Inhibition of ATR opposes glioblastoma invasion through disruption of cytoskeletal networks and integrin internalization via macropinocytosis

Author:

Derby Sarah J1,Dutton Louise1,Strathdee Karen E1,Stevenson Katrina1,Koessinger Anna12,Jackson Mark1,Tian Yuling1,Yu Wenxi1,Mclay Kathy1,Misquitta Josette1,Alsharif Sama1,Clarke Cassie J2,Gilmour Lesley1,Thomason Peter2,McGhee Ewan2,McGarrity-Cottrell Connor L3,Vanderlinden Aurelie3,Collis Spencer J3,Rominyi Ola3,Lemgruber Leandro4,Solecki Gergely5,Olson Michael6,Winkler Frank5ORCID,Carlin Leo M12,Heiland Dieter Henrik7,Inman Gareth J12,Chalmers Anthony J1,Norman Jim C12ORCID,Carruthers Ross1,Birch Joanna L1

Affiliation:

1. Wolfson Wohl Translational Cancer Research Centre, School of Cancer Sciences, University of Glasgow , Glasgow , UK

2. CRUK Scotland Institute , Glasgow , UK

3. Department of Oncology and Metabolism, The University of Sheffield Medical School , Sheffield , UK

4. Cellular Analysis Facility, College of Medical, Veterinary & Life Sciences, University of Glasgow , Glasgow , UK

5. German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) , Heidelberg , Germany

6. Department of Chemistry and Biology, Ryeson University , Toronto, Ontario , Canada

7. MILO Laboratory, Department of Neurosurgery , Freiburg , Germany

Abstract

Abstract Background Glioblastomas have highly infiltrative growth patterns that contribute to recurrence and poor survival. Despite infiltration being a critical therapeutic target, no clinically useful therapies exist that counter glioblastoma invasion. Here, we report that inhibition of ataxia telangiectasia and Rad 3 related kinase (ATR) reduces invasion of glioblastoma cells through dysregulation of cytoskeletal networks and subsequent integrin trafficking. Methods Glioblastoma motility and invasion were assessed in vitro and in vivo in response to ATR inhibition (ATRi) and ATR overexpression using time-lapse microscopy, two orthotopic glioblastoma models, and intravital imaging. Disruption to cytoskeleton networks and endocytic processing were investigated via high-throughput, super-resolution and intravital imaging. Results High ATR expression was associated with significantly poorer survival in clinical datasets while histological, protein expression, and spatial transcriptomics using glioblastoma tumor specimens revealed higher ATR expression at infiltrative margins. Pharmacological inhibition with two different compounds and RNAi targeting of ATR opposed the invasion of glioblastoma, whereas overexpression of ATR drove migration. Subsequent investigation revealed that cytoskeletal dysregulation reduced macropinocytotic internalization of integrins at growth-cone-like structures, resulting in a tumor microtube retraction defect. The biological relevance and translational potential of these findings were confirmed using two orthotopic in vivo models of glioblastoma and intravital imaging. Conclusions We demonstrate a novel role for ATR in determining invasion in glioblastoma cells and propose that pharmacological targeting of ATR could have far-reaching clinical benefits beyond radiosensitization.

Funder

UK Research and Innovation Future Leaders

Cancer Research UK

CRUK Institute

CRUK Fellowship

National Centre for the Replacement Refinement and Reduction of Animals in Research

Brain Tumour Charity

Publisher

Oxford University Press (OUP)

Subject

Cancer Research,Neurology (clinical),Oncology

Cited by 1 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

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