Cargando…

Cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors

BACKGROUND: Persistent epithelial defects (PED), associated with limbal stem cell deficiency (LSCD), require ocular surface reconstruction with a stable corneal epithelium (CE). This study investigated CE reformation using human adipose mesenchymal stem cells (ADSC), which derived epithelial progeni...

Descripción completa

Detalles Bibliográficos
Autores principales: Bandeira, Francisco, Goh, Tze-Wei, Setiawan, Melina, Yam, Gary Hin-Fai, Mehta, Jodhbir S.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6942321/
https://www.ncbi.nlm.nih.gov/pubmed/31900226
http://dx.doi.org/10.1186/s13287-019-1533-1
_version_ 1783484680228569088
author Bandeira, Francisco
Goh, Tze-Wei
Setiawan, Melina
Yam, Gary Hin-Fai
Mehta, Jodhbir S.
author_facet Bandeira, Francisco
Goh, Tze-Wei
Setiawan, Melina
Yam, Gary Hin-Fai
Mehta, Jodhbir S.
author_sort Bandeira, Francisco
collection PubMed
description BACKGROUND: Persistent epithelial defects (PED), associated with limbal stem cell deficiency (LSCD), require ocular surface reconstruction with a stable corneal epithelium (CE). This study investigated CE reformation using human adipose mesenchymal stem cells (ADSC), which derived epithelial progenitors via mesenchymal-epithelial transition (MET). METHODS: STEMPRO human ADSC were cultured with specific inhibitors antagonizing glycogen synthase kinase-3 and transforming growth factor-β signaling, followed by culture under a defined progenitor cell targeted-epithelial differentiation condition to generate epithelial-like cells (MET-Epi), which were characterized for cell viability, mesenchymal, and epithelial phenotypes using immunofluorescence and flow cytometry. Tissue-engineered (TE) MET-Epi cells on fibrin gel were transplanted to corneal surface of the rat LSCD model caused by alkali injury. Epithelial healing, corneal edema, and haze grading, CE formation were assessed by fluorescein staining, slit lamp bio-microscopy, anterior segment optical coherence tomography, and immunohistochemistry. RESULTS: CD73(high)/CD90(high)/CD105(high)/CD166(high)/CD14(negative)/CD31(negative) human ADSC underwent MET, giving viable epithelial-like progenitors expressing δNp63, CDH1 (E-cadherin), epidermal growth factor receptor, integrin-β4, and cytokeratin (CK)-5, 9. Under defined epithelial differentiation culture, these progenitors generated MET-Epi cells expressing cell junction proteins ZO1 and occludin. When transplanted onto rat corneal surface with LSCD-induced PED, TE-MET-Epi achieved more efficient epithelial healing, suppressed corneal edema, and opacities, when compared to corneas without treatment or transplanted with TE-ADSC. CE markers (CK3, 12, and CDH1) were expressed on TE-MET-Epi-transplanted corneas but not in other control groups. CONCLUSION: Human ADSC-derived epithelial-like cells, via MET, recovered the CE from PED associated with LSCD. ADSC can be a viable adult stem cell source for potential autologous epithelial cell-based therapy for corneal surface disorders.
format Online
Article
Text
id pubmed-6942321
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-69423212020-01-07 Cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors Bandeira, Francisco Goh, Tze-Wei Setiawan, Melina Yam, Gary Hin-Fai Mehta, Jodhbir S. Stem Cell Res Ther Research BACKGROUND: Persistent epithelial defects (PED), associated with limbal stem cell deficiency (LSCD), require ocular surface reconstruction with a stable corneal epithelium (CE). This study investigated CE reformation using human adipose mesenchymal stem cells (ADSC), which derived epithelial progenitors via mesenchymal-epithelial transition (MET). METHODS: STEMPRO human ADSC were cultured with specific inhibitors antagonizing glycogen synthase kinase-3 and transforming growth factor-β signaling, followed by culture under a defined progenitor cell targeted-epithelial differentiation condition to generate epithelial-like cells (MET-Epi), which were characterized for cell viability, mesenchymal, and epithelial phenotypes using immunofluorescence and flow cytometry. Tissue-engineered (TE) MET-Epi cells on fibrin gel were transplanted to corneal surface of the rat LSCD model caused by alkali injury. Epithelial healing, corneal edema, and haze grading, CE formation were assessed by fluorescein staining, slit lamp bio-microscopy, anterior segment optical coherence tomography, and immunohistochemistry. RESULTS: CD73(high)/CD90(high)/CD105(high)/CD166(high)/CD14(negative)/CD31(negative) human ADSC underwent MET, giving viable epithelial-like progenitors expressing δNp63, CDH1 (E-cadherin), epidermal growth factor receptor, integrin-β4, and cytokeratin (CK)-5, 9. Under defined epithelial differentiation culture, these progenitors generated MET-Epi cells expressing cell junction proteins ZO1 and occludin. When transplanted onto rat corneal surface with LSCD-induced PED, TE-MET-Epi achieved more efficient epithelial healing, suppressed corneal edema, and opacities, when compared to corneas without treatment or transplanted with TE-ADSC. CE markers (CK3, 12, and CDH1) were expressed on TE-MET-Epi-transplanted corneas but not in other control groups. CONCLUSION: Human ADSC-derived epithelial-like cells, via MET, recovered the CE from PED associated with LSCD. ADSC can be a viable adult stem cell source for potential autologous epithelial cell-based therapy for corneal surface disorders. BioMed Central 2020-01-03 /pmc/articles/PMC6942321/ /pubmed/31900226 http://dx.doi.org/10.1186/s13287-019-1533-1 Text en © The Author(s). 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Bandeira, Francisco
Goh, Tze-Wei
Setiawan, Melina
Yam, Gary Hin-Fai
Mehta, Jodhbir S.
Cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors
title Cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors
title_full Cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors
title_fullStr Cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors
title_full_unstemmed Cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors
title_short Cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors
title_sort cellular therapy of corneal epithelial defect by adipose mesenchymal stem cell-derived epithelial progenitors
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6942321/
https://www.ncbi.nlm.nih.gov/pubmed/31900226
http://dx.doi.org/10.1186/s13287-019-1533-1
work_keys_str_mv AT bandeirafrancisco cellulartherapyofcornealepithelialdefectbyadiposemesenchymalstemcellderivedepithelialprogenitors
AT gohtzewei cellulartherapyofcornealepithelialdefectbyadiposemesenchymalstemcellderivedepithelialprogenitors
AT setiawanmelina cellulartherapyofcornealepithelialdefectbyadiposemesenchymalstemcellderivedepithelialprogenitors
AT yamgaryhinfai cellulartherapyofcornealepithelialdefectbyadiposemesenchymalstemcellderivedepithelialprogenitors
AT mehtajodhbirs cellulartherapyofcornealepithelialdefectbyadiposemesenchymalstemcellderivedepithelialprogenitors