Cargando…

Human adipose-derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial-mesenchymal transition by cross interacting with the TGF-β/Smad and PI3K/AKT signaling pathways

The influence and underlying mechanisms of human adipose-derived stem cells (Hu-ADSCs) on breast cancer cells in the tumor microenvironment remain unclear. Understanding the association between Hu-ADSCs and cancer cells may provide targets for breast cancer treatment and reference for the clinical a...

Descripción completa

Detalles Bibliográficos
Autores principales: Wu, Simeng, Wang, Yajun, Yuan, Zhe, Wang, Siliang, Du, Hongmei, Liu, Xue, Wang, Qiushi, Zhu, Xike
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6297785/
https://www.ncbi.nlm.nih.gov/pubmed/30483746
http://dx.doi.org/10.3892/mmr.2018.9664
_version_ 1783381205435023360
author Wu, Simeng
Wang, Yajun
Yuan, Zhe
Wang, Siliang
Du, Hongmei
Liu, Xue
Wang, Qiushi
Zhu, Xike
author_facet Wu, Simeng
Wang, Yajun
Yuan, Zhe
Wang, Siliang
Du, Hongmei
Liu, Xue
Wang, Qiushi
Zhu, Xike
author_sort Wu, Simeng
collection PubMed
description The influence and underlying mechanisms of human adipose-derived stem cells (Hu-ADSCs) on breast cancer cells in the tumor microenvironment remain unclear. Understanding the association between Hu-ADSCs and cancer cells may provide targets for breast cancer treatment and reference for the clinical application of stem cells. Therefore, a Hu-ADSC and breast cancer MCF7 cell coculture system was established to investigate the paracrine effects of Hu-ADSCs on MCF7 cell migration and invasion, in addition to the potential mechanism of action by reverse transcription-quantitative polymerase chain reaction and western blotting. Hu-ADSCs enhanced MCF7 cell migration and invasion by decreasing the expression of epithelial marker E-cadherin, and increasing the expression of interstitial marker N-cadherin and epithelial-mesenchymal transition (EMT) transcription factors in vitro. The EMT effect of cocultured MCF7 cells was inhibited with the addition of anti-transforming growth factor (TGF)-β1 or phosphoinositide 3-kinase (PI3K) inhibitor LY294002, accompanied by a significant decrease in phosphorylated (p)-mothers against decapentaplegic homolog (Smad) and p-protein kinase B (AKT) expression. The data suggested that the paracrine effect of Hu-ADSCs in the tumor microenvironment promoted the EMT of MCF7 cells by cross interacting with the TGF-β/Smad and PI3K/AKT pathways.
format Online
Article
Text
id pubmed-6297785
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-62977852018-12-26 Human adipose-derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial-mesenchymal transition by cross interacting with the TGF-β/Smad and PI3K/AKT signaling pathways Wu, Simeng Wang, Yajun Yuan, Zhe Wang, Siliang Du, Hongmei Liu, Xue Wang, Qiushi Zhu, Xike Mol Med Rep Articles The influence and underlying mechanisms of human adipose-derived stem cells (Hu-ADSCs) on breast cancer cells in the tumor microenvironment remain unclear. Understanding the association between Hu-ADSCs and cancer cells may provide targets for breast cancer treatment and reference for the clinical application of stem cells. Therefore, a Hu-ADSC and breast cancer MCF7 cell coculture system was established to investigate the paracrine effects of Hu-ADSCs on MCF7 cell migration and invasion, in addition to the potential mechanism of action by reverse transcription-quantitative polymerase chain reaction and western blotting. Hu-ADSCs enhanced MCF7 cell migration and invasion by decreasing the expression of epithelial marker E-cadherin, and increasing the expression of interstitial marker N-cadherin and