Mechanical stress during confined migration causes aberrant mitoses and c-MYC amplification

Author:

Bastianello Giulia12ORCID,Kidiyoor Gururaj Rao1ORCID,Lowndes Conor1ORCID,Li Qingsen1,Bonnal Raoul1,Godwin Jeffrey1ORCID,Iannelli Fabio1,Drufuca Lorenzo2,Bason Ramona1ORCID,Orsenigo Fabrizio1ORCID,Parazzoli Dario1,Pavani Mattia1ORCID,Cancila Valeria3,Piccolo Stefano14ORCID,Scita Giorgio12ORCID,Ciliberto Andrea1,Tripodo Claudio13ORCID,Pagani Massimiliano12ORCID,Foiani Marco156

Affiliation:

1. Istituto Fondazione Italiana per la Ricerca sul Cancro di Oncologia molecolare—the Associazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology, Milano 20139, Italy

2. Università degli Studi di Milano, Milan 20122, Italy

3. Tumor Immunology Unit, Department of Health Science, University of Palermo School of Medicine, Palermo 90133, Italy

4. Department of Molecular Medicine, University of Padua, Padua 35123, Italy

5. Istituto di Genetica Molecolare, Centro Nazionale Ricerca, Pavia 27100, Italy

6. Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore

Abstract

Confined cell migration hampers genome integrity and activates the ATR and ATM mechano-transduction pathways. We investigated whether the mechanical stress generated by metastatic interstitial migration contributes to the enhanced chromosomal instability observed in metastatic tumor cells. We employed live cell imaging, micro-fluidic approaches, and scRNA-seq to follow the fate of tumor cells experiencing confined migration. We found that, despite functional ATR, ATM, and spindle assembly checkpoint (SAC) pathways, tumor cells dividing across constriction frequently exhibited altered spindle pole organization, chromosome mis-segregations, micronuclei formation, chromosome fragility, high gene copy number variation, and transcriptional de-regulation and up-regulation of c-MYC oncogenic transcriptional signature via c-MYC locus amplifications. In vivo tumor settings showed that malignant cells populating metastatic foci or infiltrating the interstitial stroma gave rise to cells expressing high levels of c-MYC. Altogether, our data suggest that mechanical stress during metastatic migration contributes to override the checkpoint controls and boosts genotoxic and oncogenic events. Our findings may explain why cancer aneuploidy often does not correlate with mutations in SAC genes and why c-MYC amplification is strongly linked to metastatic tumors.

Funder

Fondazione AIRC per la ricerca sul cancro ETS

Publisher

Proceedings of the National Academy of Sciences

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