Cargando…

miR-29b defines the pro-/anti-proliferative effects of S100A7 in breast cancer

INTRODUCTION: S100A7 (Psoriasin) is an inflammatory protein known to be upregulated in breast cancer. However, the role of S100A7 in breast cancer has been elusive, since both pro- and anti-proliferative roles have been reported in different types of breast cancer cells and animal models. To date, t...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhao, Helong, Wilkie, Tasha, Deol, Yadwinder, Sneh, Amita, Ganju, Akaansha, Basree, Mustafa, Nasser, Mohd W, Ganju, Ramesh K
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2015
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4314775/
https://www.ncbi.nlm.nih.gov/pubmed/25622979
http://dx.doi.org/10.1186/s12943-014-0275-z
_version_ 1782355365802803200
author Zhao, Helong
Wilkie, Tasha
Deol, Yadwinder
Sneh, Amita
Ganju, Akaansha
Basree, Mustafa
Nasser, Mohd W
Ganju, Ramesh K
author_facet Zhao, Helong
Wilkie, Tasha
Deol, Yadwinder
Sneh, Amita
Ganju, Akaansha
Basree, Mustafa
Nasser, Mohd W
Ganju, Ramesh K
author_sort Zhao, Helong
collection PubMed
description INTRODUCTION: S100A7 (Psoriasin) is an inflammatory protein known to be upregulated in breast cancer. However, the role of S100A7 in breast cancer has been elusive, since both pro- and anti-proliferative roles have been reported in different types of breast cancer cells and animal models. To date, the mechanism by which S100A7 differentially regulates breast cancer cell proliferation is still not clear. METHODS: We used Gene Functional Enrichment Analysis to search for the determining factor of S100A7 differential regulation. We confirmed the factor and elaborated its regulating mechanism using in vitro cell culture. We further verified the findings using xenografts of human breast cancer cells in nude mice. RESULTS: In the present study, we show that S100A7 significantly downregulates the expression of miR-29b in Estrogen Receptor (ER)-positive breast cancer cells (represented by MCF7), and significantly upregulates miR-29b in ER-negative cells (represented by MDA-MB-231). The differential regulation of miR-29b by S100A7 in ER-positive and ER-negative breast cancer is supported by the gene expression analysis of TCGA invasive breast cancer dataset. miR-29b transcription is inhibited by NF-κB, and NF-κB activation is differentially regulated by S100A7 in ER-positive and ER-negative breast cancer cells. This further leads to differential regulation of PI3K p85α and CDC42 expression, p53 activation and p53-associated anti-proliferative pathways. Reversing the S100A7-caused changes of miR-29b expression by transfecting exogenous miR-29b or miR-29b-Decoy can inhibit the effects of S100A7 on in vitro cell proliferation and tumor growth in nude mice. CONCLUSIONS: The distinct modulations of the NF-κB – miR-29b – p53 pathway make S100A7 an oncogene in ER-negative and a cancer-suppressing gene in ER-positive breast cancer cells, with miR-29b being the determining regulatory factor. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s12943-014-0275-z) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-4314775
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-43147752015-02-04 miR-29b defines the pro-/anti-proliferative effects of S100A7 in breast cancer Zhao, Helong Wilkie, Tasha Deol, Yadwinder Sneh, Amita Ganju, Akaansha Basree, Mustafa Nasser, Mohd W Ganju, Ramesh K Mol Cancer Research INTRODUCTION: S100A7 (Psoriasin) is an inflammatory protein known to be upregulated in breast cancer. However, the role of S100A7 in breast cancer has been elusive, since both pro- and anti-proliferative roles have been reported in different types of breast cancer cells and animal models. To date, the mechanism by which S100A7 differentially regulates breast cancer cell proliferation is still not clear. METHODS: We used Gene Functional Enrichment Analysis to search for the determining factor of S100A7 differential regulation. We confirmed the factor and elaborated its regulating mechanism using in vitro cell culture. We further verified the findings using xenografts of human breast cancer cells in nude mice. RESULTS: In the present study, we show that S100A7 significantly downregulates the expression of miR-29b in Estrogen Receptor (ER)-positive breast cancer cells (represented by MCF7), and significantly upregulates miR-29b in ER-negative cells (represented by MDA-MB-231). The differential regulation of miR-29b by S100A7 in ER-positive and ER-negative breast cancer is supported by the gene expression analysis of TCGA invasive breast cancer dataset. miR-29b transcription is inhibited by NF-κB, and NF-κB activation is differentially regulated by S100A7 in ER-positive and ER-negative breast cancer cells. This further leads to differential regulation of PI3K p85α and CDC42 expression, p53 activation and p53-associated anti-proliferative pathways. Reversing the S100A7-caused changes of miR-29b expression by transfecting exogenous miR-29b or miR-29b-Decoy can inhibit the effects of S100A7 on in vitro cell proliferation and tumor growth in nude mice. CONCLUSIONS: The distinct modulations of the NF-κB – miR-29b – p53 pathway make S100A7 an oncogene in ER-negative and a cancer-suppressing gene in ER-positive breast cancer cells, with miR-29b being the determining regulatory factor. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1186/s12943-014-0275-z) contains supplementary material, which is available to authorized users. BioMed Central 2015-01-27 /pmc/articles/PMC4314775/ /pubmed/25622979 http://dx.doi.org/10.1186/s12943-014-0275-z Text en © Zhao et al.; licensee BioMed Central. 2015 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. 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
Zhao, Helong
Wilkie, Tasha
Deol, Yadwinder
Sneh, Amita
Ganju, Akaansha
Basree, Mustafa
Nasser, Mohd W
Ganju, Ramesh K
miR-29b defines the pro-/anti-proliferative effects of S100A7 in breast cancer
title miR-29b defines the pro-/anti-proliferative effects of S100A7 in breast cancer
title_full miR-29b defines the pro-/anti-proliferative effects of S100A7 in breast cancer
title_fullStr miR-29b defines the pro-/anti-proliferative effects of S100A7 in breast cancer
title_full_unstemmed miR-29b defines the pro-/anti-proliferative effects of S100A7 in breast cancer
title_short miR-29b defines the pro-/anti-proliferative effects of S100A7 in breast cancer
title_sort mir-29b defines the pro-/anti-proliferative effects of s100a7 in breast cancer
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4314775/
https://www.ncbi.nlm.nih.gov/pubmed/25622979
http://dx.doi.org/10.1186/s12943-014-0275-z
work_keys_str_mv AT zhaohelong mir29bdefinestheproantiproliferativeeffectsofs100a7inbreastcancer
AT wilkietasha mir29bdefinestheproantiproliferativeeffectsofs100a7inbreastcancer
AT deolyadwinder mir29bdefinestheproantiproliferativeeffectsofs100a7inbreastcancer
AT snehamita mir29bdefinestheproantiproliferativeeffectsofs100a7inbreastcancer
AT ganjuakaansha mir29bdefinestheproantiproliferativeeffectsofs100a7inbreastcancer
AT basreemustafa mir29bdefinestheproantiproliferativeeffectsofs100a7inbreastcancer
AT nassermohdw mir29bdefinestheproantiproliferativeeffectsofs100a7inbreastcancer
AT ganjurameshk mir29bdefinestheproantiproliferativeeffectsofs100a7inbreastcancer