Please use this identifier to cite or link to this item: http://bura.brunel.ac.uk/handle/2438/19559
Full metadata record
DC FieldValueLanguage
dc.contributor.authorOsborn, DPS-
dc.contributor.authorRoccasecca, RM-
dc.contributor.authorMcMurray, F-
dc.contributor.authorHernandez-Hernandez, V-
dc.contributor.authorMukherjee, S-
dc.contributor.authorBarroso, I-
dc.contributor.authorStemple, D-
dc.contributor.authorCox, R-
dc.contributor.authorBeales, PL-
dc.contributor.authorChristou-Savina, S-
dc.date.accessioned2019-11-12T12:46:26Z-
dc.date.available2019-11-12T12:46:26Z-
dc.date.issued2014-02-04-
dc.identifiere87662-
dc.identifier.citationOsborn, D.P.S. et al. (2014) 'Loss of FTO Antagonises Wnt Signaling and Leads to Developmental Defects Associated with Ciliopathies', PLoS ONE, 9 (2), e87662, pp. 1 - 13. doi: 10.1371/journal.pone.0087662.en_US
dc.identifier.issn1932-6203-
dc.identifier.urihttps://bura.brunel.ac.uk/handle/2438/19559-
dc.description.abstractCommon intronic variants in the Human fat mass and obesity-associated gene (FTO) are found to be associated with an increased risk of obesity. Overexpression of FTO correlates with increased food intake and obesity, whilst loss-of-function results in lethality and severe developmental defects. Despite intense scientific discussions around the role of FTO in energy metabolism, the function of FTO during development remains undefined. Here, we show that loss of Fto leads to developmental defects such as growth retardation, craniofacial dysmorphism and aberrant neural crest cells migration in Zebrafish. We find that the important developmental pathway, Wnt, is compromised in the absence of FTO, both in vivo (zebrafish) and in vitro (Fto−/− MEFs and HEK293T). Canonical Wnt signalling is down regulated by abrogated β-Catenin translocation to the nucleus whilst non-canonical Wnt/Ca2+ pathway is activated via its key signal mediators CaMKII and PKCδ. Moreover, we demonstrate that loss of Fto results in short, absent or disorganised cilia leading to situs inversus, renal cystogenesis, neural crest cell defects and microcephaly in Zebrafish. Congruently, Fto knockout mice display aberrant tissue specific cilia. These data identify FTO as a protein-regulator of the balanced activation between canonical and non-canonical branches of the Wnt pathway. Furthermore, we present the first evidence that FTO plays a role in development and cilia formation/function.en_US
dc.description.sponsorshipThis work was supported by grants from the Wellcome Trust (PLB), EU-FP7 (SYSCILIA -241955) (DPSO), Medical Research Council G0801843/(SCS), the NIHR Great Ormond Street/Institute of Child Health Biomedical Research Centre. IB and DS acknowledge funding from the Wellcome Trust (grant WT098051), IB is supported by the United Kingdom NIHR Cambridge Biomedical Research Centre and the MRC Centre for Obesity and Related Metabolic Diseases-
dc.format.extente87662 - e87662-
dc.format.extent1 - 13-
dc.format.mediumElectronic-
dc.languageEnglish-
dc.language.isoenen_US
dc.publisherPublic Library of Science (PLoS)en_US
dc.rightsCopyright © 2014 Osborn et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/) which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.-
dc.rights.urihttps://creativecommons.org/licenses/by/4.0/-
dc.subjectcilia-
dc.subjectWnt signaling cascade-
dc.subjectembryos-
dc.subjectzebrafish-
dc.subjectphosphorylation-
dc.subjectenzyme-linked immunoassays-
dc.subjectin situ hybridization-
dc.subjectmouse models-
dc.titleLoss of FTO Antagonises Wnt Signaling and Leads to Developmental Defects Associated with Ciliopathiesen_US
dc.typeArticleen_US
dc.identifier.doihttps://doi.org/10.1371/journal.pone.0087662-
dc.relation.isPartOfPLoS ONE-
pubs.issue2-
pubs.publication-statusPublished online-
pubs.volume9-
dc.identifier.eissn1932-6203-
dc.rights.licensehttps://creativecommons.org/licenses/by/4.0/legalcode.en-
dc.rights.holderOsborn et al.-
Appears in Collections:Dept of Life Sciences Research Papers

Files in This Item:
File Description SizeFormat 
Fulltext.pdfCopyright © 2014 Osborn et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/) which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.2.7 MBAdobe PDFView/Open


This item is licensed under a Creative Commons License Creative Commons