ARSD, a novel ERα downstream target gene, inhibits proliferation and migration of breast cancer cells via activating Hippo/YAP pathway

Abstract Advanced breast cancer (BC), especially basal like triple-negative BC (TNBC), is a highly malignant tumor without viable treatment option, highlighting the urgent need to seek novel therapeutic targets. Arylsulfatase D (ARSD), localized at Xp22.3, is a female-biased gene due to its escaping...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Yun Lin, Chun Li, Wei Xiong, Liping Fan, Hongchao Pan, Yaochen Li
Formato: article
Lenguaje:EN
Publicado: Nature Publishing Group 2021
Materias:
Acceso en línea:https://doaj.org/article/2e1acce53f8949e5868615914889967c
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:2e1acce53f8949e5868615914889967c
record_format dspace
spelling oai:doaj.org-article:2e1acce53f8949e5868615914889967c2021-11-07T12:04:53ZARSD, a novel ERα downstream target gene, inhibits proliferation and migration of breast cancer cells via activating Hippo/YAP pathway10.1038/s41419-021-04338-82041-4889https://doaj.org/article/2e1acce53f8949e5868615914889967c2021-11-01T00:00:00Zhttps://doi.org/10.1038/s41419-021-04338-8https://doaj.org/toc/2041-4889Abstract Advanced breast cancer (BC), especially basal like triple-negative BC (TNBC), is a highly malignant tumor without viable treatment option, highlighting the urgent need to seek novel therapeutic targets. Arylsulfatase D (ARSD), localized at Xp22.3, is a female-biased gene due to its escaping from X chromosome inactivation (XCI). Unfortunately, no systematic investigation of ARSD on BC has been reported. In this study, we observed that ARSD expression was positively related to ERα status either in BC cells or tissue specimens, which were associated with good prognosis. Furthermore, we found a set of hormone-responsive lineage-specific transcription factors, FOXA1, GATA3, ERα, directly drove high expression of ARSD through chromatin looping in luminal subtype BC cells. Opposingly, ARSD still subjected to XCI in TNBC cells mediated by Xist, CpG islands methylation, and inhibitory histone modification. Unexpectedly, we also found that ectopic ARSD overexpression could inhibit proliferation and migration of TNBC cells by activating Hippo/YAP pathway, indicating that ARSD may be a molecule brake on ERα signaling pathway, which restricted ERα to be an uncontrolled active status. Combined with other peoples’ researches that Hippo signaling maintained ER expression and ER + BC growth, we believed that there should exist a regulative feedback loop formation among ERα, ARSD, and Hippo/YAP pathway. Collectively, our findings will help filling the knowledge gap about the influence of ARSD on BC and providing evidence that ARSD may serve as a potential marker to predict prognosis and as a therapeutic target.Yun LinChun LiWei XiongLiping FanHongchao PanYaochen LiNature Publishing GrouparticleCytologyQH573-671ENCell Death and Disease, Vol 12, Iss 11, Pp 1-14 (2021)
institution DOAJ
collection DOAJ
language EN
topic Cytology
QH573-671
spellingShingle Cytology
QH573-671
Yun Lin
Chun Li
Wei Xiong
Liping Fan
Hongchao Pan
Yaochen Li
ARSD, a novel ERα downstream target gene, inhibits proliferation and migration of breast cancer cells via activating Hippo/YAP pathway
description Abstract Advanced breast cancer (BC), especially basal like triple-negative BC (TNBC), is a highly malignant tumor without viable treatment option, highlighting the urgent need to seek novel therapeutic targets. Arylsulfatase D (ARSD), localized at Xp22.3, is a female-biased gene due to its escaping from X chromosome inactivation (XCI). Unfortunately, no systematic investigation of ARSD on BC has been reported. In this study, we observed that ARSD expression was positively related to ERα status either in BC cells or tissue specimens, which were associated with good prognosis. Furthermore, we found a set of hormone-responsive lineage-specific transcription factors, FOXA1, GATA3, ERα, directly drove high expression of ARSD through chromatin looping in luminal subtype BC cells. Opposingly, ARSD still subjected to XCI in TNBC cells mediated by Xist, CpG islands methylation, and inhibitory histone modification. Unexpectedly, we also found that ectopic ARSD overexpression could inhibit proliferation and migration of TNBC cells by activating Hippo/YAP pathway, indicating that ARSD may be a molecule brake on ERα signaling pathway, which restricted ERα to be an uncontrolled active status. Combined with other peoples’ researches that Hippo signaling maintained ER expression and ER + BC growth, we believed that there should exist a regulative feedback loop formation among ERα, ARSD, and Hippo/YAP pathway. Collectively, our findings will help filling the knowledge gap about the influence of ARSD on BC and providing evidence that ARSD may serve as a potential marker to predict prognosis and as a therapeutic target.
format article
author Yun Lin
Chun Li
Wei Xiong
Liping Fan
Hongchao Pan
Yaochen Li
author_facet Yun Lin
Chun Li
Wei Xiong
Liping Fan
Hongchao Pan
Yaochen Li
author_sort Yun Lin
title ARSD, a novel ERα downstream target gene, inhibits proliferation and migration of breast cancer cells via activating Hippo/YAP pathway
title_short ARSD, a novel ERα downstream target gene, inhibits proliferation and migration of breast cancer cells via activating Hippo/YAP pathway
title_full ARSD, a novel ERα downstream target gene, inhibits proliferation and migration of breast cancer cells via activating Hippo/YAP pathway
title_fullStr ARSD, a novel ERα downstream target gene, inhibits proliferation and migration of breast cancer cells via activating Hippo/YAP pathway
title_full_unstemmed ARSD, a novel ERα downstream target gene, inhibits proliferation and migration of breast cancer cells via activating Hippo/YAP pathway
title_sort arsd, a novel erα downstream target gene, inhibits proliferation and migration of breast cancer cells via activating hippo/yap pathway
publisher Nature Publishing Group
publishDate 2021
url https://doaj.org/article/2e1acce53f8949e5868615914889967c
work_keys_str_mv AT yunlin arsdanoveleradownstreamtargetgeneinhibitsproliferationandmigrationofbreastcancercellsviaactivatinghippoyappathway
AT chunli arsdanoveleradownstreamtargetgeneinhibitsproliferationandmigrationofbreastcancercellsviaactivatinghippoyappathway
AT weixiong arsdanoveleradownstreamtargetgeneinhibitsproliferationandmigrationofbreastcancercellsviaactivatinghippoyappathway
AT lipingfan arsdanoveleradownstreamtargetgeneinhibitsproliferationandmigrationofbreastcancercellsviaactivatinghippoyappathway
AT hongchaopan arsdanoveleradownstreamtargetgeneinhibitsproliferationandmigrationofbreastcancercellsviaactivatinghippoyappathway
AT yaochenli arsdanoveleradownstreamtargetgeneinhibitsproliferationandmigrationofbreastcancercellsviaactivatinghippoyappathway
_version_ 1718443537280794624