Uncovering the dosage-dependent roles of Arid1a in gastric tumorigenesis for combinatorial drug therapy

Author:

Loe Adrian Kwan Ho12ORCID,Francis Roshane12ORCID,Seo Jieun123ORCID,Du Lutao45ORCID,Wang Yunshan45ORCID,Kim Ji-Eun12ORCID,Hakim Shaheed W.6ORCID,Kim Jung-Eun12ORCID,He Housheng Hansen78ORCID,Guo Haiyang457ORCID,Kim Tae-Hee12ORCID

Affiliation:

1. Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada

2. Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada

3. Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea

4. Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, China

5. Tumor Marker Detection Engineering Laboratory of Shandong Province, Jinan, Shandong, China

6. St. Joseph’s Health Centre, Unity Health Toronto, Toronto, Ontario, Canada

7. Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada

8. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada

Abstract

Gastric cancer (GC) is one of the most common deadly cancers in the world. Although patient genomic data have identified AT-rich interaction domain 1A (ARID1A), a key chromatin remodeling complex subunit, as the second most frequently mutated gene after TP53, its in vivo role and relationship to TP53 in gastric tumorigenesis remains unclear. Establishing a novel mouse model that reflects the ARID1A heterozygous mutations found in the majority of human GC cases, we demonstrated that Arid1a heterozygosity facilitates tumor progression through a global loss of enhancers and subsequent suppression of the p53 and apoptosis pathways. Moreover, mouse genetic and single-cell analyses demonstrated that the homozygous deletion of Arid1a confers a competitive disadvantage through the activation of the p53 pathway, highlighting its distinct dosage-dependent roles. Using this unique vulnerability of Arid1a mutated GC cells, our combined treatment with the epigenetic inhibitor, TP064, and the p53 agonist, Nutlin-3, inhibited growth of Arid1a heterozygous tumor organoids, providing a novel therapeutic option for GC.

Funder

SickKids Foundation

Canadian Institutes of Health Research

Cancer Research Society

Concern Foundation

SickKids Restracomp

University of Toronto

Taishan Scholar Program of Shandong Province

National Key Research and Development Project of China

Natural Sciences and Engineering Research Council of Canada

Ontario Ministry of Research, Innovation and Science

SickKids Garron Family Cancer Centre Pitblado Basic/Translational Discovery

Publisher

Rockefeller University Press

Subject

Immunology,Immunology and Allergy

Reference59 articles.

1. Loss of ARID1A, ARID1B, and ARID2 expression during progression of gastric cancer;Aso;Anticancer Res.,2015

2. Use of mononucleotide repeat markers for detection of microsatellite instability in mouse tumors;Bacher;Mol. Carcinog.,2005

3. ARID1A-mutated ovarian cancers depend on HDAC6 activity;Bitler;Nat. Cell Biol.,2017

4. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data;Cerami;Cancer Discov.,2012

5. The gastric precancerous cascade;Correa;J. Dig. Dis.,2012

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