Cargando…

Gain-of-function mutant p53 promotes the oncogenic potential of head and neck squamous cell carcinoma cells by targeting the transcription factors FOXO3a and FOXM1

Many mutant p53 proteins exert oncogenic gain-of-function (GOF) properties that promote cancer cell invasive growth and metastasis, yet the mechanisms mediating these functions still largely remain elusive. We show here that overexpression of the GOF mutant p53 G245D and other GOF p53 mutants enhanc...

Descripción completa

Detalles Bibliográficos
Autores principales: Tanaka, Noriaki, Zhao, Mei, Tang, Lin, Patel, Ameeta A., Xi, Qing, Van, Hieu T., Takahashi, Hideaki, Osman, Abdullah A., Zhang, Jiexin, Wang, Jing, Myers, Jeffrey N., Zhou, Ge
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5844800/
https://www.ncbi.nlm.nih.gov/pubmed/29269868
http://dx.doi.org/10.1038/s41388-017-0032-z
_version_ 1783305303226318848
author Tanaka, Noriaki
Zhao, Mei
Tang, Lin
Patel, Ameeta A.
Xi, Qing
Van, Hieu T.
Takahashi, Hideaki
Osman, Abdullah A.
Zhang, Jiexin
Wang, Jing
Myers, Jeffrey N.
Zhou, Ge
author_facet Tanaka, Noriaki
Zhao, Mei
Tang, Lin
Patel, Ameeta A.
Xi, Qing
Van, Hieu T.
Takahashi, Hideaki
Osman, Abdullah A.
Zhang, Jiexin
Wang, Jing
Myers, Jeffrey N.
Zhou, Ge
author_sort Tanaka, Noriaki
collection PubMed
description Many mutant p53 proteins exert oncogenic gain-of-function (GOF) properties that promote cancer cell invasive growth and metastasis, yet the mechanisms mediating these functions still largely remain elusive. We show here that overexpression of the GOF mutant p53 G245D and other GOF p53 mutants enhances the invasive cell growth of p53-deficient head and neck squamous cell carcinoma (HNSCC) UM-SCC-1 cells both in in vitro three-dimensional culture and in an in vivo orthotopic nude mouse model of HNSCC through a novel transcription-independent mechanism. We demonstrate that the expression of the oncogenic forkhead transcription factor FOXM1 is upregulated by GOF mutant p53s. Moreover, we show that overexpression of GOF mutant p53 G245D decreases the AMP-activated protein kinase (AMPK)-mediated phosphorylation of FOXO3a, a tumor suppressive forkhead transcription factor, leading to its cytoplasmic accumulation. This downregulation of FOXO3a’s activity, in turn, leads to de-repression of FOXM1 expression. Importantly, we show that either overexpression of FOXO3a or downregulation of FOXM1 impairs both GOF mutant p53-mediated cell invasion in vitro and pulmonary metastases of UM-SCC-1 cells in vivo. Finally, not only do oral cancer patients with p53 mutations exhibit higher levels of FOXM1 expression than patients with wild-type p53, but also HNSCC patients with TP53 mutations and high levels of FOXM1 expression have the poorest survival outcomes. Given our prior demonstration that GOF mutant p53s inhibit AMPK, our current study, establishes and demonstrates a novel transcription-independent GOF mutant p53-AMPK-FOXO3a-FOXM1 signaling cascade that plays an important role in mediating mutant p53s’ gain-of-function activities in HNSCCs.
format Online
Article
Text
id pubmed-5844800
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-58448002018-06-22 Gain-of-function mutant p53 promotes the oncogenic potential of head and neck squamous cell carcinoma cells by targeting the transcription factors FOXO3a and FOXM1 Tanaka, Noriaki Zhao, Mei Tang, Lin Patel, Ameeta A. Xi, Qing Van, Hieu T. Takahashi, Hideaki Osman, Abdullah A. Zhang, Jiexin Wang, Jing Myers, Jeffrey N. Zhou, Ge Oncogene Article Many mutant p53 proteins exert oncogenic gain-of-function (GOF) properties that promote cancer cell invasive growth and metastasis, yet the mechanisms mediating these functions still largely remain elusive. We show here that overexpression of the GOF mutant p53 G245D and other GOF p53 mutants enhances the invasive cell growth of p53-deficient head and neck squamous cell carcinoma (HNSCC) UM-SCC-1 cells both in in vitro three-dimensional culture and in an in vivo orthotopic nude mouse model of