Distinct Ring1b complexes defined by DEAD-box helicases and EMT transcription factors synergistically enhance E-cadherin silencing in breast cancer

Abstract Ring1b is a core subunit of polycomb repressive complex 1 (PRC1) and is essential in several high-risk cancers. However, the epigenetic mechanism of Ring1b underlying breast cancer malignancy is poorly understood. In this study, we showed increased expression of Ring1b promoted metastasis b...

Full description

Bibliographic Details
Main Authors: Yawei Wang, Yingying Sun, Chao Shang, Lili Chen, Hongyu Chen, Dake Wang, Xianlu Zeng
Format: Article
Language:English
Published: Nature Publishing Group 2021-02-01
Series:Cell Death and Disease
Online Access:https://doi.org/10.1038/s41419-021-03491-4
id doaj-244d40385c1240bd8db99d18734644ce
record_format Article
spelling doaj-244d40385c1240bd8db99d18734644ce2021-02-21T12:05:01ZengNature Publishing GroupCell Death and Disease2041-48892021-02-0112211410.1038/s41419-021-03491-4Distinct Ring1b complexes defined by DEAD-box helicases and EMT transcription factors synergistically enhance E-cadherin silencing in breast cancerYawei Wang0Yingying Sun1Chao Shang2Lili Chen3Hongyu Chen4Dake Wang5Xianlu Zeng6The Key Laboratory of Molecular Epigenetics of the Ministry of Education, Institute of Genetics and Cytology, Northeast Normal UniversityThe Key Laboratory of Molecular Epigenetics of the Ministry of Education, Institute of Genetics and Cytology, Northeast Normal UniversityThe Key Laboratory of Molecular Epigenetics of the Ministry of Education, Institute of Genetics and Cytology, Northeast Normal UniversityThe Key Laboratory of Molecular Epigenetics of the Ministry of Education, Institute of Genetics and Cytology, Northeast Normal UniversityThe Key Laboratory of Molecular Epigenetics of the Ministry of Education, Institute of Genetics and Cytology, Northeast Normal UniversityThe Key Laboratory of Molecular Epigenetics of the Ministry of Education, Institute of Genetics and Cytology, Northeast Normal UniversityThe Key Laboratory of Molecular Epigenetics of the Ministry of Education, Institute of Genetics and Cytology, Northeast Normal UniversityAbstract Ring1b is a core subunit of polycomb repressive complex 1 (PRC1) and is essential in several high-risk cancers. However, the epigenetic mechanism of Ring1b underlying breast cancer malignancy is poorly understood. In this study, we showed increased expression of Ring1b promoted metastasis by weakening cell–cell adhesions of breast cancer cells. We confirmed that Ring1b could downregulate E-cadherin and contributed to an epigenetic rewiring via PRC1-dependent function by forming distinct complexes with DEAD-box RNA helicases (DDXs) or epithelial-mesenchymal transition transcription factors (EMT TFs) on site-specific loci of E-cadherin promoter. DDXs-Ring1b complexes moderately inhibited E-cadherin, which resulted in an early hybrid EMT state of epithelial cells, and EMT TFs-Ring1b complexes cooperated with DDXs-Ring1b complexes to further repress E-cadherin in mesenchymal-like cancer cells. Clinically, high expression of Ring1b with DDXs or EMT TFs predicted low levels of E-cadherin, metastatic behavior, and poor prognosis. These findings provide an epigenetic regulation mechanism of Ring1b complexes in E-cadherin expression. Ring1b complexes may be potential therapeutic targets and biomarkers for diagnosis and prognosis in invasion breast cancer.https://doi.org/10.1038/s41419-021-03491-4
collection DOAJ
language English
format Article
sources DOAJ
author Yawei Wang
Yingying Sun
Chao Shang
Lili Chen
Hongyu Chen
Dake Wang
Xianlu Zeng
spellingShingle Yawei Wang
Yingying Sun
Chao Shang
Lili Chen
Hongyu Chen
Dake Wang
Xianlu Zeng
