Show simple item record

Authordc.contributor.authorCáceres Lluch, Mónica 
Authordc.contributor.authorOrtiz, Liliana 
Authordc.contributor.authorRecabarren, Tatiana 
Authordc.contributor.authorRomero, Aníbal 
Authordc.contributor.authorColombo Flores, Alicia 
Authordc.contributor.authorLeiva Salcedo, Elías 
Authordc.contributor.authorVarela Lekanda, Diego 
Authordc.contributor.authorRivas, José 
Authordc.contributor.authorSilva, Ian 
Authordc.contributor.authorMorales, Diego 
Authordc.contributor.authorCampusano, Camilo 
Authordc.contributor.authorAlmarza, Óscar 
Authordc.contributor.authorSimon, Felipe 
Authordc.contributor.authorToledo Araya, Héctor 
Authordc.contributor.authorPark, Kang-Sik 
Authordc.contributor.authorTrimmer, James S. 
Authordc.contributor.authorCerda, Óscar 
Admission datedc.date.accessioned2015-09-28T18:52:18Z
Available datedc.date.available2015-09-28T18:52:18Z
Publication datedc.date.issued2015
Cita de ítemdc.identifier.citationPLoS One Volumen: 10 Número: 6 Jun 25 2015en_US
Identifierdc.identifier.otherDOI:10.1371/journal.pone.0130540
Identifierdc.identifier.urihttps://repositorio.uchile.cl/handle/2250/133919
General notedc.descriptionArtículo de publicación ISIen_US
Abstractdc.description.abstractCellular migration and contractility are fundamental processes that are regulated by a variety of concerted mechanisms such as cytoskeleton rearrangements, focal adhesion turnover, and Ca2+ oscillations. TRPM4 is a Ca2+-activated non-selective cationic channel (Ca2+-NSCC) that conducts monovalent but not divalent cations. Here, we used a mass spectrometry-based proteomics approach to identify putative TRPM4-associated proteins. Interestingly, the largest group of these proteins has actin cytoskeleton-related functions, and among these nine are specifically annotated as focal adhesion-related proteins. Consistent with these results, we found that TRPM4 localizes to focal adhesions in cells from different cellular lineages. We show that suppression of TRPM4 in MEFs impacts turnover of focal adhesions, serum-induced Ca2+ influx, focal adhesion kinase (FAK) and Rac activities, and results in reduced cellular spreading, migration and contractile behavior. Finally, we demonstrate that the inhibition of TRPM4 activity alters cellular contractility in vivo, affecting cutaneous wound healing. Together, these findings provide the first evidence, to our knowledge, for a TRP channel specifically localized to focal adhesions, where it performs a central role in modulating cellular migration and contractility.en_US
Patrocinadordc.description.sponsorshipFondecyt 11121239 11140064 1120240 1121078 1120126 NIH R01 NS042225 Conicyt Millennium Institute on Immunology and Immunotherapy P09-016-Fen_US
Lenguagedc.language.isoenen_US
Publisherdc.publisherPublic Library Scienceen_US
Type of licensedc.rightsAtribución-NoComercial-SinDerivadas 3.0 Chile*
Link to Licensedc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/3.0/cl/*
Keywordsdc.subjectCation channel TRPM4en_US
Keywordsdc.subjectCell-migrationen_US
Keywordsdc.subjectCalcium oscillationsen_US
Keywordsdc.subjectSignaling complexesen_US
Keywordsdc.subjectMicroarray analysisen_US
Keywordsdc.subjectBeta-pixen_US
Keywordsdc.subjectActivationen_US
Keywordsdc.subjectExpressionen_US
Keywordsdc.subjectZebrafishen_US
Keywordsdc.subjectMotilityen_US
Títulodc.titleTRPM4 Is a Novel Component of the Adhesome Required for Focal Adhesion Disassembly, Migration and Contractilityen_US
Document typedc.typeArtículo de revista


Files in this item

Icon

This item appears in the following Collection(s)

Show simple item record

Atribución-NoComercial-SinDerivadas 3.0 Chile
Except where otherwise noted, this item's license is described as Atribución-NoComercial-SinDerivadas 3.0 Chile