Please use this identifier to cite or link to this item: http://hdl.handle.net/2445/124162
Full metadata record
DC FieldValueLanguage
dc.contributor.authorGabasa Ferràndez, Marta-
dc.contributor.authorDuch, Paula-
dc.contributor.authorJorba, Ignasi-
dc.contributor.authorGiménez Hidalgo, Alicia-
dc.contributor.authorLugo, Roberto-
dc.contributor.authorPavelescu, Irina-
dc.contributor.authorRodriguez Pascual, Fernando-
dc.contributor.authorMolina Molina, María-
dc.contributor.authorXaubet Mir, Antonio-
dc.contributor.authorPereda, Javier-
dc.contributor.authorAlcaraz Casademunt, Jordi-
dc.date.accessioned2018-08-28T10:42:41Z-
dc.date.available2018-08-28T10:42:41Z-
dc.date.issued2017-12-15-
dc.identifier.issn1059-1524-
dc.identifier.urihttp://hdl.handle.net/2445/124162-
dc.description.abstractThe contribution of epithelial-to-mesenchymal transition (EMT) to the profibrotic stiff microenvironment and myofibroblast accumulation in pulmonary fibrosis remains unclear. We examined EMT-competent lung epithelial cells and lung fibroblasts from control (fibrosis-free) donors or patients with idiopathic pulmonary fibrosis (IPF), which is a very aggressive fibrotic disorder. Cells were cultured on profibrotic conditions including stiff substrata and TGF-beta 1, and analyzed in terms of morphology, stiffness, and expression of EMT/myofibroblast markers and fibrillar collagens. All fibroblasts acquired a robust myofibroblast phenotype on TGF-beta 1 stimulation. Yet IPF myofibroblasts exhibited higher stiffness and expression of fibrillar collagens than control fibroblasts, concomitantly with enhanced FAK(Y397) activity. FAK inhibition was sufficient to decrease fibroblast stiffness and collagen expression, supporting that FAK(Y397) hyperactivation may underlie the aberrant mechanobiology of IPF fibroblasts. In contrast, cells undergoing EMT failed to reach the values exhibited by IPF myofibroblasts in all parameters examined. Likewise, EMT could be distinguished from nonactivated control fibroblasts, suggesting that EMT does not elicit myofibroblast precursors either. Our data suggest that EMT does not contribute directly to the myofibroblast population, and may contribute to the stiff fibrotic microenvironment through their own stiffness but not their collagen expression. Our results also support that targeting FAK(Y397) may rescue normal mechanobiology in IPF.-
dc.format.extent15 p.-
dc.format.mimetypeapplication/pdf-
dc.language.isoeng-
dc.publisherAmerican Society for Cell Biology-
dc.relation.isformatofReproducció del document publicat a: https://doi.org/10.1091/mbc.E17-01-0026-
dc.relation.ispartofMolecular Biology of the Cell, 2017, vol. 28, num. 26, p. 3741-3755-
dc.relation.urihttps://doi.org/10.1091/mbc.E17-01-0026-
dc.rightscc-by-nc-sa (c) Gabasa Ferràndez, Marta et al., 2017-
dc.rights.urihttp://creativecommons.org/licenses/by-nc-sa/3.0/es-
dc.sourceArticles publicats en revistes (Biomedicina)-
dc.subject.classificationFibrosi pulmonar-
dc.subject.classificationCèl·lules epitelials-
dc.subject.otherPulmonary fibrosis-
dc.subject.otherEpithelial cells-
dc.titleEpithelial contribution to the profibrotic stiff microenvironment and myofibroblast population in lung fibrosis-
dc.typeinfo:eu-repo/semantics/article-
dc.typeinfo:eu-repo/semantics/publishedVersion-
dc.identifier.idgrec677494-
dc.date.updated2018-08-28T10:42:41Z-
dc.rights.accessRightsinfo:eu-repo/semantics/openAccess-
dc.identifier.pmid29046395-
Appears in Collections:Articles publicats en revistes (Institut d'lnvestigació Biomèdica de Bellvitge (IDIBELL))
Articles publicats en revistes (Biomedicina)
Articles publicats en revistes (IDIBAPS: Institut d'investigacions Biomèdiques August Pi i Sunyer)

Files in This Item:
File Description SizeFormat 
677494.pdf5.84 MBAdobe PDFView/Open


This item is licensed under a Creative Commons License Creative Commons