Cargando…

Bronchial airway gene expression signatures in mouse lung squamous cell carcinoma and their modulation by cancer chemopreventive agents

Due to exposure to environmental toxicants, a “field cancerization” effect occurs in the lung resulting in the development of a field of initiated but morphologically normal appearing cells in the damaged epithelium of bronchial airways with dysregulated gene expression patterns. Using a mouse model...

Descripción completa

Detalles Bibliográficos
Autores principales: Xiong, Donghai, Pan, Jing, Zhang, Qi, Szabo, Eva, Miller, Mark Steven, Lubet, Ronald A., You, Ming, Wang, Yian
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals LLC 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5386655/
https://www.ncbi.nlm.nih.gov/pubmed/27935865
http://dx.doi.org/10.18632/oncotarget.13806
_version_ 1782520810730160128
author Xiong, Donghai
Pan, Jing
Zhang, Qi
Szabo, Eva
Miller, Mark Steven
Lubet, Ronald A.
You, Ming
Wang, Yian
author_facet Xiong, Donghai
Pan, Jing
Zhang, Qi
Szabo, Eva
Miller, Mark Steven
Lubet, Ronald A.
You, Ming
Wang, Yian
author_sort Xiong, Donghai
collection PubMed
description Due to exposure to environmental toxicants, a “field cancerization” effect occurs in the lung resulting in the development of a field of initiated but morphologically normal appearing cells in the damaged epithelium of bronchial airways with dysregulated gene expression patterns. Using a mouse model of lung squamous cell carcinoma (SCC), we performed transcriptome sequencing (RNA-Seq) to profile bronchial airway gene expression and found activation of the PI3K and Myc signaling networks in cytologically normal bronchial airway epithelial cells of mice with preneopastic lung SCC lesions, which was reversed by treatment with the PI3K Inhibitor XL-147 and pioglitazone, respectively. Activated MYC signaling was also present in premalignant and tumor tissues from human lung SCC patients. In addition, we identified a key microRNA, mmu-miR-449c-5p, whose suppression significantly up-regulated Myc expression in the normal bronchial airway epithelial cells of mice with early stage SCC lesions. We developed a novel bronchial genomic classifier in mice and validated it in humans. In the classifier, Ppbp (pro-platelet basic protein) was overexpressed 115 fold in the bronchial airways of mice with preneoplastic lung SCC lesions. This is the first report that demonstrates Ppbp as a novel biomarker in the bronchial airway for lung cancer diagnosis.
format Online
Article
Text
id pubmed-5386655
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Impact Journals LLC
record_format MEDLINE/PubMed
spelling pubmed-53866552017-04-26 Bronchial airway gene expression signatures in mouse lung squamous cell carcinoma and their modulation by cancer chemopreventive agents Xiong, Donghai Pan, Jing Zhang, Qi Szabo, Eva Miller, Mark Steven Lubet, Ronald A. You, Ming Wang, Yian Oncotarget Research Paper Due to exposure to environmental toxicants, a “field cancerization” effect occurs in the lung resulting in the development of a field of initiated but morphologically normal appearing cells in the damaged epithelium of bronchial airways with dysregulated gene expression patterns. Using a mouse model of lung squamous cell carcinoma (SCC), we performed transcriptome sequencing (RNA-Seq) to profile bronchial airway gene expression and found activation of the PI3K and Myc signaling networks in cytologically normal bronchial airway epithelial cells of mice with preneopastic lung SCC lesions, which was reversed by treatment with the PI3K Inhibitor XL-147 and pioglitazone, respectively. Activated MYC signaling was also present in premalignant and tumor tissues from human lung SCC patients. In addition, we identified a key microRNA, mmu-miR-449c-5p, whose suppression significantly up-regulated Myc expression in the normal bronchial airway epithelial cells of mice with early stage SCC lesions. We developed a novel bronchial genomic classifier in mice and validated it in humans. In the classifier, Ppbp (pro-platelet basic protein) was overexpressed 115 fold in the bronchial airways of mice with preneoplastic lung SCC lesions. This is the first report that demonstrates Ppbp as a novel biomarker in the bronchial airway for lung cancer diagnosis. Impact Journals LLC 2016-12-07 /pmc/articles/PMC5386655/ /pubmed/27935865 http://dx.doi.org/10.18632/oncotarget.13806 Text en Copyright: © 2017 Xiong 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, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Xiong, Donghai
Pan, Jing
Zhang, Qi
Szabo, Eva
Miller, Mark Steven
Lubet, Ronald A.
You, Ming
Wang, Yian
Bronchial airway gene expression signatures in mouse lung squamous cell carcinoma and their modulation by cancer chemopreventive agents
title Bronchial airway gene expression signatures in mouse lung squamous cell carcinoma and their modulation by cancer chemopreventive agents
title_full Bronchial airway gene expression signatures in mouse lung squamous cell carcinoma and their modulation by cancer chemopreventive agents
title_fullStr Bronchial airway gene expression signatures in mouse lung squamous cell carcinoma and their modulation by cancer chemopreventive agents
title_full_unstemmed Bronchial airway gene expression signatures in mouse lung squamous cell carcinoma and their modulation by cancer chemopreventive agents
title_short Bronchial airway gene expression signatures in mouse lung squamous cell carcinoma and their modulation by cancer chemopreventive agents
title_sort bronchial airway gene expression signatures in mouse lung squamous cell carcinoma and their modulation by cancer chemopreventive agents
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5386655/
https://www.ncbi.nlm.nih.gov/pubmed/27935865
http://dx.doi.org/10.18632/oncotarget.13806
work_keys_str_mv AT xiongdonghai bronchialairwaygeneexpressionsignaturesinmouselungsquamouscellcarcinomaandtheirmodulationbycancerchemopreventiveagents
AT panjing bronchialairwaygeneexpressionsignaturesinmouselungsquamouscellcarcinomaandtheirmodulationbycancerchemopreventiveagents
AT zhangqi bronchialairwaygeneexpressionsignaturesinmouselungsquamouscellcarcinomaandtheirmodulationbycancerchemopreventiveagents
AT szaboeva bronchialairwaygeneexpressionsignaturesinmouselungsquamouscellcarcinomaandtheirmodulationbycancerchemopreventiveagents
AT millermarksteven bronchialairwaygeneexpressionsignaturesinmouselungsquamouscellcarcinomaandtheirmodulationbycancerchemopreventiveagents
AT lubetronalda bronchialairwaygeneexpressionsignaturesinmouselungsquamouscellcarcinomaandtheirmodulationbycancerchemopreventiveagents
AT youming bronchialairwaygeneexpressionsignaturesinmouselungsquamouscellcarcinomaandtheirmodulationbycancerchemopreventiveagents
AT wangyian bronchialairwaygeneexpressionsignaturesinmouselungsquamouscellcarcinomaandtheirmodulationbycancerchemopreventiveagents