Cargando…

HDAC Inhibition Induces Cell Cycle Arrest and Mesenchymal-Epithelial Transition in a Novel Pleural-Effusion Derived Uterine Carcinosarcoma Cell Line

Objective: Uterine carcinosarcoma (UCS) is a rare but highly aggressive malignancy with biphasic growth pattern. This morphology can be attributed to epithelial-mesenchymal transition (EMT) that often associates with tumor invasion and metastasis. Accordingly, we analyzed a novel patient-derived pre...

Descripción completa

Detalles Bibliográficos
Autores principales: Stockhammer, Paul, Okumus, Özlem, Hegedus, Luca, Rittler, Dominika, Ploenes, Till, Herold, Thomas, Kalbourtzis, Stavros, Bankfalvi, Agnes, Sucker, Antje, Kimmig, Rainer, Aigner, Clemens, Hegedus, Balazs
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8262245/
https://www.ncbi.nlm.nih.gov/pubmed/34257602
http://dx.doi.org/10.3389/pore.2021.636088
_version_ 1783719154434441216
author Stockhammer, Paul
Okumus, Özlem
Hegedus, Luca
Rittler, Dominika
Ploenes, Till
Herold, Thomas
Kalbourtzis, Stavros
Bankfalvi, Agnes
Sucker, Antje
Kimmig, Rainer
Aigner, Clemens
Hegedus, Balazs
author_facet Stockhammer, Paul
Okumus, Özlem
Hegedus, Luca
Rittler, Dominika
Ploenes, Till
Herold, Thomas
Kalbourtzis, Stavros
Bankfalvi, Agnes
Sucker, Antje
Kimmig, Rainer
Aigner, Clemens
Hegedus, Balazs
author_sort Stockhammer, Paul
collection PubMed
description Objective: Uterine carcinosarcoma (UCS) is a rare but highly aggressive malignancy with biphasic growth pattern. This morphology can be attributed to epithelial-mesenchymal transition (EMT) that often associates with tumor invasion and metastasis. Accordingly, we analyzed a novel patient-derived preclinical model to explore whether EMT is a potential target in UCS. Methods: A novel UCS cell line (PF338) was established from the malignant pleural effusion of a 59-year-old patient at time of disease progression. Immunohistochemistry was performed in primary and metastatic tumor lesions. Oncogenic mutations were identified by next-generation sequencing. Viability assays and cell cycle analyses were used to test in vitro sensitivity to different standard and novel treatments. E-cadherin, β-catenin and pSMAD2 expressions were measured by immunoblot. Results: Whereas immunohistochemistry of the metastatic tumor showed a predominantly sarcomatous vimentin positive tumor that has lost E-cadherin expression, PF338 cells demonstrated biphasic growth and carried mutations in KRAS, PIK3CA, PTEN and ARID1A. PF338 tumor cells were resistant to MEK- and TGF-β signaling-inhibition but sensitive to PIK3CA- and PARP-inhibition and first-line chemotherapeutics. Strikingly, histone deacetylase (HDAC) inhibition markedly reduced cell viability by inducing a dose-dependent G0/1 arrest and led to mesenchymal-epithelial transition as evidenced by morphological change and increased E-cadherin and β-catenin expression. Conclusions: Our data suggest that HDAC inhibition is effective in a novel UCS cell line by interfering with both viability and differentiation. These findings emphasize the dynamic manner of EMT/MET and epigenetics and the importance of molecular profiling to pave the way for novel therapies in UCS.
format Online
Article
Text
id pubmed-8262245
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-82622452021-07-12 HDAC Inhibition Induces Cell Cycle Arrest and Mesenchymal-Epithelial Transition in a Novel Pleural-Effusion Derived Uterine Carcinosarcoma Cell Line Stockhammer, Paul Okumus, Özlem Hegedus, Luca Rittler, Dominika Ploenes, Till Herold, Thomas Kalbourtzis, Stavros Bankfalvi, Agnes Sucker, Antje Kimmig, Rainer Aigner, Clemens Hegedus, Balazs Pathol Oncol Res Society Journal Archive Objective: Uterine carcinosarcoma (UCS) is a rare but highly aggressive malignancy with biphasic growth pattern. This morphology can be attributed to epithelial-mesenchymal transition (EMT) that often associates with tumor invasion and metastasis. Accordingly, we analyzed a novel patient-derived preclinical model to explore whether EMT is a potential target in UCS. Methods: A novel UCS cell line (PF338) was established from the