Please use this identifier to cite or link to this item: https://hdl.handle.net/10316/107193
DC FieldValueLanguage
dc.contributor.authorSudre, Carole H.-
dc.contributor.authorBocchetta, Martina-
dc.contributor.authorHeller, Carolin-
dc.contributor.authorConvery, Rhian-
dc.contributor.authorNeason, Mollie-
dc.contributor.authorMoore, Katrina M.-
dc.contributor.authorCash, David M.-
dc.contributor.authorThomas, David L.-
dc.contributor.authorWoollacott, Ione O. C.-
dc.contributor.authorFoiani, Martha-
dc.contributor.authorHeslegrave, Amanda-
dc.contributor.authorShafei, Rachelle-
dc.contributor.authorGreaves, Caroline-
dc.contributor.authorvan Swieten, John-
dc.contributor.authorMoreno, Fermin-
dc.contributor.authorSánchez-Valle, Raquel-
dc.contributor.authorBorroni, Barbara-
dc.contributor.authorLaforce, Robert-
dc.contributor.authorMasellis, Mario-
dc.contributor.authorTartaglia, Maria Carmela-
dc.contributor.authorGraff, Caroline-
dc.contributor.authorGalimberti, Daniela-
dc.contributor.authorRowe, James B-
dc.contributor.authorFinger, Elizabeth-
dc.contributor.authorSynofzik, Matthis-
dc.contributor.authorVandenberghe, Rik-
dc.contributor.authorde Mendonça, Alexandre-
dc.contributor.authorTagliavini, Fabrizio-
dc.contributor.authorSantana, Isabel-
dc.contributor.authorDucharme, Simon-
dc.contributor.authorButler, Chris-
dc.contributor.authorGerhard, Alex-
dc.contributor.authorLevin, Johannes-
dc.contributor.authorDanek, Adrian-
dc.contributor.authorFrisoni, Giovanni B-
dc.contributor.authorSorbi, Sandro-
dc.contributor.authorOtto, Markus-
dc.contributor.authorZetterberg, Henrik-
dc.contributor.authorOurselin, Sebastien-
dc.contributor.authorCardoso, M Jorge-
dc.contributor.authorRohrer, Jonathan D.-
dc.date.accessioned2023-06-14T09:10:33Z-
dc.date.available2023-06-14T09:10:33Z-
dc.date.issued2019-
dc.identifier.issn22131582pt
dc.identifier.urihttps://hdl.handle.net/10316/107193-
dc.description.abstractFrontotemporal dementia (FTD) is a heterogeneous group of neurodegenerative disorders with both sporadic and genetic forms. Mutations in the progranulin gene (GRN) are a common cause of genetic FTD, causing either a behavioural presentation or, less commonly, language impairment. Presence on T2-weighted images of white matter hyperintensities (WMH) has been previously shown to be more commonly associated with GRN mutations rather than other forms of FTD. The aim of the current study was to investigate the longitudinal change in WMH and the associations of WMH burden with grey matter (GM) loss, markers of neurodegeneration and cognitive function in GRN mutation carriers. 336 participants in the Genetic FTD Initiative (GENFI) study were included in the analysis: 101 presymptomatic and 32 symptomatic GRN mutation carriers, as well as 203 mutation-negative controls. 39 presymptomatic and 12 symptomatic carriers, and 73 controls also had longitudinal data available. Participants underwent MR imaging acquisition including isotropic 1 mm T1-weighted and T2-weighted sequences. WMH were automatically segmented and locally subdivided to enable a more detailed representation of the pathology distribution. Log-transformed WMH volumes were investigated in terms of their global and regional associations with imaging measures (grey matter volumes), biomarker concentrations (plasma neurofilament light chain, NfL, and glial fibrillary acidic protein, GFAP), genetic status (TMEM106B risk genotype) and cognition (tests of executive function). Analyses revealed that WMH load was higher in both symptomatic and presymptomatic groups compared with controls and this load increased over time. In particular, lesions were seen periventricularly in frontal and occipital lobes, progressing to medial layers over time. However, there was variability in the WMH load across GRN mutation carriers - in the symptomatic group 25.0% had none/mild load, 37.5% had medium and 37.5% had a severe load - a difference not fully explained by disease duration. GM atrophy was strongly associated with WMH load both globally and in separate lobes, and increased WMH burden in the frontal, periventricular and medial regions was associated with worse executive function. Furthermore, plasma NfL and to a lesser extent GFAP concentrations were seen to be associated with increased lesion burden. Lastly, the presence of the homozygous TMEM106B rs1990622 TT risk genotypic status was associated with an increased accrual of WMH per year. In summary, WMH occur in GRN mutation carriers and accumulate over time, but are variable in their severity. They are associated with increased GM atrophy and executive dysfunction. Furthermore, their presence is associated with markers of WM damage (NfL) and astrocytosis (GFAP), whilst their accrual is modified by TMEM106B genetic status. WMH load may represent a target marker for trials of disease modifying therapies in individual patients but the variability across the GRN population would prevent use of such markers as a global outcome measure across all participants in a trial.pt
