Utilize este identificador para referenciar este registo: https://hdl.handle.net/1822/24885

Registo completo
Campo DCValorIdioma
dc.contributor.authorHamm, Alexander-
dc.contributor.authorVeschini, Lorenzo-
dc.contributor.authorTakeda, Yukiji-
dc.contributor.authorCosta, Sandra Maria Araújo da-
dc.contributor.authorDelamarre, Estelle-
dc.contributor.authorSquadrito, Mario Leonardo-
dc.contributor.authorHenze, Anne-Theres-
dc.contributor.authorWenes, Mathias-
dc.contributor.authorSerneels, Jens-
dc.contributor.authorPucci, Ferdinando-
dc.contributor.authorRoncal, Carmen-
dc.contributor.authorAnisimov, Andrey-
dc.contributor.authorAlitalo, Kari-
dc.contributor.authorDe Palma, Michele-
dc.contributor.authorMazzone, Massimiliano-
dc.date.accessioned2013-07-30T13:15:27Z-
dc.date.available2013-07-30T13:15:27Z-
dc.date.issued2013-
dc.date.submitted2013-
dc.identifier.issn1757-4676por
dc.identifier.urihttps://hdl.handle.net/1822/24885-
dc.description.abstractOcclusion of the main arterial route redirects blood flow to the collateral circulation. We previously reported that macrophages genetically modified to express low levels of prolyl hydroxylase domain protein 2 (PHD2) display an arteriogenic phenotype, which promotes the formation of collateral vessels and protects the skeletal muscle from ischaemic necrosis. However, the molecular mechanisms underlying this process are unknown. Here, we demonstrate that femoral artery occlusion induces a switch in macrophage phenotype through angiopoietin-1 (ANG1)-mediated Phd2 repression. ANG blockade by a soluble trap prevented the downregulation of Phd2 expression in macrophages and their phenotypic switch, thus inhibiting collateral growth. ANG1-dependent Phd2 repression initiated a feed-forward loop mediated by the induction of the ANG receptor TIE2 in macrophages. Gene silencing and cell depletion strategies demonstrate that TIE2 induction in macrophages is required to promote their proarteriogenic functions, enabling collateral vessel formation following arterial obstruction. These results indicate an indispensable role for TIE2 in sustaining in situ programming of macrophages to a proarteriogenic, M2-like phenotype, suggesting possible new venues for the treatment of ischaemic disorders.por
dc.description.sponsorshipThe authors thank Y. Jonsson for technical assistance. The authors are thankful to P. Carmeliet for scientific discussion and support. AH was granted by Deutsche Forschungsgemeinschaft (DFG), LV by the European Society of Cardiology (ESC), SC by Fundacao para a Ciencia e a Tecnologia (FCT), ED by Fondation ARC pour la recherche sur le cancer, A-T.H by Fonds Wetenschappelijk Onderzoek (FWO). This work was supported by grants from the Fonds Wetenschappelijk Onderzoek (FWO G.0726.10 to MM) and the European Research Council (OxyMO to MM and Tie2<SUP>+</SUP> Monocytes to MDP).por
dc.language.isoengpor
dc.publisherJohn Wiley and Sonspor
dc.rightsopenAccesspor
dc.subjectArteriogenesispor
dc.subjectIschaemiapor
dc.subjectMacrophagespor
dc.subjectPHD2por
dc.subjectTIE2por
dc.titlePHD2 regulates arteriogenic macrophages through TIE2 signalingpor
dc.typearticle-
dc.peerreviewedyespor
dc.relation.publisherversionwww.onlinelibrary.wiley.compor
sdum.publicationstatuspublishedpor
oaire.citationStartPage1por
oaire.citationEndPage15por
oaire.citationIssue6por
oaire.citationTitleEMBO Molecular Medicinepor
oaire.citationVolume5por
dc.date.updated2013-07-15T09:44:16Z-
dc.identifier.doi10.1002/emmm.201302695-
dc.identifier.pmid23616286por
dc.subject.wosScience & Technologypor
sdum.journalEmbo Molecular Medicinepor
Aparece nas coleções:ICVS - Artigos em revistas internacionais / Papers in international journals

Ficheiros deste registo:
Ficheiro Descrição TamanhoFormato 
201302695_ftp.pdf1,78 MBAdobe PDFVer/Abrir

Partilhe no FacebookPartilhe no TwitterPartilhe no DeliciousPartilhe no LinkedInPartilhe no DiggAdicionar ao Google BookmarksPartilhe no MySpacePartilhe no Orkut
Exporte no formato BibTex mendeley Exporte no formato Endnote Adicione ao seu ORCID