Näytä suppeat kuvailutiedot

dc.contributor.authorMagga, Johanna
dc.contributor.authorVainio, Laura
dc.contributor.authorKilpiö, Teemu
dc.contributor.authorHulmi, Juha
dc.contributor.authorTaponen, Saija
dc.contributor.authorLin, Ruizhu
dc.contributor.authorRäsänen, Markus
dc.contributor.authorSzabó, Zoltán
dc.contributor.authorGao, Erhe
dc.contributor.authorRahtu-Korpela, Lea
dc.contributor.authorAlakoski, Tarja
dc.contributor.authorUlvila, Johanna
dc.contributor.authorLaitinen, Mika
dc.contributor.authorPasternack, Arja
dc.contributor.authorKoch, Walter J.
dc.contributor.authorAlitalo, Kari
dc.contributor.authorKivelä, Riikka
dc.contributor.authorRitvos, Olli
dc.contributor.authorKerkelä, Risto
dc.date.accessioned2019-03-18T07:08:30Z
dc.date.available2019-03-18T07:08:30Z
dc.date.issued2019
dc.identifier.citationMagga, J., Vainio, L., Kilpiö, T., Hulmi, J., Taponen, S., Lin, R., Räsänen, M., Szabó, Z., Gao, E., Rahtu-Korpela, L., Alakoski, T., Ulvila, J., Laitinen, M., Pasternack, A., Koch, W. J., Alitalo, K., Kivelä, R., Ritvos, O., & Kerkelä, R. (2019). Systemic blockade of ACVR2B ligands protects myocardium from acute ischemia-reperfusion injury. <i>Molecular Therapy</i>, <i>27</i>(3), 600-610. <a href="https://doi.org/10.1016/j.ymthe.2019.01.013" target="_blank">https://doi.org/10.1016/j.ymthe.2019.01.013</a>
dc.identifier.otherCONVID_28896998
dc.identifier.otherTUTKAID_80514
dc.identifier.urihttps://jyx.jyu.fi/handle/123456789/63160
dc.description.abstractActivin A and myostatin, members of the transforming growth factor (TGF)-β superfamily of secreted factors, are potent negative regulators of muscle growth, but their contribution to myocardial ischemia-reperfusion (IR) injury is not known. The aim of this study was to investigate if activin 2B (ACVR2B) receptor ligands contribute to myocardial IR injury. Mice were treated with soluble ACVR2B decoy receptor (ACVR2B-Fc) and subjected to myocardial ischemia followed by reperfusion for 6 or 24 h. Systemic blockade of ACVR2B ligands by ACVR2B-Fc was protective against cardiac IR injury, as evidenced by reduced infarcted area, apoptosis, and autophagy and better preserved LV systolic function following IR. ACVR2B-Fc modified cardiac metabolism, LV mitochondrial respiration, as well as cardiac phenotype toward physiological hypertrophy. Similar to its protective role in IR injury in vivo, ACVR2B-Fc antagonized SMAD2 signaling and cell death in cardiomyocytes that were subjected to hypoxic stress. ACVR2B ligand myostatin was found to exacerbate hypoxic stress. In addition to acute cardioprotection in ischemia, ACVR2B-Fc provided beneficial effects on cardiac function in prolonged cardiac stress in cardiotoxicity model. By blocking myostatin, ACVR2B-Fc potentially reduces cardiomyocyte death and modifies cardiomyocyte metabolism for hypoxic conditions to protect the heart from IR injury.fi
dc.format.mimetypeapplication/pdf
dc.language.isoeng
dc.publisherCell Press
dc.relation.ispartofseriesMolecular Therapy
dc.rightsCC BY 4.0
dc.subject.otheractivins
dc.subject.othergrowth differentiation factors
dc.subject.otherischemia-reperfusion injury
dc.titleSystemic blockade of ACVR2B ligands protects myocardium from acute ischemia-reperfusion injury
dc.typearticle
dc.identifier.urnURN:NBN:fi:jyu-201903071776
dc.contributor.laitosLiikuntatieteellinen tiedekuntafi
dc.contributor.laitosFaculty of Sport and Health Sciencesen
dc.contributor.oppiaineLiikuntafysiologiafi
dc.contributor.oppiaineExercise Physiologyen
dc.type.urihttp://purl.org/eprint/type/JournalArticle
dc.date.updated2019-03-07T13:15:12Z
dc.type.coarhttp://purl.org/coar/resource_type/c_2df8fbb1
dc.description.reviewstatuspeerReviewed
dc.format.pagerange600-610
dc.relation.issn1525-0016
dc.relation.numberinseries3
dc.relation.volume27
dc.type.versionpublishedVersion
dc.rights.copyright© 2019 The Author(s).
dc.rights.accesslevelopenAccessfi
dc.relation.grantnumber275922
dc.subject.ysoiskemia
dc.subject.ysoproteiinit
dc.subject.ysosydän
dc.subject.ysolihassolut
dc.subject.ysolihakset
dc.subject.ysokasvutekijät
dc.format.contentfulltext
jyx.subject.urihttp://www.yso.fi/onto/yso/p15852
jyx.subject.urihttp://www.yso.fi/onto/yso/p4332
jyx.subject.urihttp://www.yso.fi/onto/yso/p16731
jyx.subject.urihttp://www.yso.fi/onto/yso/p25540
jyx.subject.urihttp://www.yso.fi/onto/yso/p2784
jyx.subject.urihttp://www.yso.fi/onto/yso/p2832
dc.rights.urlhttps://creativecommons.org/licenses/by/4.0/
dc.relation.doi10.1016/j.ymthe.2019.01.013
dc.relation.funderSuomen Akatemiafi
dc.relation.funderResearch Council of Finlanden
jyx.fundingprogramAkatemiatutkija, SAfi
jyx.fundingprogramAcademy Research Fellow, AoFen
jyx.fundinginformationWe thank Marja Arbelius, Sirpa Rutanen, and Kirsi Salo (University of Oulu) for excellent technical assistance. We also acknowledge Joni Degerman, Maria Arrano de Kivikko, and Nada Bechara-Hirvonen (Wihuri Research Institute, University of Helsinki) and Tuuli Nissinen (University of Jyväskylä) for excellent technical assistance. This work was supported by research funding from Academy of Finland grants 268505 (J.M.), 275922 (J.J.H.), and 297094 (R. Kerkelä); the People Programme (Marie Curie Actions) of the European Union’s Seventh Framework Programme FP7/2007-2013 under REA grant 317250 (M.R.); the Emil Aaltonen Foundation (T.K.); and the Finnish Foundation for Cardiovascular Research (J.M., L.V., T.K., Z.S., R. Kerkelä).
dc.type.okmA1


Aineistoon kuuluvat tiedostot

Thumbnail

Aineisto kuuluu seuraaviin kokoelmiin

Näytä suppeat kuvailutiedot

CC BY 4.0
Ellei muuten mainita, aineiston lisenssi on CC BY 4.0