Cargando…

Notch1 Deficiency Induces Tumor Cell Accumulation Inside the Bronchiolar Lumen and Increases TAZ Expression in an Autochthonous Kras (LSL-G12V) Driven Lung Cancer Mouse Model

Purpose: Abrogation of Notch signaling, which is pivotal for lung development and pulmonary epithelial cell fate decisions was shown to be involved in the aggressiveness and the differentiation of lung carcinomas. Additionally, the transcription factors YAP and TAZ which are involved in the Hippo pa...

Descripción completa

Detalles Bibliográficos
Autores principales: Meder, Lydia, Florin, Alexandra, Ozretić, Luka, Nill, Marieke, Koker, Mirjam, Meemboor, Sonja, Radtke, Freddy, Diehl, Linda, Ullrich, Roland T., Odenthal, Margarete, Büttner, Reinhard, Heukamp, Lukas C.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8262161/
https://www.ncbi.nlm.nih.gov/pubmed/34257546
http://dx.doi.org/10.3389/pore.2021.596522
_version_ 1783719135749865472
author Meder, Lydia
Florin, Alexandra
Ozretić, Luka
Nill, Marieke
Koker, Mirjam
Meemboor, Sonja
Radtke, Freddy
Diehl, Linda
Ullrich, Roland T.
Odenthal, Margarete
Büttner, Reinhard
Heukamp, Lukas C.
author_facet Meder, Lydia
Florin, Alexandra
Ozretić, Luka
Nill, Marieke
Koker, Mirjam
Meemboor, Sonja
Radtke, Freddy
Diehl, Linda
Ullrich, Roland T.
Odenthal, Margarete
Büttner, Reinhard
Heukamp, Lukas C.
author_sort Meder, Lydia
collection PubMed
description Purpose: Abrogation of Notch signaling, which is pivotal for lung development and pulmonary epithelial cell fate decisions was shown to be involved in the aggressiveness and the differentiation of lung carcinomas. Additionally, the transcription factors YAP and TAZ which are involved in the Hippo pathway, were recently shown to be tightly linked with Notch signaling and to regulate the cell fate in epidermal stem cells. Thus, we aim to elucidate the effects of conditional Notch1 deficiency on carcinogenesis and TAZ expression in lung cancer. Methods: We investigated the effect of conditional Cre-recombinase mediated Notch1 knock-out on lung cancer cells in vivo using an autochthonous mouse model of lung adenocarcinomas driven by Kras (LSL-G12V) and comprehensive immunohistochemical analysis. In addition, we analyzed clinical samples and human lung cancer cell lines for TAZ expression and supported our findings by publicly available data from The Cancer Genome Atlas (TCGA). Results: In mice, we found induction of papillary adenocarcinomas and protrusions of tumor cells from the bronchiolar lining upon Notch1 deficiency. Moreover, the mutated Kras driven lung tumors with deleted Notch1 showed increased TAZ expression and focal nuclear translocation which was frequently observed in human pulmonary adenocarcinomas and squamous cell carcinomas of the lung, but not in small cell lung carcinomas. In addition, we used data from TCGA to show that putative inactivating NOTCH1 mutations co-occur with KRAS mutations and genomic amplifications in lung adenocarcinomas. Conclusion: Our in vivo study provides evidence that Notch1 deficiency in mutated Kras driven lung carcinomas contributes to lung carcinogenesis in a subgroup of patients by increasing TAZ expression who might benefit from TAZ signaling blockade.
format Online
Article
Text
id pubmed-8262161
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-82621612021-07-12 Notch1 Deficiency Induces Tumor Cell Accumulation Inside the Bronchiolar Lumen and Increases TAZ Expression in an Autochthonous Kras (LSL-G12V) Driven Lung Cancer Mouse Model Meder, Lydia Florin, Alexandra Ozretić, Luka Nill, Marieke Koker, Mirjam Meemboor, Sonja Radtke, Freddy Diehl, Linda Ullrich, Roland T. Odenthal, Margarete Büttner, Reinhard Heukamp, Lukas C. Pathol Oncol Res Society Journal Archive Purpose: Abrogation of Notch signaling, which is pivotal for lung development and pulmonary epithelial cell fate decisions was shown to be involved in the aggressiveness and the differentiation of lung carcinomas. Additionally, the transcription factors YAP and TAZ which are involved in the Hippo pathway, were recently shown to be tightly linked with Notch signaling and to regulate the cell fate in epidermal stem cells. Thus, we aim to elucidate the effects of conditional Notch1 deficiency on carcinogenesis and TAZ expression in lung cancer. Methods: We investigated the effect of conditional Cre-recombinase mediated Notch1 knock-out on lung cancer cells in vivo using an autochthonous mouse model of lung adenocarcinomas driven by Kras (LSL-G12V) and comprehensive immunohistochemical analysis. In addition, we analyzed clinical samples and human lung cancer cell lines for TAZ expression and supported our findings by publicly available data from The Cancer Genome Atlas (TCGA). Results: In mice, we found induction of papillary adenocarcinomas and protrusions of tumor cells from the bronchiolar lining upon Notch1 deficiency. Moreover, the mutated Kras driven lung tumors with deleted Notch1 showed increased TAZ expression and focal nuclear translocation which was frequently observed in human pulmonary adenocarcinomas and squamous cell carcinomas of the lung, but not in small cell lung carcinomas. In addition, we used data from TCGA to show that putative inactivating NOTCH1 mutations co-occur with KRAS mutations and genomic amplifications in lung adenocarcinomas. Conclusion: Our in vivo study provides evidence that Notch1 deficiency in mutated Kras driven lung carcinomas contributes to lung carcinogenesis in a subgroup of patients by increasing TAZ expression who might benefit from TAZ signaling blockade. Frontiers Media S.A. 2021-04-16 /pmc/articles/PMC8262161/ /pubmed/34257546 http://dx.doi.org/10.3389/pore.2021.596522 Text en Copyright © 2021 Meder, Florin, Ozretić, Nill, Koker, Meemboor, Radtke, Diehl, Ullrich, Odenthal, Büttner and Heukamp. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Society Journal Archive
Meder, Lydia
Florin, Alexandra
Ozretić, Luka
Nill, Marieke
Koker, Mirjam
Meemboor, Sonja
Radtke, Freddy
Diehl, Linda
Ullrich, Roland T.
Odenthal, Margarete
Büttner, Reinhard
Heukamp, Lukas C.
Notch1 Deficiency Induces Tumor Cell Accumulation Inside the Bronchiolar Lumen and Increases TAZ Expression in an Autochthonous Kras (LSL-G12V) Driven Lung Cancer Mouse Model
title Notch1 Deficiency Induces Tumor Cell Accumulation Inside the Bronchiolar Lumen and Increases TAZ Expression in an Autochthonous Kras (LSL-G12V) Driven Lung Cancer Mouse Model
title_full Notch1 Deficiency Induces Tumor Cell Accumulation Inside the Bronchiolar Lumen and Increases TAZ Expression in an Autochthonous Kras (LSL-G12V) Driven Lung Cancer Mouse Model
title_fullStr Notch1 Deficiency Induces Tumor Cell Accumulation Inside the Bronchiolar Lumen and Increases TAZ Expression in an Autochthonous Kras (LSL-G12V) Driven Lung Cancer Mouse Model
title_full_unstemmed Notch1 Deficiency Induces Tumor Cell Accumulation Inside the Bronchiolar Lumen and Increases TAZ Expression in an Autochthonous Kras (LSL-G12V) Driven Lung Cancer Mouse Model
title_short Notch1 Deficiency Induces Tumor Cell Accumulation Inside the Bronchiolar Lumen and Increases TAZ Expression in an Autochthonous Kras (LSL-G12V) Driven Lung Cancer Mouse Model
title_sort notch1 deficiency induces tumor cell accumulation inside the bronchiolar lumen and increases taz expression in an autochthonous kras (lsl-g12v) driven lung cancer mouse model
topic Society Journal Archive
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8262161/
https://www.ncbi.nlm.nih.gov/pubmed/34257546
http://dx.doi.org/10.3389/pore.2021.596522
work_keys_str_mv AT mederlydia notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT florinalexandra notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT ozreticluka notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT nillmarieke notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT kokermirjam notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT meemboorsonja notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT radtkefreddy notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT diehllinda notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT ullrichrolandt notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT odenthalmargarete notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT buttnerreinhard notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel
AT heukamplukasc notch1deficiencyinducestumorcellaccumulationinsidethebronchiolarlumenandincreasestazexpressioninanautochthonouskraslslg12vdrivenlungcancermousemodel