COMMD3 loss drives invasive breast cancer growth by modulating copper homeostasis

Author:

Hancock Janelle L1,Kalimutho Murugan2,Straube Jasmin1,Lim Malcolm3,Gresshoff Irma3,Saunus Jodi M3,Lee Jason1,Lakhani Sunil4,Simpson Kaylene J5,Bush Ashley I6,Anderson Robin7,Khanna Kum Kum1

Affiliation:

1. QIMR Berghofer: QIMR Berghofer Medical Research Institute

2. QIMR Berghofer Medical Research Institute

3. University Of Queensland UQCCR: The University of Queensland Centre for Clinical Research

4. UQCCR: The University of Queensland Centre for Clinical Research

5. University of Melbourne Victorian Comprehensive Cancer Centre

6. Florey Neuroscience Institutes: The Florey Institute of Neuroscience and Mental Health

7. Olivia Newton-John Cancer Centre at Austin Health: Olivia Newton-John Cancer Wellness & Research Centre

Abstract

Abstract Background Despite overall improvement in breast cancer patient outcomes from earlier diagnosis and personalised treatment approaches, some patients continue to experience recurrence and incurable metastases. It is therefore imperative to understand the molecular changes that allow transition from a non-aggressive state to a more aggressive phenotype. This transition is governed by a number of factors. Methods As crosstalk with extracellular matrix (ECM) is critical for tumour cell growth and survival, we applied high throughput shRNA screening on a validated ‘3D on-top cellular assay’ to identify novel growth suppressive mechanisms. Results A number of novel candidate genes were identified. We focused on COMMD3, a previously uncharacterised gene that suppressed invasive growth of ER + breast cancer cells in the cellular assay. Analysis of published expression data suggested that COMMD3 is normally expressed in the mammary ducts and lobules, that expression is lost in some tumours and that loss is associated with lower survival probabilities. We performed immunohistochemical analysis of an independent tumour cohort to investigate relationships between COMMD3 protein expression, phenotypic markers and disease-specific survival. This revealed an association between COMMD3 loss and shorter survival in hormone-dependent breast cancers and particularly luminal-A-like tumours (ER+/Ki67-low; 10-year survival probability 0.83 vs 0.73 for COMMD3-positive and -negative cases, respectively). Expression of COMMD3 in luminal-A-like tumours was directly associated with markers of luminal differentiation: c-KIT, ELF5, androgen receptor and tubule formation (the extent of normal glandular architecture; p < 0.05). Consistent with this, knockdown of COMMD3 induced invasive spheroid growth in ER + breast cancer cell lines in vitro, while Commd3 knockdown in the relatively indolent 4T07 TNBC mouse cell line promoted tumour expansion in syngeneic Balb/c hosts. Notably, RNA sequencing revealed a role for COMMD3 in copper signalling, via regulation of the Na+/K+-ATPase subunit, ATP1B1. Treatment of COMMD3-knockdown cells with the copper chelator, tetrathiomolybdate, significantly reduced invasive spheroid growth via induction of apoptosis. Conclusion Overall, we found that COMMD3 loss promoted-aggressive behaviour in breast cancer cells.

Publisher

Research Square Platform LLC

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