HNSCC through a novel transcription-independent mechanism. We demonstrate that the expression of the oncogenic forkhead transcription factor FOXM1 is upregulated by GOF mutant p53s. Moreover, we show that overexpression of GOF mutant p53 G245D decreases the AMP-activated protein kinase (AMPK)-mediated phosphorylation of FOXO3a, a tumor suppressive forkhead transcription factor, leading to its cytoplasmic accumulation. This downregulation of FOXO3a’s activity, in turn, leads to de-repression of FOXM1 expression. Importantly, we show that either overexpression of FOXO3a or downregulation of FOXM1 impairs both GOF mutant p53-mediated cell invasion in vitro and pulmonary metastases of UM-SCC-1 cells in vivo. Finally, not only do oral cancer patients with p53 mutations exhibit higher levels of FOXM1 expression than patients with wild-type p53, but also HNSCC patients with TP53 mutations and high levels of FOXM1 expression have the poorest survival outcomes. Given our prior demonstration that GOF mutant p53s inhibit AMPK, our current study, establishes and demonstrates a novel transcription-independent GOF mutant p53-AMPK-FOXO3a-FOXM1 signaling cascade that plays an important role in mediating mutant p53s’ gain-of-function activities in HNSCCs. Nature Publishing Group UK 2017-12-22 2018 /pmc/articles/PMC5844800/ /pubmed/29269868 http://dx.doi.org/10.1038/s41388-017-0032-z Text en © The Author(s) 2017 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as 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 images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Tanaka, Noriaki
Zhao, Mei
Tang, Lin
Patel, Ameeta A.
Xi, Qing
Van, Hieu T.
Takahashi, Hideaki
Osman, Abdullah A.
Zhang, Jiexin
Wang, Jing
Myers, Jeffrey N.
Zhou, Ge
Gain-of-function mutant p53 promotes the oncogenic potential of head and neck squamous cell carcinoma cells by targeting the transcription factors FOXO3a and FOXM1
title Gain-of-function mutant p53 promotes the oncogenic potential of head and neck squamous cell carcinoma cells by targeting the transcription factors FOXO3a and FOXM1
title_full Gain-of-function mutant p53 promotes the oncogenic potential of head and neck squamous cell carcinoma cells by targeting the transcription factors FOXO3a and FOXM1
title_fullStr Gain-of-function mutant p53 promotes the oncogenic potential of head and neck squamous cell carcinoma cells by targeting the transcription factors FOXO3a and FOXM1
title_full_unstemmed Gain-of-function mutant p53 promotes the oncogenic potential of head and neck squamous cell carcinoma cells by targeting the transcription factors FOXO3a and FOXM1
title_short Gain-of-function mutant p53 promotes the oncogenic potential of head and neck squamous cell carcinoma cells by targeting the transcription factors FOXO3a and FOXM1
title_sort gain-of-function mutant p53 promotes the oncogenic potential of head and neck squamous cell carcinoma cells by targeting the transcription factors foxo3a and foxm1
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5844800/
https://www.ncbi.nlm.nih.gov/pubmed/29269868
http://dx.doi.org/10.1038/s41388-017-0032-z
work_keys_str_mv AT tanakanoriaki gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT zhaomei gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT tanglin gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT patelameetaa gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT xiqing gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT vanhieut gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT takahashihideaki gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT osmanabdullaha gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT zhangjiexin gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT wangjing gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT myersjeffreyn gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1
AT zhouge gainoffunctionmutantp53promotestheoncogenicpotentialofheadandnecksquamouscellcarcinomacellsbytargetingthetranscriptionfactorsfoxo3aandfoxm1