Cargando…

Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-β1/ZEB1/miR-200c signaling pathway

Ursolic acid (UA) is a biologically active compound, commonly used in traditional Chinese medicine (TCM). It has been reported to exhibit strong anticancer properties against a variety of cancers. Our previous studies showed that UA promoted apoptosis in colorectal cancer (CRC) cells and inhibited c...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Ling, Cai, Qiao-Yan, Liu, Jianxin, Peng, Jun, Chen, You-Qin, Sferra, Thomas J., Lin, Jiu-Mao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6676672/
https://www.ncbi.nlm.nih.gov/pubmed/31452805
http://dx.doi.org/10.3892/ol.2019.10604
_version_ 1783440811141103616
author Zhang, Ling
Cai, Qiao-Yan
Liu, Jianxin
Peng, Jun
Chen, You-Qin
Sferra, Thomas J.
Lin, Jiu-Mao
author_facet Zhang, Ling
Cai, Qiao-Yan
Liu, Jianxin
Peng, Jun
Chen, You-Qin
Sferra, Thomas J.
Lin, Jiu-Mao
author_sort Zhang, Ling
collection PubMed
description Ursolic acid (UA) is a biologically active compound, commonly used in traditional Chinese medicine (TCM). It has been reported to exhibit strong anticancer properties against a variety of cancers. Our previous studies showed that UA promoted apoptosis in colorectal cancer (CRC) cells and inhibited cellular proliferation and angiogenesis. However, the effect and underlying molecular mechanism of UA in CRC progression remain unclear. In the present study, the role of UA in suppressing the migration and invasion of human colon cancer HCT116 and HCT-8 cells was investigated, using Transwell assays. In addition, to evaluate whether the anticancer properties of UA were mediated by the regulation of a double-negative feedback loop consisting of the transforming growth factor-β1 (TGF-β1)/zinc finger E-box-binding homeobox (ZEB1) pathway and microRNA (miR)-200a/b/c, reverse transcription-quantitative PCR and western blot analysis were performed. The results indicated that UA treatment significantly suppressed cellular growth, migration and invasion in HCT116 and HCT-8 cells in a dose-dependent manner. Furthermore, following UA treatment, several crucial mediators of the TGF-β1 signaling pathway, including TGF-β1, phosphorylated (p)-Smad2/3, p-focal adhesion kinase and ZEB1, were significantly downregulated in the HCT116 and HCT-8 cell lines compared with the control group. Furthermore, the ratio of N-cadherin/E-cadherin, two proteins directly downstream of the TGF-β1 signaling pathway, was found to be downregulated in UA treated CRC cells. Finally, UA significantly upregulated miR200a/b/c, with miR-200c exhibiting the highest increase in expression levels following UA treatment. Collectively, the present study suggested that inhibition of CRC cell invasion by UA occurred via regulation of the TGF-β1/ZEB1/miR-200c signaling network, which may be one of the mechanisms by which UA appears to be an effective therapeutic agent against colon cancer.
format Online
Article
Text
id pubmed-6676672
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-66766722019-08-26 Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-β1/ZEB1/miR-200c signaling pathway Zhang, Ling Cai, Qiao-Yan Liu, Jianxin Peng, Jun Chen, You-Qin Sferra, Thomas J. Lin, Jiu-Mao Oncol Lett Articles Ursolic acid (UA) is a biologically active compound, commonly used in traditional Chinese medicine (TCM). It has been reported to exhibit strong anticancer properties against a variety of cancers. Our previous studies showed that UA promoted apoptosis in colorectal cancer (CRC) cells and inhibited cellular proliferation and angiogenesis. However, the effect and underlying molecular mechanism of UA in CRC progression remain unclear. In the present study, the role of UA in suppressing the migration and invasion of human colon cancer HCT116 and HCT-8 cells was investigated, using Transwell assays. In addition, to evaluate whether the anticancer properties of UA were mediated by the regulation of a double-negative feedback loop consisting of the transforming growth factor-β1 (TGF-β1)/zinc finger E-box-binding homeobox (ZEB1) pathway and microRNA (miR)-200a/b/c, reverse transcription-quantitative PCR and western blot analysis were performed. The results indicated that UA treatment significantly suppressed cellular growth, migration and invasion in HCT116 and HCT-8 cells in a dose-dependent manner. Furthermore, following UA treatment, several crucial mediators of the TGF-β1 signaling pathway, including TGF-β1, phosphorylated (p)-Smad2/3, p-focal adhesion kinase and ZEB1, were significantly downregulated in the HCT116 and HCT-8 cell lines compared with the control group. Furthermore, the ratio of N-cadherin/E-cadherin, two proteins directly downstream of the TGF-β1 signaling pathway, was found to be downregulated in UA treated CRC cells. Finally, UA significantly upregulated miR200a/b/c, with miR-200c exhibiting the highest increase in expression levels following UA treatment. Collectively, the present study suggested that inhibition of CRC cell invasion by UA occurred via regulation of the TGF-β1/ZEB1/miR-200c signaling network, which may be one of the mechanisms by which UA appears to be an effective therapeutic agent against colon cancer. D.A. Spandidos 2019-09 2019-07-11 /pmc/articles/PMC6676672/ /pubmed/31452805 http://dx.doi.org/10.3892/ol.2019.10604 Text en Copyright: © Zhang 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
Zhang, Ling
Cai, Qiao-Yan
Liu, Jianxin
Peng, Jun
Chen, You-Qin
Sferra, Thomas J.
Lin, Jiu-Mao
Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-β1/ZEB1/miR-200c signaling pathway
title Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-β1/ZEB1/miR-200c signaling pathway
title_full Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-β1/ZEB1/miR-200c signaling pathway
title_fullStr Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-β1/ZEB1/miR-200c signaling pathway
title_full_unstemmed Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-β1/ZEB1/miR-200c signaling pathway
title_short Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-β1/ZEB1/miR-200c signaling pathway
title_sort ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the tgf-β1/zeb1/mir-200c signaling pathway
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6676672/
https://www.ncbi.nlm.nih.gov/pubmed/31452805
http://dx.doi.org/10.3892/ol.2019.10604
work_keys_str_mv AT zhangling ursolicacidsuppressestheinvasivepotentialofcolorectalcancercellsbyregulatingthetgfb1zeb1mir200csignalingpathway
AT caiqiaoyan ursolicacidsuppressestheinvasivepotentialofcolorectalcancercellsbyregulatingthetgfb1zeb1mir200csignalingpathway
AT liujianxin ursolicacidsuppressestheinvasivepotentialofcolorectalcancercellsbyregulatingthetgfb1zeb1mir200csignalingpathway
AT pengjun ursolicacidsuppressestheinvasivepotentialofcolorectalcancercellsbyregulatingthetgfb1zeb1mir200csignalingpathway
AT chenyouqin ursolicacidsuppressestheinvasivepotentialofcolorectalcancercellsbyregulatingthetgfb1zeb1mir200csignalingpathway
AT sferrathomasj ursolicacidsuppressestheinvasivepotentialofcolorectalcancercellsbyregulatingthetgfb1zeb1mir200csignalingpathway
AT linjiumao ursolicacidsuppressestheinvasivepotentialofcolorectalcancercellsbyregulatingthetgfb1zeb1mir200csignalingpathway