Cargando…

Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis

BACKGROUND: A promising arsenal of histone deacetylase (HDAC)-targeted treatment has emerged in the past decade, as the abnormal targeting or retention of HDACs to DNA regulatory regions often occurs in many cancers. Head and neck squamous cell carcinoma (HNSCC) is one of the most aggressive maligna...

Descripción completa

Detalles Bibliográficos
Autores principales: He, Leilei, Gao, Lixia, Shay, Chloe, Lang, Liwei, Lv, Fenglin, Teng, Yong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6379952/
https://www.ncbi.nlm.nih.gov/pubmed/30777099
http://dx.doi.org/10.1186/s13046-019-1080-8
_version_ 1783396218211139584
author He, Leilei
Gao, Lixia
Shay, Chloe
Lang, Liwei
Lv, Fenglin
Teng, Yong
author_facet He, Leilei
Gao, Lixia
Shay, Chloe
Lang, Liwei
Lv, Fenglin
Teng, Yong
author_sort He, Leilei
collection PubMed
description BACKGROUND: A promising arsenal of histone deacetylase (HDAC)-targeted treatment has emerged in the past decade, as the abnormal targeting or retention of HDACs to DNA regulatory regions often occurs in many cancers. Head and neck squamous cell carcinoma (HNSCC) is one of the most aggressive malignancies worldwide associated with poor overall survival in late-stage patients. HDAC inhibitors have great potential to treat this devastating disease; however, few has been studied regarding the beneficial role of HDAC inhibition in anti-HNSCC therapy and the underlying molecular mechanisms remain elusive. METHODS: Cell migration and invasion were examined by wound closure and Transwell assays. Protein levels and interactions were assessed by Western blotting and immunoprecipitation. HDAC activity was measured with the fluorometric HDAC Activity Assay. Phospho-receptor tyrosine kinase (RTK) profiling was determined by the Proteome Profiler Human Phospho-RTK Array. RESULTS: ADP-ribosylation factor 1 (Arf1), a small GTPase coordinating vesicle-mediated intracellular trafficking, can be inactivated by HDAC inhibitors through histone acetylation-independent degradation of epidermal growth factor receptor (EGFR) in HNSCC cells. Mechanistically, high levels of Arf1 activity are maintained by binding to phosphorylated EGFR which is localized on HNSCC cell plasma membrane. Decreased EGFR phosphorylation is associated with reduced EGFR protein levels in the presence of TSA, which inactivates Arf1 and eventually inhibits invasion in HNSCC cells. CONCLUSIONS: Our insights explore the critical role of EGFR-Arf1 complex in driving HNSCC progression, and demonstrate the selective action of HDAC inhibitors on this specific axis for suppressing HNSCC invasion. This novel finding represents the first example of modulating the EGFR-Arf1 complex in HNSCC by small molecule agents. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13046-019-1080-8) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6379952
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-63799522019-02-28 Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis He, Leilei Gao, Lixia Shay, Chloe Lang, Liwei Lv, Fenglin Teng, Yong J Exp Clin Cancer Res Research BACKGROUND: A promising arsenal of histone deacetylase (HDAC)-targeted treatment has emerged in the past decade, as the abnormal targeting or retention of HDACs to DNA regulatory regions often occurs in many cancers. Head and neck squamous cell carcinoma (HNSCC) is one of the most aggressive malignancies worldwide associated with poor overall survival in late-stage patients. HDAC inhibitors have great potential to treat this devastating disease; however, few has been studied regarding the beneficial role of HDAC inhibition in anti-HNSCC therapy and the underlying molecular mechanisms remain elusive. METHODS: Cell migration and invasion were examined by wound closure and Transwell assays. Protein levels and interactions were assessed by Western blotting and immunoprecipitation. HDAC activity was measured with the fluorometric HDAC Activity Assay. Phospho-receptor tyrosine kinase (RTK) profiling was determined by the Proteome Profiler Human Phospho-RTK Array. RESULTS: ADP-ribosylation factor 1 (Arf1), a small GTPase coordinating vesicle-mediated intracellular trafficking, can be inactivated by HDAC inhibitors through histone acetylation-independent degradation of epidermal growth factor receptor (EGFR) in HNSCC cells. Mechanistically, high levels of Arf1 activity are maintained by binding to phosphorylated EGFR which is localized on HNSCC cell plasma membrane. Decreased EGFR phosphorylation is associated with reduced EGFR protein levels in the presence of TSA, which inactivates Arf1 and eventually inhibits invasion in HNSCC cells. CONCLUSIONS: Our insights explore the critical role of EGFR-Arf1 complex in driving HNSCC progression, and demonstrate the selective action of HDAC inhibitors on this specific axis for suppressing HNSCC invasion. This novel finding represents the first example of modulating the EGFR-Arf1 complex in HNSCC by small molecule agents. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13046-019-1080-8) contains supplementary material, which is available to authorized users. BioMed Central 2019-02-18 /pmc/articles/PMC6379952/ /pubmed/30777099 http://dx.doi.org/10.1186/s13046-019-1080-8 Text en © The Author(s). 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
He, Leilei
Gao, Lixia
Shay, Chloe
Lang, Liwei
Lv, Fenglin
Teng, Yong
Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis
title Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis
title_full Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis
title_fullStr Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis
title_full_unstemmed Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis
title_short Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis
title_sort histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the egfr-arf1 axis
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6379952/
https://www.ncbi.nlm.nih.gov/pubmed/30777099
http://dx.doi.org/10.1186/s13046-019-1080-8
work_keys_str_mv AT heleilei histonedeacetylaseinhibitorssuppressaggressivenessofheadandnecksquamouscellcarcinomaviahistoneacetylationindependentblockadeoftheegfrarf1axis
AT gaolixia histonedeacetylaseinhibitorssuppressaggressivenessofheadandnecksquamouscellcarcinomaviahistoneacetylationindependentblockadeoftheegfrarf1axis
AT shaychloe histonedeacetylaseinhibitorssuppressaggressivenessofheadandnecksquamouscellcarcinomaviahistoneacetylationindependentblockadeoftheegfrarf1axis
AT langliwei histonedeacetylaseinhibitorssuppressaggressivenessofheadandnecksquamouscellcarcinomaviahistoneacetylationindependentblockadeoftheegfrarf1axis
AT lvfenglin histonedeacetylaseinhibitorssuppressaggressivenessofheadandnecksquamouscellcarcinomaviahistoneacetylationindependentblockadeoftheegfrarf1axis
AT tengyong histonedeacetylaseinhibitorssuppressaggressivenessofheadandnecksquamouscellcarcinomaviahistoneacetylationindependentblockadeoftheegfrarf1axis