Cargando…

Differentiation of mesenchymal stem cells from human amniotic fluid to cardiomyocyte-like cells

Ischemic heart disease (IHD) is a major factor influencing worldwide mortality rates. Furthermore, IHD has become a significant health problem among the Thai population. Stem cell therapy using mesenchymal stem cells (MSCs) is an alternative therapeutic method that has been applied to improve the qu...

Descripción completa

Detalles Bibliográficos
Autores principales: Markmee, Runchana, Aungsuchawan, Sirinda, Narakornsak, Suteera, Tancharoen, Waleephan, Bumrungkit, Kanokkarn, Pangchaidee, Nataporn, Pothacharoen, Peraphan, Puaninta, Chaniporn
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5865810/
https://www.ncbi.nlm.nih.gov/pubmed/28849052
http://dx.doi.org/10.3892/mmr.2017.7333
_version_ 1783308748873269248
author Markmee, Runchana
Aungsuchawan, Sirinda
Narakornsak, Suteera
Tancharoen, Waleephan
Bumrungkit, Kanokkarn
Pangchaidee, Nataporn
Pothacharoen, Peraphan
Puaninta, Chaniporn
author_facet Markmee, Runchana
Aungsuchawan, Sirinda
Narakornsak, Suteera
Tancharoen, Waleephan
Bumrungkit, Kanokkarn
Pangchaidee, Nataporn
Pothacharoen, Peraphan
Puaninta, Chaniporn
author_sort Markmee, Runchana
collection PubMed
description Ischemic heart disease (IHD) is a major factor influencing worldwide mortality rates. Furthermore, IHD has become a significant health problem among the Thai population. Stem cell therapy using mesenchymal stem cells (MSCs) is an alternative therapeutic method that has been applied to improve the quality of life of patients. Amniotic fluid (AF) contains a heterogeneous cell population, including MSCs, which are multipotent stem cells that have the capability to differentiate into mesenchymal lineages. The purpose of the present study was to evaluate the MSC characteristics of human (h)AF and determine its potency regarding cardiogenic differentiation. MSC characterization following flow cytometric analysis revealed that the cells expressed MSC markers, cluster of differentiation (CD)44, CD90, human leukocyte antigen-ABC and CD73. The results of the alamar blue assay demonstrated that cell proliferation rate continuously increased from the early cultivation phase up to 5-fold during days 1 to 5 of cell culturing. The highest rate of cell proliferation was observed on day 17 with a 30-fold increase compared with that on day 1. During the cardiogenic induction stage, morphological changes were observed between day 0 and day 21, and it was revealed that the hAF derived-MSCs in the cardiogenic-induced group exhibited myotube-like morphology after 7 days of cell culturing. Following cardiogenic induction, immunohistochemistry staining was performed on day 21, and reverse transcription-quantitative polymerase chain reaction on day 7 and 21. These steps were performed to detect the protein and gene expression levels of cardiac specific proteins (GATA4, cardiac troponin T, Nkx2.5 and Connexin43). The results of the present study indicated that hAF-MSCs possess the potential to differentiate into cardiomyocyte-like cells. Thus, it was concluded that hAF may be a suitable source of MSCs for stem cell therapy and tissue engineering.
format Online
Article
Text
id pubmed-5865810
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-58658102018-03-27 Differentiation of mesenchymal stem cells from human amniotic fluid to cardiomyocyte-like cells Markmee, Runchana Aungsuchawan, Sirinda Narakornsak, Suteera Tancharoen, Waleephan Bumrungkit, Kanokkarn Pangchaidee, Nataporn Pothacharoen, Peraphan Puaninta, Chaniporn Mol Med Rep Articles Ischemic heart disease (IHD) is a major factor influencing worldwide mortality rates. Furthermore, IHD has become a significant health problem among the Thai population. Stem cell therapy using mesenchymal stem cells (MSCs) is an alternative