Cargando…

MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells

MiR-29b has been reported to be both a suppressor and a promoter in breast cancer (BC) cells proliferation and metastasis. Significant efforts have been made to explain the seemingly contradictory effects of miR-29b on BC, but no answer has yet been clearly verified. In this study, we overexpressed...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Hua, An, Xinglan, Yu, Hao, Zhang, Sheng, Tang, Bo, Zhang, Xueming, Li, Ziyi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5731940/
https://www.ncbi.nlm.nih.gov/pubmed/29254230
http://dx.doi.org/10.18632/oncotarget.22183
_version_ 1783286595517939712
author Wang, Hua
An, Xinglan
Yu, Hao
Zhang, Sheng
Tang, Bo
Zhang, Xueming
Li, Ziyi
author_facet Wang, Hua
An, Xinglan
Yu, Hao
Zhang, Sheng
Tang, Bo
Zhang, Xueming
Li, Ziyi
author_sort Wang, Hua
collection PubMed
description MiR-29b has been reported to be both a suppressor and a promoter in breast cancer (BC) cells proliferation and metastasis. Significant efforts have been made to explain the seemingly contradictory effects of miR-29b on BC, but no answer has yet been clearly verified. In this study, we overexpressed and knocked down miR-29b in BC cell lines, modulated expression of its downstream target gene TET1 and downregulated a downstream target gene of TET1, ZEB2, to explore the regulatory mechanism of miR-29b in BC cell proliferation, migration and epithelial-mesenchymal transition (EMT). Our results showed lower expression of miR-29b in BC samples and cell lines. Functional assays showed that miR-29b overexpression resulted in a higher cell proliferation, greater colony formation, higher migration rate and EMT. A dual luciferase assay identified TET1 as a direct target of miR-29b. As the promoting effects of miR-29b in the proliferation and metastasis of MDA-MB-231 and MCF-7, knockdown of TET1 also led to increased proliferation, colony formation, invasion and EMT. Further, we found that TET1 bound to the promoter of ZEB2, and siTET1 enhanced ZEB2 expression. Disruption of ZEB2 expression inhibited BC cells proliferation, colony formation and invasion. Our results establish the miR-29b/TET1/ZEB2 pathway in BC cell proliferation, migration and provide a theoretical basis for further research on the molecular mechanisms and new clinical treatments for BC.
format Online
Article
Text
id pubmed-5731940
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-57319402017-12-17 MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells Wang, Hua An, Xinglan Yu, Hao Zhang, Sheng Tang, Bo Zhang, Xueming Li, Ziyi Oncotarget Research Paper MiR-29b has been reported to be both a suppressor and a promoter in breast cancer (BC) cells proliferation and metastasis. Significant efforts have been made to explain the seemingly contradictory effects of miR-29b on BC, but no answer has yet been clearly verified. In this study, we overexpressed and knocked down miR-29b in BC cell lines, modulated expression of its downstream target gene TET1 and downregulated a downstream target gene of TET1, ZEB2, to explore the regulatory mechanism of miR-29b in BC cell proliferation, migration and epithelial-mesenchymal transition (EMT). Our results showed lower expression of miR-29b in BC samples and cell lines. Functional assays showed that miR-29b overexpression resulted in a higher cell proliferation, greater colony formation, higher migration rate and EMT. A dual luciferase assay identified TET1 as a direct target of miR-29b. As the promoting effects of miR-29b in the proliferation and metastasis of MDA-MB-231 and MCF-7, knockdown of TET1 also led to increased proliferation, colony formation, invasion and EMT. Further, we found that TET1 bound to the promoter of ZEB2, and siTET1 enhanced ZEB2 expression. Disruption of ZEB2 expression inhibited BC cells proliferation, colony formation and invasion. Our results establish the miR-29b/TET1/ZEB2 pathway in BC cell proliferation, migration and provide a theoretical basis for further research on the molecular mechanisms and new clinical treatments for BC. Impact Journals LLC 2017-10-31 /pmc/articles/PMC5731940/ /pubmed/29254230 http://dx.doi.org/10.18632/oncotarget.22183 Text en Copyright: © 2017 Wang et al. http://creativecommons.org/licenses/by/3.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (http://creativecommons.org/licenses/by/3.0/) (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Wang, Hua
An, Xinglan
Yu, Hao
Zhang, Sheng
Tang, Bo
Zhang, Xueming
Li, Ziyi
MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells
title MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells
title_full MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells
title_fullStr MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells
title_full_unstemmed MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells
title_short MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells
title_sort mir-29b/tet1/zeb2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5731940/
https://www.ncbi.nlm.nih.gov/pubmed/29254230
http://dx.doi.org/10.18632/oncotarget.22183
work_keys_str_mv AT wanghua mir29btet1zeb2signalingaxisregulatesmetastaticpropertiesandepithelialmesenchymaltransitioninbreastcancercells
AT anxinglan mir29btet1zeb2signalingaxisregulatesmetastaticpropertiesandepithelialmesenchymaltransitioninbreastcancercells
AT yuhao mir29btet1zeb2signalingaxisregulatesmetastaticpropertiesandepithelialmesenchymaltransitioninbreastcancercells
AT zhangsheng mir29btet1zeb2signalingaxisregulatesmetastaticpropertiesandepithelialmesenchymaltransitioninbreastcancercells
AT tangbo mir29btet1zeb2signalingaxisregulatesmetastaticpropertiesandepithelialmesenchymaltransitioninbreastcancercells
AT zhangxueming mir29btet1zeb2signalingaxisregulatesmetastaticpropertiesandepithelialmesenchymaltransitioninbreastcancercells
AT liziyi mir29btet1zeb2signalingaxisregulatesmetastaticpropertiesandepithelialmesenchymaltransitioninbreastcancercells