malignant pleural effusion of a 59-year-old patient at time of disease progression. Immunohistochemistry was performed in primary and metastatic tumor lesions. Oncogenic mutations were identified by next-generation sequencing. Viability assays and cell cycle analyses were used to test in vitro sensitivity to different standard and novel treatments. E-cadherin, β-catenin and pSMAD2 expressions were measured by immunoblot. Results: Whereas immunohistochemistry of the metastatic tumor showed a predominantly sarcomatous vimentin positive tumor that has lost E-cadherin expression, PF338 cells demonstrated biphasic growth and carried mutations in KRAS, PIK3CA, PTEN and ARID1A. PF338 tumor cells were resistant to MEK- and TGF-β signaling-inhibition but sensitive to PIK3CA- and PARP-inhibition and first-line chemotherapeutics. Strikingly, histone deacetylase (HDAC) inhibition markedly reduced cell viability by inducing a dose-dependent G0/1 arrest and led to mesenchymal-epithelial transition as evidenced by morphological change and increased E-cadherin and β-catenin expression. Conclusions: Our data suggest that HDAC inhibition is effective in a novel UCS cell line by interfering with both viability and differentiation. These findings emphasize the dynamic manner of EMT/MET and epigenetics and the importance of molecular profiling to pave the way for novel therapies in UCS. Frontiers Media S.A. 2021-03-26 /pmc/articles/PMC8262245/ /pubmed/34257602 http://dx.doi.org/10.3389/pore.2021.636088 Text en Copyright © 2021 Stockhammer, Okumus, Hegedus, Rittler, Ploenes, Herold, Kalbourtzis, Bankfalvi, Sucker, Kimmig, Aigner and Hegedus. 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
Stockhammer, Paul
Okumus, Özlem
Hegedus, Luca
Rittler, Dominika
Ploenes, Till
Herold, Thomas
Kalbourtzis, Stavros
Bankfalvi, Agnes
Sucker, Antje
Kimmig, Rainer
Aigner, Clemens
Hegedus, Balazs
HDAC Inhibition Induces Cell Cycle Arrest and Mesenchymal-Epithelial Transition in a Novel Pleural-Effusion Derived Uterine Carcinosarcoma Cell Line
title HDAC Inhibition Induces Cell Cycle Arrest and Mesenchymal-Epithelial Transition in a Novel Pleural-Effusion Derived Uterine Carcinosarcoma Cell Line
title_full HDAC Inhibition Induces Cell Cycle Arrest and Mesenchymal-Epithelial Transition in a Novel Pleural-Effusion Derived Uterine Carcinosarcoma Cell Line
title_fullStr HDAC Inhibition Induces Cell Cycle Arrest and Mesenchymal-Epithelial Transition in a Novel Pleural-Effusion Derived Uterine Carcinosarcoma Cell Line
title_full_unstemmed HDAC Inhibition Induces Cell Cycle Arrest and Mesenchymal-Epithelial Transition in a Novel Pleural-Effusion Derived Uterine Carcinosarcoma Cell Line
title_short HDAC Inhibition Induces Cell Cycle Arrest and Mesenchymal-Epithelial Transition in a Novel Pleural-Effusion Derived Uterine Carcinosarcoma Cell Line
title_sort hdac inhibition induces cell cycle arrest and mesenchymal-epithelial transition in a novel pleural-effusion derived uterine carcinosarcoma cell line
topic Society Journal Archive
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8262245/
https://www.ncbi.nlm.nih.gov/pubmed/34257602
http://dx.doi.org/10.3389/pore.2021.636088
work_keys_str_mv AT stockhammerpaul hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT okumusozlem hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT hegedusluca hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT rittlerdominika hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT ploenestill hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT heroldthomas hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT kalbourtzisstavros hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT bankfalviagnes hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT suckerantje hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT kimmigrainer hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT aignerclemens hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline
AT hegedusbalazs hdacinhibitioninducescellcyclearrestandmesenchymalepithelialtransitioninanovelpleuraleffusionderiveduterinecarcinosarcomacellline