Show simple item record

dc.contributor.authorRoss, JA
dc.contributor.authorArcos-Villacis, N
dc.contributor.authorBattey, E
dc.contributor.authorBoogerd, C
dc.contributor.authorOrellana, CA
dc.contributor.authorMarhuenda, E
dc.contributor.authorSwiatlowska, P
dc.contributor.authorHodzic, D
dc.contributor.authorPrin, F
dc.contributor.authorMohun, T
dc.contributor.authorCatibog, N
dc.contributor.authorTapia, O
dc.contributor.authorGerace, L
dc.contributor.authorIskratsch, T
dc.contributor.authorShah, AM
dc.contributor.authorStroud, MJ
dc.date.accessioned2024-01-18T10:11:52Z
dc.date.available2023-02-08
dc.date.available2024-01-18T10:11:52Z
dc.date.issued2023
dc.identifier.issn0008-6363
dc.identifier.urihttps://qmro.qmul.ac.uk/xmlui/handle/123456789/94022
dc.description.abstractAims: Nuclear envelope integrity is essential for the compartmentalization of the nucleus and cytoplasm. Importantly, mutations in genes encoding nuclear envelope (NE) and associated proteins are the second highest cause of familial dilated cardiomyopathy. One such NE protein that causes cardiomyopathy in humans and affects mouse heart development is Lem2. However, its role in the heart remains poorly understood. Methods and results: We generated mice in which Lem2 was specifically ablated either in embryonic cardiomyocytes (Lem2 cKO) or in adult cardiomyocytes (Lem2 iCKO) and carried out detailed physiological, tissue, and cellular analyses. High-resolution episcopic microscopy was used for three-dimensional reconstructions and detailed morphological analyses. RNA-sequencing and immunofluorescence identified altered pathways and cellular phenotypes, and cardiomyocytes were isolated to interrogate nuclear integrity in more detail. In addition, echocardiography provided a physiological assessment of Lem2 iCKO adult mice. We found that Lem2 was essential for cardiac development, and hearts from Lem2 cKO mice were morphologically and transcriptionally underdeveloped. Lem2 cKO hearts displayed high levels of DNA damage, nuclear rupture, and apoptosis. Crucially, we found that these defects were driven by muscle contraction as they were ameliorated by inhibiting myosin contraction and L-type calcium channels. Conversely, reducing Lem2 levels to ∼45% in adult cardiomyocytes did not lead to overt cardiac dysfunction up to 18 months of age. Conclusions: Our data suggest that Lem2 is critical for integrity at the nascent NE in foetal hearts, and protects the nucleus from the mechanical forces of muscle contraction. In contrast, the adult heart is not detectably affected by partial Lem2 depletion, perhaps owing to a more established NE and increased adaptation to mechanical stress. Taken together, these data provide insights into mechanisms underlying cardiomyopathy in patients with mutations in Lem2 and cardio-laminopathies in general.en_US
dc.format.extent2074 - 2088
dc.publisherOxford University Pressen_US
dc.relation.ispartofCARDIOVASCULAR RESEARCH
dc.rightsThis is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
dc.rightsAttribution 3.0 United States*
dc.rights.urihttp://creativecommons.org/licenses/by/3.0/us/*
dc.subjectHeart failureen_US
dc.subjectCardiac developmenten_US
dc.subjectNuclear Envelopeen_US
dc.subjectLINC complexen_US
dc.subjectLaminopathyen_US
dc.titleLem2 is essential for cardiac development by maintaining nuclear integrityen_US
dc.typeArticleen_US
dc.rights.holder© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology.
dc.identifier.doi10.1093/cvr/cvad061
pubs.author-urlhttps://www.webofscience.com/api/gateway?GWVersion=2&SrcApp=PARTNER_APP&SrcAuth=LinksAMR&KeyUT=WOS:000979127000001&DestLinkType=FullRecord&DestApp=ALL_WOS&UsrCustomerID=612ae0d773dcbdba3046f6df545e9f6aen_US
pubs.issue11en_US
pubs.notesNot knownen_US
pubs.publication-statusPublisheden_US
pubs.volume119en_US
rioxxterms.funderDefault funderen_US
rioxxterms.identifier.projectDefault projecten_US
qmul.funderInvestigating the cardiomyocyte rigidity sensing mechanism with micro patterned surfaces and nanopillars::Biotechnology and Biological Sciences Research Councilen_US
qmul.funderInvestigating the cardiomyocyte rigidity sensing mechanism with micro patterned surfaces and nanopillars::Biotechnology and Biological Sciences Research Councilen_US


Files in this item

Thumbnail
Thumbnail

This item appears in the following Collection(s)

Show simple item record

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com
Except where otherwise noted, this item's license is described as This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com