ZMIZ1 enhances ERα-dependent expression of E2F2 in breast cancer

Author:

Zhao Weiye1,Rose Susanna F1,Blake Ryan2,Godicelj Aňze3,Cullen Amy E2,Stenning Jack1,Beevors Lucy4,Gehrung Marcel2,Kumar Sanjeev5,Kishore Kamal2,Sawle Ashley2,Eldridge Matthew2,Giorgi Federico M6,Bridge Katherine S17,Markowetz Florian2,Holding Andrew N178ORCID

Affiliation:

1. Department of Biology, University of York, York, UK

2. CRUK Cambridge Institute, University of Cambridge, Cambridge, UK

3. Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Smith Building, Boston, Massachusetts, USA

4. The Institute of Metabolism and Systems Research (IMSR), University of Birmingham, College of Medical and Dental Sciences, Birmingham, UK

5. Chris O’Brien Lifehouse, Sydney, New South Wales, Australia

6. Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy

7. York Biomedical Research Institute, University of York, York, UK

8. The Alan Turing Institute, Kings Cross, London, UK

Abstract

The estrogen receptor-α (ER) drives 75% of breast cancers. On activation, the ER recruits and assembles a 1–2 MDa transcriptionally active complex. These complexes can modulate tumour growth, and understanding the roles of individual proteins within these complexes can help identify new therapeutic targets. Here, we present the discovery of ER and ZMIZ1 within the same multi-protein assembly by quantitative proteomics, and validated by proximity ligation assay. We characterise ZMIZ1 function by demonstrating a significant decrease in the proliferation of ER-positive cancer cell lines. To establish a role for the ER-ZMIZ1 interaction, we measured the transcriptional changes in the estrogen response post-ZMIZ1 knockdown using an RNA-seq time-course over 24 h. Gene set enrichment analysis of the ZMIZ1-knockdown data identified a specific delay in the response of estradiol-induced cell cycle genes. Integration of ENCODE data with our RNA-seq results identified that ER and ZMIZ1 both bind the promoter of E2F2. We therefore propose that ER and ZMIZ1 interact to enable the efficient estrogenic response at subset of cell cycle genes via a novel ZMIZ1–ER–E2F2 signalling axis. Finally, we show that high ZMIZ1 expression is predictive of worse patient outcome, ER and ZMIZ1 are co-expressed in breast cancer patients in TCGA and METABRIC, and the proteins are co-localised within the nuclei of tumour cell in patient biopsies. In conclusion, we establish that ZMIZ1 is a regulator of the estrogenic cell cycle response and provide evidence of the biological importance of the ER–ZMIZ1 interaction in ER-positive patient tumours, supporting potential clinical relevance.

Publisher

Bioscientifica

Reference48 articles.

1. Proximity ligation assay (PLA);Alam,2022

2. Functional characterization of somatic mutations in cancer using network-based inference of protein activity;Alvarez,2016

3. Proteomic analysis of archival breast cancer clinical specimens identifies biological subtypes with distinct survival outcomes;Asleh,2022

4. Prioritization of cancer therapeutic targets using CRISPR-Cas9 screens;Behan,2019

5. AKT alters genome-wide estrogen receptor alpha binding and impacts estrogen signaling in breast cancer;Bhat-Nakshatri,2008

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3