Cargando…

Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non‐small‐cell lung cancer cells

Recent discoveries have revealed that human cancer involves aberrant epigenetic alterations. We and others have previously shown that the histone methyltransferase EZH2, the catalytic subunit of polycomb repressive complex 2 (PRC2), is frequently overexpressed in non‐small‐cell lung cancer (NSCLC) a...

Descripción completa

Detalles Bibliográficos
Autores principales: Takashina, Taichi, Kinoshita, Ichiro, Kikuchi, Junko, Shimizu, Yasushi, Sakakibara‐Konishi, Jun, Oizumi, Satoshi, Nishimura, Masaharu, Dosaka‐Akita, Hirotoshi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4946723/
https://www.ncbi.nlm.nih.gov/pubmed/27116120
http://dx.doi.org/10.1111/cas.12957
_version_ 1782443063948345344
author Takashina, Taichi
Kinoshita, Ichiro
Kikuchi, Junko
Shimizu, Yasushi
Sakakibara‐Konishi, Jun
Oizumi, Satoshi
Nishimura, Masaharu
Dosaka‐Akita, Hirotoshi
author_facet Takashina, Taichi
Kinoshita, Ichiro
Kikuchi, Junko
Shimizu, Yasushi
Sakakibara‐Konishi, Jun
Oizumi, Satoshi
Nishimura, Masaharu
Dosaka‐Akita, Hirotoshi
author_sort Takashina, Taichi
collection PubMed
description Recent discoveries have revealed that human cancer involves aberrant epigenetic alterations. We and others have previously shown that the histone methyltransferase EZH2, the catalytic subunit of polycomb repressive complex 2 (PRC2), is frequently overexpressed in non‐small‐cell lung cancer (NSCLC) and that an EZH2 inhibitor, 3‐deazaneplanocin A, inhibits the proliferation of NSCLC cells. Transcriptional silencing by EZH2 was recently shown to be required for the activity of histone deacetylases (HDACs) that interact with another PRC2 protein, EED. To develop a more effective epigenetic therapy for NSCLC, we determined the effects of co‐treatment with 3‐deazaneplanocin A and the HDAC inhibitor vorinostat (SAHA) in NSCLC cells. The co‐treatment synergistically suppressed the proliferation of all tested NSCLC cell lines, regardless of their epidermal growth factor receptor (EGFR) status. The synergistic effect was associated with slightly decreased histone H3 lysine 27 trimethylation, modestly increased histone acetylation, and the depletion of EZH2 and other PRC2 proteins. The co‐treatment resulted in an accumulation of p27Kip1, decrease in cyclin A, and increased apoptotic fraction in an additive/synergistic manner. Interestingly, the co‐treatment strongly suppressed EGFR signaling, not only in EGFR‐wild‐type NSCLC cells, but also in EGFR‐mutant cells, mainly through dephosphorylation of EGFR. Furthermore, the co‐treatment suppressed the in vivo tumor growth of EGFR‐mutant, EGFR–tyrosine kinase‐resistant H1975 cells more effectively than did each agent alone, without visible toxicity. These results suggest that the combined pharmacological targeting of EZH2 and HDACs may provide more effective epigenetic therapeutics for NSCLC.
format Online
Article
Text
id pubmed-4946723
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-49467232016-07-27 Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non‐small‐cell lung cancer cells Takashina, Taichi Kinoshita, Ichiro Kikuchi, Junko Shimizu, Yasushi Sakakibara‐Konishi, Jun Oizumi, Satoshi Nishimura, Masaharu Dosaka‐Akita, Hirotoshi Cancer Sci Original Articles Recent discoveries have revealed that human cancer involves aberrant epigenetic alterations. We and others have previously shown that the histone methyltransferase EZH2, the catalytic subunit of polycomb repressive complex 2 (PRC2), is frequently overexpressed in non‐small‐cell lung cancer (NSCLC) and that an EZH2 inhibitor, 3‐deazaneplanocin A, inhibits the proliferation of NSCLC cells. Transcriptional silencing by EZH2 was recently shown to be required for