dc.description.sponsorshipThis work was funded by the UK Medical Research Council, the Italian Ministry of Health, and the Canadian Institutes of Health Research as part of a Centres of Excellence in Neurodegeneration grant (CoEN015), the Wellcome/EPSRC Centre for Medical Engineering [WT 203148/Z/16/Z], IMI2 grant AMYPAD [115952], the MSCA-ITN-Demo [721820], and the Wellcome Flagship Programme in High-Dimensional Neurology. The Dementia Research Centre is supported by Alzheimer's Research UK, Brain Research Trust, and The Wolfson Foundation. This work was supported by the NIHR Queen Square Dementia Biomedical Research Unit and the NIHR UCL/H Biomedical Research Centre. CHS is supported by an Alzheimer's Society Junior Fellowship (AS-JF-17-011). JDR is supported by an MRC Clinician Scientist Fellowship (MR/ M008525/1) and has received funding from the NIHR Rare Disease Translational Research Collaboration (BRC149/NS/MH). KD is supported by an Alzheimer's Society PhD Studentship (AS-PhD-2015-005). JBR is supported by the Wellcome Trust (103838) and the NIHR Cambridge Biomedical Research Centre. MM is supported by the Canadian Institutes of Health Research and the Ontario Research Fund. RL is supported by Réseau de médecine génétique appliquée, Fonds de recherche du Québec—Santé (FRQS). FT is supported by the Italian Ministry of Health. DG is supported by the Fondazione Monzino and the Italian Ministry of Health, Ricerca Corrente. SS is supported by Cassa di Risparmio di Firenze (CRF 2013/0199) and the Ministry of Health RF- 2010-2319722. SO is supported by the Engineering and Physical Sciences Research Council (EP/H046410/1, EP/J020990/1, EP/ K005278), the Medical Research Council (MR/J01107X/1), the EU-FP7 project VPH-DARE@IT (FP7-ICT-2011-9-601055), and the National Institute for Health Research University College London Hospitals Biomedical Research Centre (NIHR BRC UCLH/UCL High Impact Initiative BW.mn.BRC10269). JvS is supported by The Netherlands Organisation for Health Research and Development Memorable grant (733050103) and Netherlands Alzheimer Foundation Memorable grant (733050103).pt
dc.language.isoengpt
dc.publisherElsevierpt
dc.relationinfo:eu-repo/grantAgreement/EC/FP7/601055/EU/VPH Dementia Research Enabled by ITpt
dc.rightsopenAccesspt
dc.rights.urihttp://creativecommons.org/licenses/by/4.0/pt
dc.subjectFrontotemporal dementiapt
dc.subjectWhite matter hyperintensitiespt
dc.subjectDementiapt
dc.subjectProgranulinpt
dc.subject.meshAdultpt
dc.subject.meshAgedpt
dc.subject.meshAsymptomatic Diseasespt
dc.subject.meshCase-Control Studiespt
dc.subject.meshDisease Progressionpt
dc.subject.meshFemalept
dc.subject.meshFrontotemporal Dementiapt
dc.subject.meshGlial Fibrillary Acidic Proteinpt
dc.subject.meshGray Matterpt
dc.subject.meshHeterozygotept
dc.subject.meshHumanspt
dc.subject.meshLongitudinal Studiespt
dc.subject.meshMagnetic Resonance Imagingpt
dc.subject.meshMalept
dc.subject.meshMembrane Proteinspt
dc.subject.meshMiddle Agedpt
dc.subject.meshMutationpt
dc.subject.meshNerve Tissue Proteinspt
dc.subject.meshNeurofilament Proteinspt
dc.subject.meshOrgan Sizept
dc.subject.meshProdromal Symptomspt
dc.subject.meshProgranulinspt
dc.subject.meshTrail Making Testpt
dc.subject.meshWhite Matterpt
dc.subject.meshExecutive Functionpt
dc.titleWhite matter hyperintensities in progranulin-associated frontotemporal dementia: A longitudinal GENFI studypt
dc.typearticle-
degois.publication.firstPage102077pt
degois.publication.titleNeuroImage: Clinicalpt
dc.peerreviewedyespt
dc.identifier.doi10.1016/j.nicl.2019.102077pt
degois.publication.volume24pt
dc.date.embargo2019-01-01*
uc.date.periodoEmbargo0pt
item.grantfulltextopen-
item.cerifentitytypePublications-
item.languageiso639-1en-
item.openairetypearticle-
item.openairecristypehttp://purl.org/coar/resource_type/c_18cf-
item.fulltextCom Texto completo-
crisitem.author.researchunitCNC - Center for Neuroscience and Cell Biology-
crisitem.author.orcid0000-0002-8114-9434-
Appears in Collections:FMUC Medicina - Artigos em Revistas Internacionais
I&D CNC - Artigos em Revistas Internacionais
Show simple item record

SCOPUSTM   
Citations

26
checked on May 6, 2024

WEB OF SCIENCETM
Citations

27
checked on May 2, 2024

Page view(s)

79
checked on May 7, 2024

Download(s)

39
checked on May 7, 2024

Google ScholarTM

Check

Altmetric

Altmetric


This item is licensed under a Creative Commons License Creative Commons