Distinct Ring1b complexes defined by DEAD-box helicases and EMT transcription factors synergistically enhance E-cadherin silencing in breast cancer
Cell Death and Disease
author_facet Yawei Wang
Yingying Sun
Chao Shang
Lili Chen
Hongyu Chen
Dake Wang
Xianlu Zeng
author_sort Yawei Wang
title Distinct Ring1b complexes defined by DEAD-box helicases and EMT transcription factors synergistically enhance E-cadherin silencing in breast cancer
title_short Distinct Ring1b complexes defined by DEAD-box helicases and EMT transcription factors synergistically enhance E-cadherin silencing in breast cancer
title_full Distinct Ring1b complexes defined by DEAD-box helicases and EMT transcription factors synergistically enhance E-cadherin silencing in breast cancer
title_fullStr Distinct Ring1b complexes defined by DEAD-box helicases and EMT transcription factors synergistically enhance E-cadherin silencing in breast cancer
title_full_unstemmed Distinct Ring1b complexes defined by DEAD-box helicases and EMT transcription factors synergistically enhance E-cadherin silencing in breast cancer
title_sort distinct ring1b complexes defined by dead-box helicases and emt transcription factors synergistically enhance e-cadherin silencing in breast cancer
publisher Nature Publishing Group
series Cell Death and Disease
issn 2041-4889
publishDate 2021-02-01
description Abstract Ring1b is a core subunit of polycomb repressive complex 1 (PRC1) and is essential in several high-risk cancers. However, the epigenetic mechanism of Ring1b underlying breast cancer malignancy is poorly understood. In this study, we showed increased expression of Ring1b promoted metastasis by weakening cell–cell adhesions of breast cancer cells. We confirmed that Ring1b could downregulate E-cadherin and contributed to an epigenetic rewiring via PRC1-dependent function by forming distinct complexes with DEAD-box RNA helicases (DDXs) or epithelial-mesenchymal transition transcription factors (EMT TFs) on site-specific loci of E-cadherin promoter. DDXs-Ring1b complexes moderately inhibited E-cadherin, which resulted in an early hybrid EMT state of epithelial cells, and EMT TFs-Ring1b complexes cooperated with DDXs-Ring1b complexes to further repress E-cadherin in mesenchymal-like cancer cells. Clinically, high expression of Ring1b with DDXs or EMT TFs predicted low levels of E-cadherin, metastatic behavior, and poor prognosis. These findings provide an epigenetic regulation mechanism of Ring1b complexes in E-cadherin expression. Ring1b complexes may be potential therapeutic targets and biomarkers for diagnosis and prognosis in invasion breast cancer.
url https://doi.org/10.1038/s41419-021-03491-4
work_keys_str_mv AT yaweiwang distinctring1bcomplexesdefinedbydeadboxhelicasesandemttranscriptionfactorssynergisticallyenhanceecadherinsilencinginbreastcancer
AT yingyingsun distinctring1bcomplexesdefinedbydeadboxhelicasesandemttranscriptionfactorssynergisticallyenhanceecadherinsilencinginbreastcancer
AT chaoshang distinctring1bcomplexesdefinedbydeadboxhelicasesandemttranscriptionfactorssynergisticallyenhanceecadherinsilencinginbreastcancer
AT lilichen distinctring1bcomplexesdefinedbydeadboxhelicasesandemttranscriptionfactorssynergisticallyenhanceecadherinsilencinginbreastcancer
AT hongyuchen distinctring1bcomplexesdefinedbydeadboxhelicasesandemttranscriptionfactorssynergisticallyenhanceecadherinsilencinginbreastcancer
AT dakewang distinctring1bcomplexesdefinedbydeadboxhelicasesandemttranscriptionfactorssynergisticallyenhanceecadherinsilencinginbreastcancer
AT xianluzeng distinctring1bcomplexesdefinedbydeadboxhelicasesandemttranscriptionfactorssynergisticallyenhanceecadherinsilencinginbreastcancer
_version_ 1724258473877700608