Chromatin compartment dynamics in a haploinsufficient model of cardiac laminopathy

Author:

Bertero Alessandro123ORCID,Fields Paul A.123ORCID,Smith Alec S.T.234,Leonard Andrea235,Beussman Kevin235ORCID,Sniadecki Nathan J.2345ORCID,Kim Deok-Ho234,Tse Hung-Fat6,Pabon Lil123ORCID,Shendure Jay78,Noble William S.7,Murry Charles E.12349ORCID

Affiliation:

1. Department of Pathology, University of Washington, Seattle, WA

2. Center for Cardiovascular Biology, University of Washington, Seattle, WA

3. Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA

4. Department of Bioengineering, University of Washington, Seattle, WA

5. Department of Mechanical Engineering, University of Washington, Seattle, WA

6. Cardiology Division, Department of Medicine, University of Hong Kong, Pok Fu Lam, Hong Kong

7. Department of Genome Sciences, University of Washington, Seattle, WA

8. Howard Hughes Medical Institute, Seattle, WA

9. Department of Medicine/Cardiology, University of Washington, Seattle, WA

Abstract

Mutations in A-type nuclear lamins cause dilated cardiomyopathy, which is postulated to result from dysregulated gene expression due to changes in chromatin organization into active and inactive compartments. To test this, we performed genome-wide chromosome conformation analyses in human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs) with a haploinsufficient mutation for lamin A/C. Compared with gene-corrected cells, mutant hiPSC-CMs have marked electrophysiological and contractile alterations, with modest gene expression changes. While large-scale changes in chromosomal topology are evident, differences in chromatin compartmentalization are limited to a few hotspots that escape segregation to the nuclear lamina and inactivation during cardiogenesis. These regions exhibit up-regulation of multiple noncardiac genes including CACNA1A, encoding for neuronal P/Q-type calcium channels. Pharmacological inhibition of the resulting current partially mitigates the electrical alterations. However, chromatin compartment changes do not explain most gene expression alterations in mutant hiPSC-CMs. Thus, global errors in chromosomal compartmentation are not the primary pathogenic mechanism in heart failure due to lamin A/C haploinsufficiency.

Funder

European Molecular Biology Organization

National Institutes of Health

National Science Foundation

Foundation Leducq Transatlantic Network of Excellence

Publisher

Rockefeller University Press

Subject

Cell Biology

Reference113 articles.

1. Structure and properties of omega-agatoxin IVB, a new antagonist of P-type calcium channels;Adams;Mol. Pharmacol.,1993

2. Blank spots on the map: some current questions on nuclear organization and genome architecture;Adriaens;Histochem. Cell Biol.,2018

3. Genetics and genotype-phenotype correlations in Finnish patients with dilated cardiomyopathy;Akinrinade;Eur. Heart J.,2015

4. Nuclear lamins are not required for lamina-associated domain organization in mouse embryonic stem cells;Amendola;EMBO Rep.,2015

5. Cardiac ion channels in health and disease;Amin;Heart Rhythm.,2010

Cited by 42 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