Inhibition of breast cancer metastasis suppressor 1 promotes a mesenchymal phenotype in lung epithelial cells that express oncogenic K-RasV12 and loss of p53.

Expression of the breast cancer metastasis suppressor 1 (BRMS1) protein is dramatically reduced in non-small cell lung cancer (NSCLC) cells and in primary human tumors. Although BRMS1 is a known suppressor of metastasis, the mechanisms through which BRMS1 functions to regulate cell migration and inv...

Full description

Bibliographic Details
Main Authors: Emily H Hall, Yuan Liu, Aizhen Xiao, Lisa Shock, David L Brautigan, Marty W Mayo, Prasad S Adusumilli, David R Jones
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2014-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3999110?pdf=render
id doaj-ef5b830b4b08433dabc7f4d8c047e57e
record_format Article
spelling doaj-ef5b830b4b08433dabc7f4d8c047e57e2020-11-24T21:34:28ZengPublic Library of Science (PLoS)PLoS ONE1932-62032014-01-0194e9586910.1371/journal.pone.0095869Inhibition of breast cancer metastasis suppressor 1 promotes a mesenchymal phenotype in lung epithelial cells that express oncogenic K-RasV12 and loss of p53.Emily H HallYuan LiuAizhen XiaoLisa ShockDavid L BrautiganMarty W MayoPrasad S AdusumilliDavid R JonesExpression of the breast cancer metastasis suppressor 1 (BRMS1) protein is dramatically reduced in non-small cell lung cancer (NSCLC) cells and in primary human tumors. Although BRMS1 is a known suppressor of metastasis, the mechanisms through which BRMS1 functions to regulate cell migration and invasion in response to specific NSCLC driver mutations are poorly understood. To experimentally address this, we utilized immortalized human bronchial epithelial cells in which p53 was knocked down in the presence of oncogenic K-RasV12 (HBEC3-p53KD-K-RasV12). These genetic alterations are commonly found in NSCLC and are associated with a poor prognosis. To determine the importance of BRMS1 for cytoskeletal function, cell migration and invasion in our model system we stably knocked down BRMS1. Here, we report that loss of BRMS1 in HBEC3-p53KD-K-RasV12 cells results in a dramatic increase in cell migration and invasion compared to controls that expressed BRMS1. Moreover, the loss of BRMS1 resulted in additional morphological changes including F-actin re-distribution, paxillin accumulation at the leading edge of the lamellapodium, and cellular shape changes resembling mesenchymal phenotypes. Importantly, re-expression of BRMS1 restores, in part, cell migration and invasion; however it does not fully reestablish the epithelial phenotype. These finding suggests that loss of BRMS1 results in a permanent, largely irreversible, mesenchymal phenotype associated with increased cell migration and invasion. Collectively, in NSCLC cells without p53 and expression of oncogenic K-Ras our study identifies BRMS1 as a key regulator required to maintain a cellular morphology and cytoskeletal architecture consistent with an epithelial phenotype.http://europepmc.org/articles/PMC3999110?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Emily H Hall
Yuan Liu
Aizhen Xiao
Lisa Shock
David L Brautigan
Marty W Mayo
Prasad S Adusumilli
David R Jones
spellingShingle Emily H Hall
Yuan Liu
Aizhen Xiao
Lisa Shock
David L Brautigan
Marty W Mayo
Prasad S Adusumilli
David R Jones
Inhibition of breast cancer metastasis suppressor 1 promotes a mesenchymal phenotype in lung epithelial cells that express oncogenic K-RasV12 and loss of p53.
PLoS ONE
author_facet Emily H Hall
Yuan Liu
Aizhen Xiao
Lisa Shock
David L Brautigan
Marty W Mayo
Prasad S Adusumilli
David R Jones
author_sort Emily H Hall
title Inhibition of breast cancer metastasis suppressor 1 promotes a mesenchymal phenotype in lung epithelial cells that express oncogenic K-RasV12 and loss of p53.
title_short Inhibition of breast cancer metastasis suppressor 1 promotes a mesenchymal phenotype in lung epithelial cells that express oncogenic K-RasV12 and loss of p53.
