Cargando…

Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer

The rising of a highly aggressive subtype of castration-resistant prostate cancer (CRPC) named treatment-induced neuroendocrine prostate cancer (t-NEPC) after androgen deprivation therapy (ADT) is well known for its features of the neuroendocrine differentiation (NED) and androgen receptor (AR) inde...

Descripción completa

Detalles Bibliográficos
Autores principales: Long, Zhi, Deng, Liang, Li, Chao, He, Qiangrong, He, Yao, Hu, Xiheng, Cai, Yi, Gan, Yu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7790822/
https://www.ncbi.nlm.nih.gov/pubmed/33414441
http://dx.doi.org/10.1038/s41419-020-03326-8
_version_ 1783633503138611200
author Long, Zhi
Deng, Liang
Li, Chao
He, Qiangrong
He, Yao
Hu, Xiheng
Cai, Yi
Gan, Yu
author_facet Long, Zhi
Deng, Liang
Li, Chao
He, Qiangrong
He, Yao
Hu, Xiheng
Cai, Yi
Gan, Yu
author_sort Long, Zhi
collection PubMed
description The rising of a highly aggressive subtype of castration-resistant prostate cancer (CRPC) named treatment-induced neuroendocrine prostate cancer (t-NEPC) after androgen deprivation therapy (ADT) is well known for its features of the neuroendocrine differentiation (NED) and androgen receptor (AR) independence. However, t-NEPC is still largely unknown. Here, we found that EHF is notably depressed in t-NEPC tumors, patient-derived xenografts, transgenic mice, and cell models. Results from cell lines uncovered that ADT represses EHF expression, which is required for the ADT-induced NED. Mechanism dissection revealed that ADT decreases the EHF transcription via relieving the AR binding to different androgen-responsive elements, which then promotes the expression and enzymatic activity of enhancer of zeste homolog 2 (EZH2), consequently catalyzing tri-methylation lysine 27 of histone H3 for transcriptional repression of its downstream genes to promote the NED. Furthermore, preclinical studies from cell and mice models proved that recovery of EHF expression or using EZH2 inhibitor can attenuate aggressive properties of CRPC cells, hinder the progression of t-NEPC, and promote the response of CPRC cells to enzalutamide. Together, we elucidate that the ADT/AR/EHF/EZH2 signaling is required for the ADT-enhanced NED and plays a critical role in the progression of t-NEPC.
format Online
Article
Text
id pubmed-7790822
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-77908222021-01-14 Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer Long, Zhi Deng, Liang Li, Chao He, Qiangrong He, Yao Hu, Xiheng Cai, Yi Gan, Yu Cell Death Dis Article The rising of a highly aggressive subtype of castration-resistant prostate cancer (CRPC) named treatment-induced neuroendocrine prostate cancer (t-NEPC) after androgen deprivation therapy (ADT) is well known for its features of the neuroendocrine differentiation (NED) and androgen receptor (AR) independence. However, t-NEPC is still largely unknown. Here, we found that EHF is notably depressed in t-NEPC tumors, patient-derived xenografts, transgenic mice, and cell models. Results from cell lines uncovered that ADT represses EHF expression, which is required for the ADT-induced NED. Mechanism dissection revealed that ADT decreases the EHF transcription via relieving the AR binding to different androgen-responsive elements, which then promotes the expression and enzymatic activity of enhancer of zeste homolog 2 (EZH2), consequently catalyzing tri-methylation lysine 27 of histone H3 for transcriptional repression of its downstream genes to promote the NED. Furthermore, preclinical studies from cell and mice models proved that recovery of EHF expression or using EZH2 inhibitor can attenuate aggressive properties of CRPC cells, hinder the progression of t-NEPC, and promote the response of CPRC cells to enzalutamide. Together, we elucidate that the ADT/AR/EHF/EZH2 signaling is required for the ADT-enhanced NED and plays a critical role in the progression of t-NEPC. Nature Publishing Group UK 2021-01-05 /pmc/articles/PMC7790822/ /pubmed/33414441 http://dx.doi.org/10.1038/s41419-020-03326-8 Text en © The Author(s) 2021 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
Long, Zhi
Deng, Liang
Li, Chao
He, Qiangrong
He, Yao
Hu, Xiheng
Cai, Yi
Gan, Yu
Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer
title Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer
title_full Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer
title_fullStr Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer
title_full_unstemmed Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer
title_short Loss of EHF facilitates the development of treatment-induced neuroendocrine prostate cancer
title_sort loss of ehf facilitates the development of treatment-induced neuroendocrine prostate cancer
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7790822/
https://www.ncbi.nlm.nih.gov/pubmed/33414441
http://dx.doi.org/10.1038/s41419-020-03326-8
work_keys_str_mv AT longzhi lossofehffacilitatesthedevelopmentoftreatmentinducedneuroendocrineprostatecancer
AT dengliang lossofehffacilitatesthedevelopmentoftreatmentinducedneuroendocrineprostatecancer
AT lichao lossofehffacilitatesthedevelopmentoftreatmentinducedneuroendocrineprostatecancer
AT heqiangrong lossofehffacilitatesthedevelopmentoftreatmentinducedneuroendocrineprostatecancer
AT heyao lossofehffacilitatesthedevelopmentoftreatmentinducedneuroendocrineprostatecancer
AT huxiheng lossofehffacilitatesthedevelopmentoftreatmentinducedneuroendocrineprostatecancer
AT caiyi lossofehffacilitatesthedevelopmentoftreatmentinducedneuroendocrineprostatecancer
AT ganyu lossofehffacilitatesthedevelopmentoftreatmentinducedneuroendocrineprostatecancer