Please use this identifier to cite or link to this item: http://hdl.handle.net/2445/194080
Full metadata record
DC FieldValueLanguage
dc.contributor.authorSirés, Anna-
dc.contributor.authorPazo-González, Mateo-
dc.contributor.authorLópez-Soriano, Joaquín-
dc.contributor.authorMéndez, Ana-
dc.contributor.authorde la Rosa, Enrique J.-
dc.contributor.authorde la Villa, Pedro-
dc.contributor.authorComella, Joan X.-
dc.contributor.authorHernández-Sánchez, Catalina-
dc.contributor.authorSolé, Montse-
dc.date.accessioned2023-02-23T18:44:34Z-
dc.date.available2023-02-23T18:44:34Z-
dc.date.issued2023-02-02-
dc.identifier.issn2073-4409-
dc.identifier.urihttp://hdl.handle.net/2445/194080-
dc.description.abstractThe short and long isoforms of FAIM (FAIM-S and FAIM-L) hold important functions in the central nervous system, and their expression levels are specifically enriched in the retina. We previously described that Faim knockout (KO) mice present structural and molecular alterations in the retina compatible with a neurodegenerative phenotype. Here, we aimed to study Faim KO retinal functions and molecular mechanisms leading to its alterations. Electroretinographic recordings showed that aged Faim KO mice present functional loss of rod photoreceptor and ganglion cells. Additionally, we found a significant delay in dark adaptation from early adult ages. This functional deficit is exacerbated by luminic stress, which also caused histopathological alterations. Interestingly, Faim KO mice present abnormal Arrestin-1 redistribution upon light reception, and we show that Arrestin-1 is ubiquitinated, a process that is abrogated by either FAIM-S or FAIM-L in vitro. Our results suggest that FAIM assists Arrestin-1 light-dependent translocation by a process that likely involves ubiquitination. In the absence of FAIM, this impairment could be the cause of dark adaptation delay and increased light sensitivity. Multiple retinal diseases are linked to deficits in photoresponse termination, and hence, investigating the role of FAIM could shed light onto the underlying mechanisms of their pathophysiology.-
dc.format.extent28 p.-
dc.format.mimetypeapplication/pdf-
dc.language.isoeng-
dc.publisherMDPI-
dc.relation.isformatofReproducció del document publicat a: https://doi.org/10.3390/cells12030487-
dc.relation.ispartofCells, 2023, vol. 12, num. 3, p. 487-
dc.relation.urihttps://doi.org/10.3390/cells12030487-
dc.rightscc-by (c) Sirés, Anna et al., 2023-
dc.rights.urihttps://creativecommons.org/licenses/by/4.0/-
dc.sourceArticles publicats en revistes (Ciències Fisiològiques)-
dc.subject.classificationRetina-
dc.subject.classificationRatolins (Animals de laboratori)-
dc.subject.classificationFotoreceptors-
dc.subject.otherRetina-
dc.subject.otherMice (Laboratory animals)-
dc.subject.otherPhotoreceptors-
dc.titleThe absence of FAIM leads to a delay in dark adaptation and hampers arrestin-1 translocation upon light reception in the retina-
dc.typeinfo:eu-repo/semantics/article-
dc.typeinfo:eu-repo/semantics/publishedVersion-
dc.identifier.idgrec730515-
dc.date.updated2023-02-23T18:44:35Z-
dc.rights.accessRightsinfo:eu-repo/semantics/openAccess-
dc.identifier.pmid36766830-
Appears in Collections:Articles publicats en revistes (Ciències Fisiològiques)
Articles publicats en revistes (Institut d'lnvestigació Biomèdica de Bellvitge (IDIBELL))

Files in This Item:
File Description SizeFormat 
730515.pdf5.78 MBAdobe PDFView/Open


This item is licensed under a Creative Commons License Creative Commons