Cargando…

Induced pluripotent stem cell intervention rescues ventricular wall motion disparity, achieving biological cardiac resynchronization post-infarction

Dyssynchronous myocardial motion aggravates cardiac pump function. Cardiac resynchronization using pacing devices is a standard-of-care in the management of heart failure. Post-infarction, however, scar tissue formation impedes the efficacy of device-based therapy. The present study tests a regenera...

Descripción completa

Detalles Bibliográficos
Autores principales: Yamada, Satsuki, Nelson, Timothy J, Kane, Garvan C, Martinez-Fernandez, Almudena, Crespo-Diaz, Ruben J, Ikeda, Yasuhiro, Perez-Terzic, Carmen, Terzic, Andre
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Blackwell Science Inc 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3779120/
https://www.ncbi.nlm.nih.gov/pubmed/23568891
http://dx.doi.org/10.1113/jphysiol.2013.252288
_version_ 1782285199522922496
author Yamada, Satsuki
Nelson, Timothy J
Kane, Garvan C
Martinez-Fernandez, Almudena
Crespo-Diaz, Ruben J
Ikeda, Yasuhiro
Perez-Terzic, Carmen
Terzic, Andre
author_facet Yamada, Satsuki
Nelson, Timothy J
Kane, Garvan C
Martinez-Fernandez, Almudena
Crespo-Diaz, Ruben J
Ikeda, Yasuhiro
Perez-Terzic, Carmen
Terzic, Andre
author_sort Yamada, Satsuki
collection PubMed
description Dyssynchronous myocardial motion aggravates cardiac pump function. Cardiac resynchronization using pacing devices is a standard-of-care in the management of heart failure. Post-infarction, however, scar tissue formation impedes the efficacy of device-based therapy. The present study tests a regenerative approach aimed at targeting the origin of abnormal motion to prevent dyssynchronous organ failure. Induced pluripotent stem (iPS) cells harbour a reparative potential, and were here bioengineered from somatic fibroblasts reprogrammed with the stemness factors OCT3/4, SOX2, KLF4, and c-MYC. In a murine infarction model, within 30 min of coronary ligation, iPS cells were delivered to mapped infarcted areas. Focal deformation and dysfunction underlying progressive heart failure was resolved prospectively using speckle-tracking imaging. Tracked at high temporal and spatial resolution, regional iPS cell transplantation restored, within 10 days post-infarction, the contractility of targeted infarcted foci and nullified conduction delay in adjacent non-infarcted regions. Local iPS cell therapy, but not delivery of parental fibroblasts or vehicle, prevented or normalized abnormal strain patterns correcting the decrease in peak strain, disparity of time-to-peak strain, and pathological systolic stretch. Focal benefit of iPS cell intervention translated into improved left ventricular conduction and contractility, reduced scar, and reversal of structural remodelling, protecting from organ decompensation. Thus, in ischaemic cardiomyopathy, targeted iPS cell transplantation synchronized failing ventricles, offering a regenerative strategy to achieve biological resynchronization.
format Online
Article
Text
id pubmed-3779120
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher Blackwell Science Inc
record_format MEDLINE/PubMed
spelling pubmed-37791202014-05-22 Induced pluripotent stem cell intervention rescues ventricular wall motion disparity, achieving biological cardiac resynchronization post-infarction Yamada, Satsuki Nelson, Timothy J Kane, Garvan C Martinez-Fernandez, Almudena Crespo-Diaz, Ruben J Ikeda, Yasuhiro Perez-Terzic, Carmen Terzic, Andre J Physiol Research Papers Dyssynchronous myocardial motion aggravates cardiac pump function. Cardiac resynchronization using pacing devices is a standard-of-care in the management of heart failure. Post-infarction, however, scar tissue formation impedes the efficacy of device-based therapy. The present study tests a regenerative approach aimed at targeting the origin of abnormal motion to prevent dyssynchronous organ failure. Induced pluripotent stem (iPS) cells harbour a reparative potential, and were here bioengineered from somatic fibroblasts reprogrammed with the stemness factors OCT3/4, SOX2, KLF4, and