Please use this identifier to cite or link to this item: http://hdl.handle.net/2445/179846
Full metadata record
DC FieldValueLanguage
dc.contributor.authorMonguió Tortajada, Marta-
dc.contributor.authorPrat Vidal, Cristina-
dc.contributor.authorMoron Font, Miriam-
dc.contributor.authorClos Sansalvador, Marta-
dc.contributor.authorCalle, Alexandra-
dc.contributor.authorGastelurrutia, Paloma-
dc.contributor.authorCserkoova, Adriana-
dc.contributor.authorMorancho, Anna-
dc.contributor.authorRamírez, Miguel Ángel-
dc.contributor.authorRosell, Anna-
dc.contributor.authorBayés Genís, Antoni-
dc.contributor.authorGálvez Montón, Carolina-
dc.contributor.authorBorràs, Francesc E.-
dc.contributor.authorRoura, Santiago-
dc.date.accessioned2021-09-03T06:44:40Z-
dc.date.available2021-09-03T06:44:40Z-
dc.date.issued2021-07-01-
dc.identifier.urihttp://hdl.handle.net/2445/179846-
dc.description.abstractThe administration of extracellular vesicles (EV) from mesenchymal stromal cells (MSC) is a promising cell-free nanotherapy for tissue repair after myocardial infarction (MI). However, the optimal EV delivery strategy remains undetermined. Here, we designed a novel MSC-EV delivery, using 3D scaffolds engineered from decellularised cardiac tissue as a cell-free product for cardiac repair. EV from porcine cardiac adipose tissue-derived MSC (cATMSC) were purified by size exclusion chromatography (SEC), functionally analysed and loaded to scaffolds. cATMSC-EV markedly reduced polyclonal proliferation and pro-inflammatory cytokines production (IFNγ, TNFα, IL12p40) of allogeneic PBMC. Moreover, cATMSC-EV recruited outgrowth endothelial cells (OEC) and allogeneic MSC, and promoted angiogenesis. Fluorescently labelled cATMSC-EV were mixed with peptide hydrogel, and were successfully retained in decellularised scaffolds. Then, cATMSC-EV-embedded pericardial scaffolds were administered in vivo over the ischemic myocardium in a pig model of MI. Six days from implantation, the engineered scaffold efficiently integrated into the post-infarcted myocardium. cATMSC-EV were detected within the construct and MI core, and promoted an increase in vascular density and reduction in macrophage and T cell infiltration within the damaged myocardium. The confined administration of multifunctional MSC-EV within an engineered pericardial scaffold ensures local EV dosage and release, and generates a vascularised bioactive niche for cell recruitment, engraftment and modulation of short-term post-ischemic inflammation.-
dc.format.extent14 p.-
dc.format.mimetypeapplication/pdf-
dc.language.isoeng-
dc.publisherElsevier BV-
dc.relation.isformatofReproducció del document publicat a: https://doi.org/10.1016/j.bioactmat.2021.02.026-
dc.relation.ispartofBioactive Materials, 2021, vol. 6, num. 10, p. 3314-3327-
dc.relation.urihttps://doi.org/10.1016/j.bioactmat.2021.02.026-
dc.rightscc by-nc-nd (c) Monguió Tortajada, Marta et al., 2021-
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/3.0/es/*
dc.sourceArticles publicats en revistes (Institut d'lnvestigació Biomèdica de Bellvitge (IDIBELL))-
dc.subject.classificationEnginyeria de teixits-
dc.subject.classificationInfart de miocardi-
dc.subject.otherTissue engineering-
dc.subject.otherMyocardial infarction-
dc.titleLocal administration of porcine immunomodulatory, chemotactic and angiogenic extracellular vesicles using engineered cardiac scaffolds for myocardial infarction-
dc.typeinfo:eu-repo/semantics/article-
dc.typeinfo:eu-repo/semantics/publishedVersion-
dc.date.updated2021-07-29T08:15:23Z-
dc.rights.accessRightsinfo:eu-repo/semantics/openAccess-
dc.identifier.pmid33778207-
Appears in Collections:Articles publicats en revistes (Institut d'lnvestigació Biomèdica de Bellvitge (IDIBELL))

Files in This Item:
File Description SizeFormat 
1-s2.0-S2452199X21000840-main.pdf12.54 MBAdobe PDFView/Open


This item is licensed under a Creative Commons License Creative Commons