Cargando…

Ubiquitin-Specific Peptidase 8 Modulates Cell Proliferation and Induces Cell Cycle Arrest and Apoptosis in Breast Cancer by Stabilizing Estrogen Receptor Alpha

Breast cancer (BC) is the most common neoplastic and lethal malignancy in women. Although antiendocrine therapy is the main treatment for estrogen receptor alpha (ERα)-positive BC, the development of resistance is a major clinical complication. In this study, we aimed to explore the role of ubiquiti...

Descripción completa

Detalles Bibliográficos
Autores principales: Zheng, Lewei, Yang, Qian, Li, Chengxin, Xu, Gaoran, Yuan, Qianqian, Hou, Jinxuan, Wu, Gaosong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9839415/
https://www.ncbi.nlm.nih.gov/pubmed/36644233
http://dx.doi.org/10.1155/2023/8483325
_version_ 1784869479669301248
author Zheng, Lewei
Yang, Qian
Li, Chengxin
Xu, Gaoran
Yuan, Qianqian
Hou, Jinxuan
Wu, Gaosong
author_facet Zheng, Lewei
Yang, Qian
Li, Chengxin
Xu, Gaoran
Yuan, Qianqian
Hou, Jinxuan
Wu, Gaosong
author_sort Zheng, Lewei
collection PubMed
description Breast cancer (BC) is the most common neoplastic and lethal malignancy in women. Although antiendocrine therapy is the main treatment for estrogen receptor alpha (ERα)-positive BC, the development of resistance is a major clinical complication. In this study, we aimed to explore the role of ubiquitin-specific peptidase 8 (USP8) in ERα signaling and identify potential targets for endocrine resistance. Public databases were used to analyze USP8 expression, prognosis, clinical characteristics, and immune cell infiltration. Immunohistochemistry and western blot assays were used to detect protein levels and ERα signaling. Quantitative reverse transcription-PCR was used to measure ERα target gene expression. The cell counting kit-8, wound-healing, clone formation, and Transwell assays were used to investigate the effects of USP8 depletion or inhibition on cell proliferation, migration, and invasion. An immunofluorescence assay was used for localizing USP8 and ERα, and a protein stability assay was performed for detecting the degradation of ERα protein. The cell cycle and apoptosis were assessed using flow cytometry. USP8 was highly expressed in the luminal subtype of BC and was associated with poor prognosis. The infiltration levels of many immune cells were positively correlated with USP8 expression. Depletion of USP8 dramatically decreased the ERα signaling activity and weakened the proliferation, migration, and invasion capabilities of BC cells. USP8 knockdown markedly induced apoptosis and cell cycle arrest (G0/G1). Colocalization analysis and protein stability assays indicated a probable mechanism by which USP8 regulates ERα. Our study demonstrates that USP8 might be crucial in BC development and may be considered a potential target for treating ER-positive BC malignancies in vitro.
format Online
Article
Text
id pubmed-9839415
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Hindawi
record_format MEDLINE/PubMed
spelling pubmed-98394152023-01-14 Ubiquitin-Specific Peptidase 8 Modulates Cell Proliferation and Induces Cell Cycle Arrest and Apoptosis in Breast Cancer by Stabilizing Estrogen Receptor Alpha Zheng, Lewei Yang, Qian Li, Chengxin Xu, Gaoran Yuan, Qianqian Hou, Jinxuan Wu, Gaosong J Oncol Research Article Breast cancer (BC) is the most common neoplastic and lethal malignancy in women. Although antiendocrine therapy is the main treatment for estrogen receptor alpha (ERα)-positive BC, the development of resistance is a major clinical complication. In this study, we aimed to explore the role of ubiquitin-specific peptidase 8 (USP8) in ERα signaling and identify potential targets for endocrine resistance. Public databases were used to analyze USP8 expression, prognosis, clinical characteristics, and immune cell infiltration. Immunohistochemistry and western blot assays were used to detect protein levels