c-MYC. In a murine infarction model, within 30 min of coronary ligation, iPS cells were delivered to mapped infarcted areas. Focal deformation and dysfunction underlying progressive heart failure was resolved prospectively using speckle-tracking imaging. Tracked at high temporal and spatial resolution, regional iPS cell transplantation restored, within 10 days post-infarction, the contractility of targeted infarcted foci and nullified conduction delay in adjacent non-infarcted regions. Local iPS cell therapy, but not delivery of parental fibroblasts or vehicle, prevented or normalized abnormal strain patterns correcting the decrease in peak strain, disparity of time-to-peak strain, and pathological systolic stretch. Focal benefit of iPS cell intervention translated into improved left ventricular conduction and contractility, reduced scar, and reversal of structural remodelling, protecting from organ decompensation. Thus, in ischaemic cardiomyopathy, targeted iPS cell transplantation synchronized failing ventricles, offering a regenerative strategy to achieve biological resynchronization. Blackwell Science Inc 2013-09-01 2013-04-08 /pmc/articles/PMC3779120/ /pubmed/23568891 http://dx.doi.org/10.1113/jphysiol.2013.252288 Text en © 2013 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited. https://creativecommons.org/licenses/by/4.0/ © 2013 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Papers
Yamada, Satsuki
Nelson, Timothy J
Kane, Garvan C
Martinez-Fernandez, Almudena
Crespo-Diaz, Ruben J
Ikeda, Yasuhiro
Perez-Terzic, Carmen
Terzic, Andre
Induced pluripotent stem cell intervention rescues ventricular wall motion disparity, achieving biological cardiac resynchronization post-infarction
title Induced pluripotent stem cell intervention rescues ventricular wall motion disparity, achieving biological cardiac resynchronization post-infarction
title_full Induced pluripotent stem cell intervention rescues ventricular wall motion disparity, achieving biological cardiac resynchronization post-infarction
title_fullStr Induced pluripotent stem cell intervention rescues ventricular wall motion disparity, achieving biological cardiac resynchronization post-infarction
title_full_unstemmed Induced pluripotent stem cell intervention rescues ventricular wall motion disparity, achieving biological cardiac resynchronization post-infarction
title_short Induced pluripotent stem cell intervention rescues ventricular wall motion disparity, achieving biological cardiac resynchronization post-infarction
title_sort induced pluripotent stem cell intervention rescues ventricular wall motion disparity, achieving biological cardiac resynchronization post-infarction
topic Research Papers
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3779120/
https://www.ncbi.nlm.nih.gov/pubmed/23568891
http://dx.doi.org/10.1113/jphysiol.2013.252288
work_keys_str_mv AT yamadasatsuki inducedpluripotentstemcellinterventionrescuesventricularwallmotiondisparityachievingbiologicalcardiacresynchronizationpostinfarction
AT nelsontimothyj inducedpluripotentstemcellinterventionrescuesventricularwallmotiondisparityachievingbiologicalcardiacresynchronizationpostinfarction
AT kanegarvanc inducedpluripotentstemcellinterventionrescuesventricularwallmotiondisparityachievingbiologicalcardiacresynchronizationpostinfarction
AT martinezfernandezalmudena inducedpluripotentstemcellinterventionrescuesventricularwallmotiondisparityachievingbiologicalcardiacresynchronizationpostinfarction
AT crespodiazrubenj inducedpluripotentstemcellinterventionrescuesventricularwallmotiondisparityachievingbiologicalcardiacresynchronizationpostinfarction
AT ikedayasuhiro inducedpluripotentstemcellinterventionrescuesventricularwallmotiondisparityachievingbiologicalcardiacresynchronizationpostinfarction
AT perezterziccarmen inducedpluripotentstemcellinterventionrescuesventricularwallmotiondisparityachievingbiologicalcardiacresynchronizationpostinfarction
AT terzicandre inducedpluripotentstemcellinterventionrescuesventricularwallmotiondisparityachievingbiologicalcardiacresynchronizationpostinfarction