therapeutic method that has been applied to improve the quality of life of patients. Amniotic fluid (AF) contains a heterogeneous cell population, including MSCs, which are multipotent stem cells that have the capability to differentiate into mesenchymal lineages. The purpose of the present study was to evaluate the MSC characteristics of human (h)AF and determine its potency regarding cardiogenic differentiation. MSC characterization following flow cytometric analysis revealed that the cells expressed MSC markers, cluster of differentiation (CD)44, CD90, human leukocyte antigen-ABC and CD73. The results of the alamar blue assay demonstrated that cell proliferation rate continuously increased from the early cultivation phase up to 5-fold during days 1 to 5 of cell culturing. The highest rate of cell proliferation was observed on day 17 with a 30-fold increase compared with that on day 1. During the cardiogenic induction stage, morphological changes were observed between day 0 and day 21, and it was revealed that the hAF derived-MSCs in the cardiogenic-induced group exhibited myotube-like morphology after 7 days of cell culturing. Following cardiogenic induction, immunohistochemistry staining was performed on day 21, and reverse transcription-quantitative polymerase chain reaction on day 7 and 21. These steps were performed to detect the protein and gene expression levels of cardiac specific proteins (GATA4, cardiac troponin T, Nkx2.5 and Connexin43). The results of the present study indicated that hAF-MSCs possess the potential to differentiate into cardiomyocyte-like cells. Thus, it was concluded that hAF may be a suitable source of MSCs for stem cell therapy and tissue engineering. D.A. Spandidos 2017-11 2017-08-23 /pmc/articles/PMC5865810/ /pubmed/28849052 http://dx.doi.org/10.3892/mmr.2017.7333 Text en Copyright: © Markmee et al. This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Markmee, Runchana
Aungsuchawan, Sirinda
Narakornsak, Suteera
Tancharoen, Waleephan
Bumrungkit, Kanokkarn
Pangchaidee, Nataporn
Pothacharoen, Peraphan
Puaninta, Chaniporn
Differentiation of mesenchymal stem cells from human amniotic fluid to cardiomyocyte-like cells
title Differentiation of mesenchymal stem cells from human amniotic fluid to cardiomyocyte-like cells
title_full Differentiation of mesenchymal stem cells from human amniotic fluid to cardiomyocyte-like cells
title_fullStr Differentiation of mesenchymal stem cells from human amniotic fluid to cardiomyocyte-like cells
title_full_unstemmed Differentiation of mesenchymal stem cells from human amniotic fluid to cardiomyocyte-like cells
title_short Differentiation of mesenchymal stem cells from human amniotic fluid to cardiomyocyte-like cells
title_sort differentiation of mesenchymal stem cells from human amniotic fluid to cardiomyocyte-like cells
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5865810/
https://www.ncbi.nlm.nih.gov/pubmed/28849052
http://dx.doi.org/10.3892/mmr.2017.7333
work_keys_str_mv AT markmeerunchana differentiationofmesenchymalstemcellsfromhumanamnioticfluidtocardiomyocytelikecells
AT aungsuchawansirinda differentiationofmesenchymalstemcellsfromhumanamnioticfluidtocardiomyocytelikecells
AT narakornsaksuteera differentiationofmesenchymalstemcellsfromhumanamnioticfluidtocardiomyocytelikecells
AT tancharoenwaleephan differentiationofmesenchymalstemcellsfromhumanamnioticfluidtocardiomyocytelikecells
AT bumrungkitkanokkarn differentiationofmesenchymalstemcellsfromhumanamnioticfluidtocardiomyocytelikecells
AT pangchaideenataporn differentiationofmesenchymalstemcellsfromhumanamnioticfluidtocardiomyocytelikecells
AT pothacharoenperaphan differentiationofmesenchymalstemcellsfromhumanamnioticfluidtocardiomyocytelikecells
AT puanintachaniporn differentiationofmesenchymalstemcellsfromhumanamnioticfluidtocardiomyocytelikecells