Basic Study
Copyright ©The Author(s) 2020. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Feb 26, 2020; 12(2): 123-138
Published online Feb 26, 2020. doi: 10.4252/wjsc.v12.i2.123
Clonal isolation of endothelial colony-forming cells from early gestation chorionic villi of human placenta for fetal tissue regeneration
Kewa Gao, Siqi He, Priyadarsini Kumar, Diana Farmer, Jianda Zhou, Aijun Wang
Kewa Gao, Siqi He, Jianda Zhou, Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha 410013, Hunan Province, China
Kewa Gao, Siqi He, Priyadarsini Kumar, Diana Farmer, Aijun Wang, Surgical Bioengineering Laboratory, Department of Surgery, University of California Davis, Sacramento, CA 95817, United States
Kewa Gao, Siqi He, Priyadarsini Kumar, Diana Farmer, Aijun Wang, Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, United States
Aijun Wang, Department of Biomedical Engineering, University of California Davis, Davis, CA 95817, United States
Author contributions: This study was designed and supervised by Wang A, Zhou J and Farmer D; experiments were performed by Gao K, He S and Kumar P; data analysis was conducted by Gao K and Wang A; funding was obtained by Farmer D and Wang A; the manuscript was written and revised by Gao K, He S, Kumar P, Farmer D, Zhou J and Wang A.
Supported by the Shriners Hospital for Children Postdoctoral Research Fellowship award, No. 84704-NCA-19; UC Davis School of Medicine Dean’s Fellowship award and funding from the NIH, No. 5R01NS100761-02 and No.R03HD091601-01; the California Institute of Regenerative Medicine, No. PC1-08103 and No. CLIN1-11404; Shriners Hospitals for Children, No. 85120-NCA-16, No. 85119-NCA-18, No. 85108-NCA-19 and No. 87200-NCA-19; March of Dimes Foundation, No. 5FY1682.
Institutional review board statement: The study was submitted to the UCD Institutional Review Board (IRB) and determined to be exempt from review.
Conflict-of-interest statement: Authors of this manuscript have no conflicts of interest to disclose.
Data sharing statement: We agree to share the research methods and data in this study to public.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Aijun Wang, PhD, Associate Professor, Surgical Bioengineering Laboratory, Department of Surgery, University of California, Davis School of Medicine, 4625 2nd Ave., Room 3005, Sacramento, CA 95817, United States. aawang@ucdavis.edu
Received: July 5, 2019
Peer-review started: July 17, 2019
First decision: August 23, 2019
Revised: December 3, 2019
Accepted: December 23, 2019
Article in press: December 23, 2019
Published online: February 26, 2020
ARTICLE HIGHLIGHTS
Research background

Fetal medicine and fetal surgery have been substantially developed for the treatment of congenital defects. Stem cell transplantation is an important means of tissue reconstruction and repair of genetic defects. Endothelial colony-forming cells (ECFCs) represent a promising cell candidate for their unique role in facilitating the formation of angiogenesis and vascularization.

Research motivation

ECFCs isolated from the chorionic villus tissue of early gestation placentas can serve as a source of cells for prenatal autologous fetal cell transplantation, as well as provide a basis for studying the development, physiological function, congenital disease, and fetal treatment of the developing fetus and placenta.

Research objectives

The objective of this study is to establish an isolation protocol to obtain ECFCs from the chorionic villus of human early gestation placentas, as well as to investigate the characterization of these cells and their potential applications in gene delivery and tissue engineering.

Research methods

Dissected chorionic villus tissues were enzymatically digested to obtain single cells. Then, magnetic bead sorting, monoclonal culture and colony isolation were performed to obtain chorionic villi-derived ECFCs (CV-ECFCs). Immunohistochemical identification, flow cytometry, Matrigel tube formation assays, LDL uptake assays and lentiviral transduction were carried out to characterize the morphology, phenotype and function of the purified and expanded CV-ECFCs.

Research results

Using the established isolation protocol, we were able to obtain 1.8 × 107 pure CV-ECFCs from a single cell colony culture within 6-8 wks. CV-ECFCs showed typical endothelial phenotypes and functions. CV-ECFCs have demonstrated the ability to be transduced with lentiviruses, and function as carriers for gene therapy. They also possess good biocompatibility with biomaterial delivery vehicles, such as small intestinal submucosa extracellular matrix for potential tissue engineering applications.

Research conclusions

This study shows that ECFCs are present in early gestation placental chorionic villi, and can be isolated and expanded to a significant number in a short period of time. These CV-ECFCs possess typical endothelial cell phenotypes and functions, and hold the potential of being used in gene therapy and tissue engineering applications.

Research perspectives

CV-ECFCs isolated from early gestation placentas provide a new source of ECFCs for the fetal treatment of congenital disorders. Combined with existing in utero treatment technologies, this cell therapy could be widely applied toward a variety of diseases and conditions. In future research, we will further explore the in vivo applications of these cells in various animal models. Investigating the phenotype and functions of CV-ECFCs will also facilitate our understanding of the development, cellular composition, and function of the developing placenta and its interaction with the developing fetus.