologies such as diabetes and renal and heart diseases, representing one of the major health problems. A specific subset of amniotic fluid stem (AFS) cells presenting the surface antigen CD117 (or c-Kit, the receptor for the stem cell factor, or SCF) has been isolated, demonstrated to be broadly multipotent [4, 5], and can be reprogrammed without viral transduction [6] Because of their fetal but non-embryonic origin, AFS cells overcome many ethical concerns; they are easy to obtain from routinely scheduled procedures during the second trimester of pregnancy (i.e., amniocentesis and amnioreduction) [7], retain immunoprivileged properties [8] and do not form teratoma once injected in vivo [4]. With particular attention to a long-term culture, hypoxia inhibits senescence, increases the proliferation rate, influences the potential of differentiation and modulates the paracrine effects of stem cells, causing upregulation of various secreted factors [16]

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