Cargando…

Single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction

AIMS: A better understanding of the pathways that regulate regeneration of the coronary vasculature is of fundamental importance for the advancement of strategies to treat patients with heart disease. Here, we aimed to investigate the origin and clonal dynamics of endothelial cells (ECs) associated...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Ziwen, Solomonidis, Emmanouil G, Meloni, Marco, Taylor, Richard S, Duffin, Rodger, Dobie, Ross, Magalhaes, Marlene S, Henderson, Beth E P, Louwe, Pieter A, D’Amico, Gabriela, Hodivala-Dilke, Kairbaan M, Shah, Ajay M, Mills, Nicholas L, Simons, Benjamin D, Gray, Gillian A, Henderson, Neil C, Baker, Andrew H, Brittan, Mairi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Oxford University Press 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6685329/
https://www.ncbi.nlm.nih.gov/pubmed/31162546
http://dx.doi.org/10.1093/eurheartj/ehz305
_version_ 1783442382842232832
author Li, Ziwen
Solomonidis, Emmanouil G
Meloni, Marco
Taylor, Richard S
Duffin, Rodger
Dobie, Ross
Magalhaes, Marlene S
Henderson, Beth E P
Louwe, Pieter A
D’Amico, Gabriela
Hodivala-Dilke, Kairbaan M
Shah, Ajay M
Mills, Nicholas L
Simons, Benjamin D
Gray, Gillian A
Henderson, Neil C
Baker, Andrew H
Brittan, Mairi
author_facet Li, Ziwen
Solomonidis, Emmanouil G
Meloni, Marco
Taylor, Richard S
Duffin, Rodger
Dobie, Ross
Magalhaes, Marlene S
Henderson, Beth E P
Louwe, Pieter A
D’Amico, Gabriela
Hodivala-Dilke, Kairbaan M
Shah, Ajay M
Mills, Nicholas L
Simons, Benjamin D
Gray, Gillian A
Henderson, Neil C
Baker, Andrew H
Brittan, Mairi
author_sort Li, Ziwen
collection PubMed
description AIMS: A better understanding of the pathways that regulate regeneration of the coronary vasculature is of fundamental importance for the advancement of strategies to treat patients with heart disease. Here, we aimed to investigate the origin and clonal dynamics of endothelial cells (ECs) associated with neovascularization in the adult mouse heart following myocardial infarction (MI). Furthermore, we sought to define murine cardiac endothelial heterogeneity and to characterize the transcriptional profiles of pro-angiogenic resident ECs in the adult mouse heart, at single-cell resolution. METHODS AND RESULTS: An EC-specific multispectral lineage-tracing mouse (Pdgfb-iCreER(T2)-R26R-Brainbow2.1) was used to demonstrate that structural integrity of adult cardiac endothelium following MI was maintained through clonal proliferation by resident ECs in the infarct border region, without significant contributions from bone marrow cells or endothelial-to-mesenchymal transition. Ten transcriptionally discrete heterogeneous EC states, as well as the pathways through which each endothelial state is likely to enhance neovasculogenesis and tissue regeneration following ischaemic injury were defined. Plasmalemma vesicle-associated protein (Plvap) was selected for further study, which showed an endothelial-specific and increased expression in both the ischaemic mouse and human heart, and played a direct role in regulating human endothelial proliferation in vitro. CONCLUSION: We present a single-cell gene expression atlas of cardiac specific resident ECs, and the transcriptional hierarchy underpinning endogenous vascular repair following MI. These data provide a rich resource that could assist in the development of new therapeutic interventions to augment endogenous myocardial perfusion and enhance regeneration in the injured heart.