epithelial-mesenchymal transition (EMT) transcription factors in vitro. The EMT effect of cocultured MCF7 cells was inhibited with the addition of anti-transforming growth factor (TGF)-β1 or phosphoinositide 3-kinase (PI3K) inhibitor LY294002, accompanied by a significant decrease in phosphorylated (p)-mothers against decapentaplegic homolog (Smad) and p-protein kinase B (AKT) expression. The data suggested that the paracrine effect of Hu-ADSCs in the tumor microenvironment promoted the EMT of MCF7 cells by cross interacting with the TGF-β/Smad and PI3K/AKT pathways. D.A. Spandidos 2019-01 2018-11-19 /pmc/articles/PMC6297785/ /pubmed/30483746 http://dx.doi.org/10.3892/mmr.2018.9664 Text en Copyright: © Wu 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
Wu, Simeng
Wang, Yajun
Yuan, Zhe
Wang, Siliang
Du, Hongmei
Liu, Xue
Wang, Qiushi
Zhu, Xike
Human adipose-derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial-mesenchymal transition by cross interacting with the TGF-β/Smad and PI3K/AKT signaling pathways
title Human adipose-derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial-mesenchymal transition by cross interacting with the TGF-β/Smad and PI3K/AKT signaling pathways
title_full Human adipose-derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial-mesenchymal transition by cross interacting with the TGF-β/Smad and PI3K/AKT signaling pathways
title_fullStr Human adipose-derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial-mesenchymal transition by cross interacting with the TGF-β/Smad and PI3K/AKT signaling pathways
title_full_unstemmed Human adipose-derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial-mesenchymal transition by cross interacting with the TGF-β/Smad and PI3K/AKT signaling pathways
title_short Human adipose-derived mesenchymal stem cells promote breast cancer MCF7 cell epithelial-mesenchymal transition by cross interacting with the TGF-β/Smad and PI3K/AKT signaling pathways
title_sort human adipose-derived mesenchymal stem cells promote breast cancer mcf7 cell epithelial-mesenchymal transition by cross interacting with the tgf-β/smad and pi3k/akt signaling pathways
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6297785/
https://www.ncbi.nlm.nih.gov/pubmed/30483746
http://dx.doi.org/10.3892/mmr.2018.9664
work_keys_str_mv AT wusimeng humanadiposederivedmesenchymalstemcellspromotebreastcancermcf7cellepithelialmesenchymaltransitionbycrossinteractingwiththetgfbsmadandpi3kaktsignalingpathways
AT wangyajun humanadiposederivedmesenchymalstemcellspromotebreastcancermcf7cellepithelialmesenchymaltransitionbycrossinteractingwiththetgfbsmadandpi3kaktsignalingpathways
AT yuanzhe humanadiposederivedmesenchymalstemcellspromotebreastcancermcf7cellepithelialmesenchymaltransitionbycrossinteractingwiththetgfbsmadandpi3kaktsignalingpathways
AT wangsiliang humanadiposederivedmesenchymalstemcellspromotebreastcancermcf7cellepithelialmesenchymaltransitionbycrossinteractingwiththetgfbsmadandpi3kaktsignalingpathways
AT duhongmei humanadiposederivedmesenchymalstemcellspromotebreastcancermcf7cellepithelialmesenchymaltransitionbycrossinteractingwiththetgfbsmadandpi3kaktsignalingpathways
AT liuxue humanadiposederivedmesenchymalstemcellspromotebreastcancermcf7cellepithelialmesenchymaltransitionbycrossinteractingwiththetgfbsmadandpi3kaktsignalingpathways
AT wangqiushi humanadiposederivedmesenchymalstemcellspromotebreastcancermcf7cellepithelialmesenchymaltransitionbycrossinteractingwiththetgfbsmadandpi3kaktsignalingpathways
AT zhuxike humanadiposederivedmesenchymalstemcellspromotebreastcancermcf7cellepithelialmesenchymaltransitionbycrossinteractingwiththetgfbsmadandpi3kaktsignalingpathways