Cargando…

PPARγ maintains ERBB2-positive breast cancer stem cells

Overexpression of the adverse prognostic marker ERBB2 occurs in 30% of breast cancers and is associated with aggressive disease and poor outcomes. Our recent findings have shown that NR1D1 and the peroxisome proliferator-activated receptor-γ (PPARγ)-binding protein (PBP) act through a common pathway...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, X, Sun, Y, Wong, J, Conklin, D S
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3898098/
https://www.ncbi.nlm.nih.gov/pubmed/23770845
http://dx.doi.org/10.1038/onc.2013.217
_version_ 1782300359606140928
author Wang, X
Sun, Y
Wong, J
Conklin, D S
author_facet Wang, X
Sun, Y
Wong, J
Conklin, D S
author_sort Wang, X
collection PubMed
description Overexpression of the adverse prognostic marker ERBB2 occurs in 30% of breast cancers and is associated with aggressive disease and poor outcomes. Our recent findings have shown that NR1D1 and the peroxisome proliferator-activated receptor-γ (PPARγ)-binding protein (PBP) act through a common pathway in upregulating several genes in the de novo fatty acid synthesis network, which is highly active in ERBB2-positive breast cancer cells. NR1D1 and PBP are functionally related to PPARγ, a well-established positive regulator of adipogenesis and lipid storage. Here, we report that inhibition of the PPARγ pathway reduces the aldehyde dehydrogenase (ALDH)-positive population in ERBB2-positive breast cancer cells. Results from in vitro tumorsphere formation assays demonstrate that the PPARγ antagonists GW9662 and T0070907 decrease tumorsphere formation in ERBB2-positive cells, but not other breast cells. We show that the mechanism by which GW9662 treatment causes a reduction in ALDH-positive population cells is partially due to ROS, as it can be rescued by treatment with N-acetyl-cysteine. Furthermore, global gene expression analyses show that GW9662 treatment suppresses the expression of several lipogenic genes, including ACLY, MIG12, FASN and NR1D1, and the stem-cell related genes KLF4 and ALDH in BT474 cells. Antagonist treatment also decreases the level of acetylation in histone 3 and histone 4 in BT474 cells, compared with MCF7 cells. In vivo, GW9662 pre-treatment inhibits the tumor-seeding ability of BT474 cells. Together, these results show that the PPARγ pathway is critical for the cancer stem cell properties of ERBB2-positive breast cancer cells.
format Online
Article
Text
id pubmed-3898098
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-38980982014-01-24 PPARγ maintains ERBB2-positive breast cancer stem cells Wang, X Sun, Y Wong, J Conklin, D S Oncogene Original Article Overexpression of the adverse prognostic marker ERBB2 occurs in 30% of breast cancers and is associated with aggressive disease and poor outcomes. Our recent findings have shown that NR1D1 and the peroxisome proliferator-activated receptor-γ (PPARγ)-binding protein (PBP) act through a common pathway in upregulating several genes in the de novo fatty acid synthesis network, which is highly active in ERBB2-positive breast cancer cells. NR1D1 and PBP are functionally related to PPARγ, a well-established positive regulator of adipogenesis and lipid storage. Here, we report that inhibition of the PPARγ pathway reduces the aldehyde dehydrogenase (ALDH)-positive population in ERBB2-positive breast cancer cells. Results from in vitro tumorsphere formation assays demonstrate that the PPARγ antagonists GW9662 and T0070907 decrease tumorsphere formation in ERBB2-positive cells, but not other breast cells. We show that the mechanism by which GW9662 treatment causes a reduction in ALDH-positive population cells is partially due to ROS, as it can be rescued by treatment with N-acetyl-cysteine. Furthermore, global gene expression analyses show that GW9662 treatment suppresses the expression of several lipogenic genes, including ACLY, MIG12, FASN and NR1D1, and the stem-cell related genes KLF4 and ALDH in BT474 cells. Antagonist treatment also decreases the level of acetylation in histone 3 and histone 4 in BT474 cells, compared with MCF7 cells. In vivo, GW9662 pre-treatment inhibits the tumor-seeding ability of BT474 cells. Together, these results show that the PPARγ pathway is critical for the cancer stem cell properties of ERBB2-positive breast cancer cells. Nature Publishing Group 2013-12-05 2013-06-17 /pmc/articles/PMC3898098/ /pubmed/23770845 http://dx.doi.org/10.1038/onc.2013.217 Text en Copyright © 2013 Macmillan Publishers Limited http://creativecommons.org/licenses/by-nc-nd/3.0/ This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivs 3.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/3.0/
spellingShingle Original Article
Wang, X
Sun, Y
Wong, J
Conklin, D S
PPARγ maintains ERBB2-positive breast cancer stem cells
title PPARγ maintains ERBB2-positive breast cancer stem cells
title_full PPARγ maintains ERBB2-positive breast cancer stem cells
title_fullStr PPARγ maintains ERBB2-positive breast cancer stem cells
title_full_unstemmed PPARγ maintains ERBB2-positive breast cancer stem cells
title_short PPARγ maintains ERBB2-positive breast cancer stem cells
title_sort pparγ maintains erbb2-positive breast cancer stem cells
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3898098/
https://www.ncbi.nlm.nih.gov/pubmed/23770845
http://dx.doi.org/10.1038/onc.2013.217
work_keys_str_mv AT wangx ppargmaintainserbb2positivebreastcancerstemcells
AT suny ppargmaintainserbb2positivebreastcancerstemcells
AT wongj ppargmaintainserbb2positivebreastcancerstemcells
AT conklinds ppargmaintainserbb2positivebreastcancerstemcells