title_full Inhibition of breast cancer metastasis suppressor 1 promotes a mesenchymal phenotype in lung epithelial cells that express oncogenic K-RasV12 and loss of p53.
title_fullStr Inhibition of breast cancer metastasis suppressor 1 promotes a mesenchymal phenotype in lung epithelial cells that express oncogenic K-RasV12 and loss of p53.
title_full_unstemmed Inhibition of breast cancer metastasis suppressor 1 promotes a mesenchymal phenotype in lung epithelial cells that express oncogenic K-RasV12 and loss of p53.
title_sort inhibition of breast cancer metastasis suppressor 1 promotes a mesenchymal phenotype in lung epithelial cells that express oncogenic k-rasv12 and loss of p53.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2014-01-01
description Expression of the breast cancer metastasis suppressor 1 (BRMS1) protein is dramatically reduced in non-small cell lung cancer (NSCLC) cells and in primary human tumors. Although BRMS1 is a known suppressor of metastasis, the mechanisms through which BRMS1 functions to regulate cell migration and invasion in response to specific NSCLC driver mutations are poorly understood. To experimentally address this, we utilized immortalized human bronchial epithelial cells in which p53 was knocked down in the presence of oncogenic K-RasV12 (HBEC3-p53KD-K-RasV12). These genetic alterations are commonly found in NSCLC and are associated with a poor prognosis. To determine the importance of BRMS1 for cytoskeletal function, cell migration and invasion in our model system we stably knocked down BRMS1. Here, we report that loss of BRMS1 in HBEC3-p53KD-K-RasV12 cells results in a dramatic increase in cell migration and invasion compared to controls that expressed BRMS1. Moreover, the loss of BRMS1 resulted in additional morphological changes including F-actin re-distribution, paxillin accumulation at the leading edge of the lamellapodium, and cellular shape changes resembling mesenchymal phenotypes. Importantly, re-expression of BRMS1 restores, in part, cell migration and invasion; however it does not fully reestablish the epithelial phenotype. These finding suggests that loss of BRMS1 results in a permanent, largely irreversible, mesenchymal phenotype associated with increased cell migration and invasion. Collectively, in NSCLC cells without p53 and expression of oncogenic K-Ras our study identifies BRMS1 as a key regulator required to maintain a cellular morphology and cytoskeletal architecture consistent with an epithelial phenotype.
url http://europepmc.org/articles/PMC3999110?pdf=render
work_keys_str_mv AT emilyhhall inhibitionofbreastcancermetastasissuppressor1promotesamesenchymalphenotypeinlungepithelialcellsthatexpressoncogenickrasv12andlossofp53
AT yuanliu inhibitionofbreastcancermetastasissuppressor1promotesamesenchymalphenotypeinlungepithelialcellsthatexpressoncogenickrasv12andlossofp53
AT aizhenxiao inhibitionofbreastcancermetastasissuppressor1promotesamesenchymalphenotypeinlungepithelialcellsthatexpressoncogenickrasv12andlossofp53
AT lisashock inhibitionofbreastcancermetastasissuppressor1promotesamesenchymalphenotypeinlungepithelialcellsthatexpressoncogenickrasv12andlossofp53
AT davidlbrautigan inhibitionofbreastcancermetastasissuppressor1promotesamesenchymalphenotypeinlungepithelialcellsthatexpressoncogenickrasv12andlossofp53
AT martywmayo inhibitionofbreastcancermetastasissuppressor1promotesamesenchymalphenotypeinlungepithelialcellsthatexpressoncogenickrasv12andlossofp53
AT prasadsadusumilli inhibitionofbreastcancermetastasissuppressor1promotesamesenchymalphenotypeinlungepithelialcellsthatexpressoncogenickrasv12andlossofp53
AT davidrjones inhibitionofbreastcancermetastasissuppressor1promotesamesenchymalphenotypeinlungepithelialcellsthatexpressoncogenickrasv12andlossofp53
_version_ 1725949292046188544