the activity of histone deacetylases (HDACs) that interact with another PRC2 protein, EED. To develop a more effective epigenetic therapy for NSCLC, we determined the effects of co‐treatment with 3‐deazaneplanocin A and the HDAC inhibitor vorinostat (SAHA) in NSCLC cells. The co‐treatment synergistically suppressed the proliferation of all tested NSCLC cell lines, regardless of their epidermal growth factor receptor (EGFR) status. The synergistic effect was associated with slightly decreased histone H3 lysine 27 trimethylation, modestly increased histone acetylation, and the depletion of EZH2 and other PRC2 proteins. The co‐treatment resulted in an accumulation of p27Kip1, decrease in cyclin A, and increased apoptotic fraction in an additive/synergistic manner. Interestingly, the co‐treatment strongly suppressed EGFR signaling, not only in EGFR‐wild‐type NSCLC cells, but also in EGFR‐mutant cells, mainly through dephosphorylation of EGFR. Furthermore, the co‐treatment suppressed the in vivo tumor growth of EGFR‐mutant, EGFR–tyrosine kinase‐resistant H1975 cells more effectively than did each agent alone, without visible toxicity. These results suggest that the combined pharmacological targeting of EZH2 and HDACs may provide more effective epigenetic therapeutics for NSCLC. John Wiley and Sons Inc. 2016-06-13 2016-07 /pmc/articles/PMC4946723/ /pubmed/27116120 http://dx.doi.org/10.1111/cas.12957 Text en © 2016 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association. This is an open access article under the terms of the Creative Commons Attribution‐NonCommercial (http://creativecommons.org/licenses/by-nc/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.
spellingShingle Original Articles
Takashina, Taichi
Kinoshita, Ichiro
Kikuchi, Junko
Shimizu, Yasushi
Sakakibara‐Konishi, Jun
Oizumi, Satoshi
Nishimura, Masaharu
Dosaka‐Akita, Hirotoshi
Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non‐small‐cell lung cancer cells
title Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non‐small‐cell lung cancer cells
title_full Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non‐small‐cell lung cancer cells
title_fullStr Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non‐small‐cell lung cancer cells
title_full_unstemmed Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non‐small‐cell lung cancer cells
title_short Combined inhibition of EZH2 and histone deacetylases as a potential epigenetic therapy for non‐small‐cell lung cancer cells
title_sort combined inhibition of ezh2 and histone deacetylases as a potential epigenetic therapy for non‐small‐cell lung cancer cells
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4946723/
https://www.ncbi.nlm.nih.gov/pubmed/27116120
http://dx.doi.org/10.1111/cas.12957
work_keys_str_mv AT takashinataichi combinedinhibitionofezh2andhistonedeacetylasesasapotentialepigenetictherapyfornonsmallcelllungcancercells
AT kinoshitaichiro combinedinhibitionofezh2andhistonedeacetylasesasapotentialepigenetictherapyfornonsmallcelllungcancercells
AT kikuchijunko combinedinhibitionofezh2andhistonedeacetylasesasapotentialepigenetictherapyfornonsmallcelllungcancercells
AT shimizuyasushi combinedinhibitionofezh2andhistonedeacetylasesasapotentialepigenetictherapyfornonsmallcelllungcancercells
AT sakakibarakonishijun combinedinhibitionofezh2andhistonedeacetylasesasapotentialepigenetictherapyfornonsmallcelllungcancercells
AT oizumisatoshi combinedinhibitionofezh2andhistonedeacetylasesasapotentialepigenetictherapyfornonsmallcelllungcancercells
AT nishimuramasaharu combinedinhibitionofezh2andhistonedeacetylasesasapotentialepigenetictherapyfornonsmallcelllungcancercells
AT dosakaakitahirotoshi combinedinhibitionofezh2andhistonedeacetylasesasapotentialepigenetictherapyfornonsmallcelllungcancercells