and ERα signaling. Quantitative reverse transcription-PCR was used to measure ERα target gene expression. The cell counting kit-8, wound-healing, clone formation, and Transwell assays were used to investigate the effects of USP8 depletion or inhibition on cell proliferation, migration, and invasion. An immunofluorescence assay was used for localizing USP8 and ERα, and a protein stability assay was performed for detecting the degradation of ERα protein. The cell cycle and apoptosis were assessed using flow cytometry. USP8 was highly expressed in the luminal subtype of BC and was associated with poor prognosis. The infiltration levels of many immune cells were positively correlated with USP8 expression. Depletion of USP8 dramatically decreased the ERα signaling activity and weakened the proliferation, migration, and invasion capabilities of BC cells. USP8 knockdown markedly induced apoptosis and cell cycle arrest (G0/G1). Colocalization analysis and protein stability assays indicated a probable mechanism by which USP8 regulates ERα. Our study demonstrates that USP8 might be crucial in BC development and may be considered a potential target for treating ER-positive BC malignancies in vitro. Hindawi 2023-01-04 /pmc/articles/PMC9839415/ /pubmed/36644233 http://dx.doi.org/10.1155/2023/8483325 Text en Copyright © 2023 Lewei Zheng et al. https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Zheng, Lewei
Yang, Qian
Li, Chengxin
Xu, Gaoran
Yuan, Qianqian
Hou, Jinxuan
Wu, Gaosong
Ubiquitin-Specific Peptidase 8 Modulates Cell Proliferation and Induces Cell Cycle Arrest and Apoptosis in Breast Cancer by Stabilizing Estrogen Receptor Alpha
title Ubiquitin-Specific Peptidase 8 Modulates Cell Proliferation and Induces Cell Cycle Arrest and Apoptosis in Breast Cancer by Stabilizing Estrogen Receptor Alpha
title_full Ubiquitin-Specific Peptidase 8 Modulates Cell Proliferation and Induces Cell Cycle Arrest and Apoptosis in Breast Cancer by Stabilizing Estrogen Receptor Alpha
title_fullStr Ubiquitin-Specific Peptidase 8 Modulates Cell Proliferation and Induces Cell Cycle Arrest and Apoptosis in Breast Cancer by Stabilizing Estrogen Receptor Alpha
title_full_unstemmed Ubiquitin-Specific Peptidase 8 Modulates Cell Proliferation and Induces Cell Cycle Arrest and Apoptosis in Breast Cancer by Stabilizing Estrogen Receptor Alpha
title_short Ubiquitin-Specific Peptidase 8 Modulates Cell Proliferation and Induces Cell Cycle Arrest and Apoptosis in Breast Cancer by Stabilizing Estrogen Receptor Alpha
title_sort ubiquitin-specific peptidase 8 modulates cell proliferation and induces cell cycle arrest and apoptosis in breast cancer by stabilizing estrogen receptor alpha
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9839415/
https://www.ncbi.nlm.nih.gov/pubmed/36644233
http://dx.doi.org/10.1155/2023/8483325
work_keys_str_mv AT zhenglewei ubiquitinspecificpeptidase8modulatescellproliferationandinducescellcyclearrestandapoptosisinbreastcancerbystabilizingestrogenreceptoralpha
AT yangqian ubiquitinspecificpeptidase8modulatescellproliferationandinducescellcyclearrestandapoptosisinbreastcancerbystabilizingestrogenreceptoralpha
AT lichengxin ubiquitinspecificpeptidase8modulatescellproliferationandinducescellcyclearrestandapoptosisinbreastcancerbystabilizingestrogenreceptoralpha
AT xugaoran ubiquitinspecificpeptidase8modulatescellproliferationandinducescellcyclearrestandapoptosisinbreastcancerbystabilizingestrogenreceptoralpha
AT yuanqianqian ubiquitinspecificpeptidase8modulatescellproliferationandinducescellcyclearrestandapoptosisinbreastcancerbystabilizingestrogenreceptoralpha
AT houjinxuan ubiquitinspecificpeptidase8modulatescellproliferationandinducescellcyclearrestandapoptosisinbreastcancerbystabilizingestrogenreceptoralpha
AT wugaosong ubiquitinspecificpeptidase8modulatescellproliferationandinducescellcyclearrestandapoptosisinbreastcancerbystabilizingestrogenreceptoralpha