File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Conversion of stem cells from apical papilla into endothelial cells by small molecules and growth factors

TitleConversion of stem cells from apical papilla into endothelial cells by small molecules and growth factors
Authors
KeywordsSCAP
Endothelial differentiation
Small molecules
Chemical reprogramming
Angiogenesis
Issue Date2021
PublisherBioMed Central Ltd. The Journal's web site is located at http://www.stemcellres.com
Citation
Stem Cell Research & Therapy, 2021, v. 12, p. article no. 266 How to Cite?
AbstractObjectives: Recently, a new strategy has been developed to directly reprogram one cell type towards another targeted cell type using small molecule compounds. Human fibroblasts have been chemically reprogrammed into neuronal cells, Schwann cells and cardiomyocyte-like cells by different small molecule combinations. This study aimed to explore whether stem cells from apical papilla (SCAP) could be reprogrammed into endothelial cells (ECs) using the same strategy. Materials and methods: The expression level of endothelial-specific genes and proteins after chemical induction of SCAP was assessed by RT-PCR, western blotting, flow cytometry and immunofluorescence. The in vitro functions of SCAP-derived chemical-induced endothelial cells (SCAP-ECs) were evaluated by tube-like structure formation assay, acetylated low-density lipoprotein (ac-LDL) uptake and NO secretion detection. The proliferation and the migration ability of SCAP-ECs were evaluated by CCK-8 and Transwell assay. LPS stimulation was used to mimic the inflammatory environment in demonstrating the ability of SCAP-ECs to express adhesion molecules. The in vivo Matrigel plug angiogenesis assay was performed to assess the function of SCAP-ECs in generating vascular structures using the immune-deficient mouse model. Results: SCAP-ECs expressed upregulated endothelial-specific genes and proteins; displayed endothelial transcriptional networks; exhibited the ability to form functional tubular-like structures, uptake ac-LDL and secrete NO in vitro; and contributed to generate blood vessels in vivo. The SCAP-ECs could also express adhesion molecules in the pro-inflammatory environment and have a similar migration and proliferation ability as HUVECs. Conclusions: Our study demonstrates that the set of small molecules and growth factors could significantly promote endothelial transdifferentiation of SCAP, which provides a promising candidate cell source for vascular engineering and treatment of ischemic diseases.
Persistent Identifierhttp://hdl.handle.net/10722/300584
ISSN
2021 Impact Factor: 8.079
2020 SCImago Journal Rankings: 1.599
PubMed Central ID
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorYI, B-
dc.contributor.authorDing, T-
dc.contributor.authorJiang, S-
dc.contributor.authorGong, T-
dc.contributor.authorChopra, H-
dc.contributor.authorSha, O-
dc.contributor.authorDissanayaka, WL-
dc.contributor.authorGe, S-
dc.contributor.authorZhang, C-
dc.date.accessioned2021-06-18T14:54:06Z-
dc.date.available2021-06-18T14:54:06Z-
dc.date.issued2021-
dc.identifier.citationStem Cell Research & Therapy, 2021, v. 12, p. article no. 266-
dc.identifier.issn1757-6512-
dc.identifier.urihttp://hdl.handle.net/10722/300584-
dc.description.abstractObjectives: Recently, a new strategy has been developed to directly reprogram one cell type towards another targeted cell type using small molecule compounds. Human fibroblasts have been chemically reprogrammed into neuronal cells, Schwann cells and cardiomyocyte-like cells by different small molecule combinations. This study aimed to explore whether stem cells from apical papilla (SCAP) could be reprogrammed into endothelial cells (ECs) using the same strategy. Materials and methods: The expression level of endothelial-specific genes and proteins after chemical induction of SCAP was assessed by RT-PCR, western blotting, flow cytometry and immunofluorescence. The in vitro functions of SCAP-derived chemical-induced endothelial cells (SCAP-ECs) were evaluated by tube-like structure formation assay, acetylated low-density lipoprotein (ac-LDL) uptake and NO secretion detection. The proliferation and the migration ability of SCAP-ECs were evaluated by CCK-8 and Transwell assay. LPS stimulation was used to mimic the inflammatory environment in demonstrating the ability of SCAP-ECs to express adhesion molecules. The in vivo Matrigel plug angiogenesis assay was performed to assess the function of SCAP-ECs in generating vascular structures using the immune-deficient mouse model. Results: SCAP-ECs expressed upregulated endothelial-specific genes and proteins; displayed endothelial transcriptional networks; exhibited the ability to form functional tubular-like structures, uptake ac-LDL and secrete NO in vitro; and contributed to generate blood vessels in vivo. The SCAP-ECs could also express adhesion molecules in the pro-inflammatory environment and have a similar migration and proliferation ability as HUVECs. Conclusions: Our study demonstrates that the set of small molecules and growth factors could significantly promote endothelial transdifferentiation of SCAP, which provides a promising candidate cell source for vascular engineering and treatment of ischemic diseases.-
dc.languageeng-
dc.publisherBioMed Central Ltd. The Journal's web site is located at http://www.stemcellres.com-
dc.relation.ispartofStem Cell Research & Therapy-
dc.rightsStem Cell Research & Therapy. Copyright © BioMed Central Ltd.-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectSCAP-
dc.subjectEndothelial differentiation-
dc.subjectSmall molecules-
dc.subjectChemical reprogramming-
dc.subjectAngiogenesis-
dc.titleConversion of stem cells from apical papilla into endothelial cells by small molecules and growth factors-
dc.typeArticle-
dc.identifier.emailDissanayaka, WL: warunad@hku.hk-
dc.identifier.emailZhang, C: zhangcf@hku.hk-
dc.identifier.authorityDissanayaka, WL=rp02216-
dc.identifier.authorityZhang, C=rp01408-
dc.description.naturepublished_or_final_version-
dc.identifier.doi10.1186/s13287-021-02350-5-
dc.identifier.pmid33941255-
dc.identifier.pmcidPMC8091697-
dc.identifier.scopuseid_2-s2.0-85105216129-
dc.identifier.hkuros322786-
dc.identifier.volume12-
dc.identifier.spagearticle no. 266-
dc.identifier.epagearticle no. 266-
dc.identifier.isiWOS:000656353500006-
dc.publisher.placeUnited Kingdom-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats