Afficher la notice abrégée

dc.rights.licenseopenen_US
hal.structure.identifierParis-Centre de Recherche Cardiovasculaire [PARCC (UMR_S 970/ U970)]
dc.contributor.authorAL SAYED, Zeina
hal.structure.identifierParis-Centre de Recherche Cardiovasculaire [PARCC (UMR_S 970/ U970)]
dc.contributor.authorPEREIRA, Céline
hal.structure.identifierInstitut Jacques Monod [IJM (UMR_7592)]
dc.contributor.authorLE BORGNE, Rémi
hal.structure.identifierChimie biologique des membranes et ciblage thérapeutique [CBMCT - UMR 3666 / U1143]
dc.contributor.authorVIARIS DE LESEGNO, Christine
hal.structure.identifierParis-Centre de Recherche Cardiovasculaire [PARCC (UMR_S 970/ U970)]
dc.contributor.authorJOUVE, Charlène
hal.structure.identifierPlate-forme de bioimagerie ultrastructurale - Ultrastructural BioImaging Core Facility
dc.contributor.authorPÉNARD, Esthel
hal.structure.identifierPlate-forme de bioimagerie ultrastructurale - Ultrastructural BioImaging Core Facility
dc.contributor.authorMALLET, Adeline
hal.structure.identifierParis-Centre de Recherche Cardiovasculaire [PARCC (UMR_S 970/ U970)]
dc.contributor.authorMASURKAR, Nihar
hal.structure.identifierInstitut du Thorax [Nantes]
dc.contributor.authorLOUSSOUARN, Gildas
hal.structure.identifierInstitut Jacques Monod [IJM (UMR_7592)]
dc.contributor.authorVERBAVATZ, Jean-Marc
hal.structure.identifierChimie biologique des membranes et ciblage thérapeutique [CBMCT - UMR 3666 / U1143]
dc.contributor.authorLAMAZE, Christophe
hal.structure.identifierBordeaux population health [BPH]
dc.contributor.authorTREGOUET, David-Alexandre
hal.structure.identifierParis-Centre de Recherche Cardiovasculaire [PARCC (UMR_S 970/ U970)]
hal.structure.identifierCIC - HEGP [CIC 1418]
dc.contributor.authorHULOT, Jean-Sébastien
dc.date.accessioned2024-06-13T14:01:37Z
dc.date.available2024-06-13T14:01:37Z
dc.date.issued2024-04-29
dc.identifier.issn0009-7322en_US
dc.identifier.urihttps://oskar-bordeaux.fr/handle/20.500.12278/200466
dc.description.abstractEnBACKGROUND: Drug-induced QT prolongation (diLQT) is a feared side effect that could expose susceptible individuals to fatal arrhythmias. The occurrence of diLQT is primarily attributed to unintended drug interactions with cardiac ion channels, notably the hERG (human ether-a-go-go-related gene) channels that generate the delayed-rectifier potassium current (I Kr ) and thereby regulate the late repolarization phase. There is an important interindividual susceptibility to develop diLQT, which is of unknown origin but can be reproduced in patient-specific induced pluripotent stem cell–derived cardiomyocytes (iPS-CMs). We aimed to investigate the dynamics of hERG channels in response to sotalol and to identify regulators of the susceptibility to developing diLQT. METHODS: We measured electrophysiological activity and cellular distribution of hERG channels after hERG blocker treatment in iPS-CMs derived from patients with highest sensitivity (HS) or lowest sensitivity (LS) to sotalol administration in vivo (ie, on the basis of the measure of the maximal change in QT interval 3 hours after administration). Specific small interfering RNAs and CAVIN1-T2A-GFP adenovirus were used to manipulate CAVIN1 expression. RESULTS: Whereas HS and LS iPS-CMs showed similar electrophysiological characteristics at baseline, the late repolarization phase was prolonged and I Kr significantly decreased after exposure of HS iPS-CMs to low sotalol concentrations. I Kr reduction was caused by a rapid translocation of hERG channel from the membrane to the cytoskeleton-associated fractions upon sotalol application. CAVIN1 , essential for caveolae biogenesis, was 2× more highly expressed in HS iPS-CMs, and its knockdown by small interfering RNA reduced their sensitivity to sotalol. CAVIN1 overexpression in LS iPS-CMs using adenovirus showed reciprocal effects. We found that treatment with sotalol promoted translocation of the hERG channel from the plasma membrane to the cytoskeleton fractions in a process dependent on CAVIN1 (caveolae associated protein 1) expression. CAVIN1 silencing reduced the number of caveolae at the membrane and abrogated the translocation of hERG channel in sotalol-treated HS iPS-CMs. CAVIN1 also controlled cardiomyocyte responses to other hERG blockers, such as E4031, vandetanib, and clarithromycin. CONCLUSIONS: Our study identifies unbridled turnover of the potassium channel hERG as a mechanism supporting the interindividual susceptibility underlying diLQT development and demonstrates how this phenomenon is finely tuned by CAVIN1.
dc.description.sponsorshipUn rôle nouveau de la cavéoline dans le transport de la sphingomyéline à la membrane plasmiqueen_US
dc.language.isoENen_US
dc.subject.encaveolae
dc.subject.encardiac myocytes
dc.subject.ensensitivity and specificity
dc.title.enCAVIN1-Mediated hERG Dynamics: A Novel Mechanism Underlying the Interindividual Variability in Drug-Induced Long QT
dc.title.alternativeCirculationen_US
dc.typeArticle de revueen_US
dc.identifier.doi10.1161/CIRCULATIONAHA.123.063917en_US
dc.subject.halSciences du Vivant [q-bio]/Santé publique et épidémiologieen_US
dc.identifier.pubmed38682330en_US
bordeaux.journalCirculationen_US
bordeaux.hal.laboratoriesBordeaux Population Health Research Center (BPH) - UMR 1219en_US
bordeaux.institutionUniversité de Bordeauxen_US
bordeaux.institutionINSERMen_US
bordeaux.teamELEANOR_BPHen_US
bordeaux.peerReviewedouien_US
bordeaux.inpressnonen_US
bordeaux.import.sourcehal
hal.identifierpasteur-04596797
hal.version1
hal.popularnonen_US
hal.audienceInternationaleen_US
hal.exportfalse
workflow.import.sourcehal
dc.rights.ccPas de Licence CCen_US
bordeaux.COinSctx_ver=Z39.88-2004&rft_val_fmt=info:ofi/fmt:kev:mtx:journal&rft.jtitle=Circulation&rft.date=2024-04-29&rft.eissn=0009-7322&rft.issn=0009-7322&rft.au=AL%20SAYED,%20Zeina&PEREIRA,%20C%C3%A9line&LE%20BORGNE,%20R%C3%A9mi&VIARIS%20DE%20LESEGNO,%20Christine&JOUVE,%20Charl%C3%A8ne&rft.genre=article


Fichier(s) constituant ce document

Thumbnail

Ce document figure dans la(les) collection(s) suivante(s)

Afficher la notice abrégée