format Online
Article
Text
id pubmed-6685329
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Oxford University Press
record_format MEDLINE/PubMed
spelling pubmed-66853292019-08-12 Single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction Li, Ziwen Solomonidis, Emmanouil G Meloni, Marco Taylor, Richard S Duffin, Rodger Dobie, Ross Magalhaes, Marlene S Henderson, Beth E P Louwe, Pieter A D’Amico, Gabriela Hodivala-Dilke, Kairbaan M Shah, Ajay M Mills, Nicholas L Simons, Benjamin D Gray, Gillian A Henderson, Neil C Baker, Andrew H Brittan, Mairi Eur Heart J Basic Science AIMS: A better understanding of the pathways that regulate regeneration of the coronary vasculature is of fundamental importance for the advancement of strategies to treat patients with heart disease. Here, we aimed to investigate the origin and clonal dynamics of endothelial cells (ECs) associated with neovascularization in the adult mouse heart following myocardial infarction (MI). Furthermore, we sought to define murine cardiac endothelial heterogeneity and to characterize the transcriptional profiles of pro-angiogenic resident ECs in the adult mouse heart, at single-cell resolution. METHODS AND RESULTS: An EC-specific multispectral lineage-tracing mouse (Pdgfb-iCreER(T2)-R26R-Brainbow2.1) was used to demonstrate that structural integrity of adult cardiac endothelium following MI was maintained through clonal proliferation by resident ECs in the infarct border region, without significant contributions from bone marrow cells or endothelial-to-mesenchymal transition. Ten transcriptionally discrete heterogeneous EC states, as well as the pathways through which each endothelial state is likely to enhance neovasculogenesis and tissue regeneration following ischaemic injury were defined. Plasmalemma vesicle-associated protein (Plvap) was selected for further study, which showed an endothelial-specific and increased expression in both the ischaemic mouse and human heart, and played a direct role in regulating human endothelial proliferation in vitro. CONCLUSION: We present a single-cell gene expression atlas of cardiac specific resident ECs, and the transcriptional hierarchy underpinning endogenous vascular repair following MI. These data provide a rich resource that could assist in the development of new therapeutic interventions to augment endogenous myocardial perfusion and enhance regeneration in the injured heart. Oxford University Press 2019-08-07 2019-06-04 /pmc/articles/PMC6685329/ /pubmed/31162546 http://dx.doi.org/10.1093/eurheartj/ehz305 Text en © The Author(s) 2019. Published by Oxford University Press on behalf of the European Society of Cardiology. http://creativecommons.org/licenses/by/4.0/ This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Basic Science
Li, Ziwen
Solomonidis, Emmanouil G
Meloni, Marco
Taylor, Richard S
Duffin, Rodger
Dobie, Ross
Magalhaes, Marlene S
Henderson, Beth E P
Louwe, Pieter A
D’Amico, Gabriela
Hodivala-Dilke, Kairbaan M
Shah, Ajay M
Mills, Nicholas L
Simons, Benjamin D
Gray, Gillian A
Henderson, Neil C
Baker, Andrew H
Brittan, Mairi
Single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction
title Single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction
title_full Single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction
title_fullStr Single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction
title_full_unstemmed Single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction
title_short Single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction
title_sort single-cell transcriptome analyses reveal novel targets modulating cardiac neovascularization by resident endothelial cells following myocardial infarction
topic Basic Science
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6685329/
https://www.ncbi.nlm.nih.gov/pubmed/31162546
http://dx.doi.org/10.1093/eurheartj/ehz305
work_keys_str_mv AT liziwen singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT solomonidisemmanouilg singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT melonimarco singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT taylorrichards singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT duffinrodger singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT dobieross singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT magalhaesmarlenes singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT hendersonbethep singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT louwepietera singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT damicogabriela singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT hodivaladilkekairbaanm singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT shahajaym singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT millsnicholasl singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT simonsbenjamind singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT graygilliana singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT hendersonneilc singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT bakerandrewh singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction
AT brittanmairi singlecelltranscriptomeanalysesrevealnoveltargetsmodulatingcardiacneovascularizationbyresidentendothelialcellsfollowingmyocardialinfarction