15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The CBI‐R detects early behavioural impairment in genetic frontotemporal dementia

      research-article
      1 , 2 , 1 , 1 , 1 , 1 , 1 , 1 , 1 , 3 , 3 , 4 , 5 , 6 , 7 , 8 , 9 , 10 , 11 , 12 , 13 , 14 , 15 , 16 , 17 , 18 , 19 , 20 , 21 , 22 , 23 , 24 , 25 , 26 , 27 , 28 , 29 , 30 , 31 , 32 , 33 , 34 , 35 , 36 , 37 , 38 , 39 , 40 , 41 , 42 , 43 , 1 , , Genetic FTD Initiative (GENFI)
      Annals of Clinical and Translational Neurology
      John Wiley and Sons Inc.

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction

          Behavioural dysfunction is a key feature of genetic frontotemporal dementia (FTD) but validated clinical scales measuring behaviour are lacking at present.

          Methods

          We assessed behaviour using the revised version of the Cambridge Behavioural Inventory (CBI‐R) in 733 participants from the Genetic FTD Initiative study: 466 mutation carriers (195 C9orf72, 76 MAPT, 195 GRN) and 267 non‐mutation carriers (controls). All mutation carriers were stratified according to their global CDR plus NACC FTLD score into three groups: asymptomatic (CDR = 0), prodromal (CDR = 0.5) and symptomatic (CDR = 1+). Mixed‐effects models adjusted for age, education, sex and family clustering were used to compare between the groups. Neuroanatomical correlates of the individual domains were assessed within each genetic group.

          Results

          CBI‐R total scores were significantly higher in all CDR 1+ mutation carrier groups compared with controls [ C9orf72 mean 70.5 (standard deviation 27.8), GRN 56.2 (33.5), MAPT 62.1 (36.9)] as well as their respective CDR 0.5 groups [ C9orf72 13.5 (14.4), GRN 13.3 (13.5), MAPT 9.4 (10.4)] and CDR 0 groups [ C9orf72 6.0 (7.9), GRN 3.6 (6.0), MAPT 8.5 (13.3)]. The C9orf72 and GRN 0.5 groups scored significantly higher than the controls. The greatest impairment was seen in the Motivation domain for the C9orf72 and GRN symptomatic groups, whilst in the symptomatic MAPTgroup, the highest‐scoring domains were Stereotypic and Motor Behaviours and Memory and Orientation. Neural correlates of each CBI‐R domain largely overlapped across the different mutation carrier groups.

          Conclusions

          The CBI‐R detects early behavioural change in genetic FTD, suggesting that it could be a useful measure within future clinical trials.

          Related collections

          Most cited references37

          • Record: found
          • Abstract: found
          • Article: not found

          Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia.

          Based on the recent literature and collective experience, an international consortium developed revised guidelines for the diagnosis of behavioural variant frontotemporal dementia. The validation process retrospectively reviewed clinical records and compared the sensitivity of proposed and earlier criteria in a multi-site sample of patients with pathologically verified frontotemporal lobar degeneration. According to the revised criteria, 'possible' behavioural variant frontotemporal dementia requires three of six clinically discriminating features (disinhibition, apathy/inertia, loss of sympathy/empathy, perseverative/compulsive behaviours, hyperorality and dysexecutive neuropsychological profile). 'Probable' behavioural variant frontotemporal dementia adds functional disability and characteristic neuroimaging, while behavioural variant frontotemporal dementia 'with definite frontotemporal lobar degeneration' requires histopathological confirmation or a pathogenic mutation. Sixteen brain banks contributed cases meeting histopathological criteria for frontotemporal lobar degeneration and a clinical diagnosis of behavioural variant frontotemporal dementia, Alzheimer's disease, dementia with Lewy bodies or vascular dementia at presentation. Cases with predominant primary progressive aphasia or extra-pyramidal syndromes were excluded. In these autopsy-confirmed cases, an experienced neurologist or psychiatrist ascertained clinical features necessary for making a diagnosis according to previous and proposed criteria at presentation. Of 137 cases where features were available for both proposed and previously established criteria, 118 (86%) met 'possible' criteria, and 104 (76%) met criteria for 'probable' behavioural variant frontotemporal dementia. In contrast, 72 cases (53%) met previously established criteria for the syndrome (P < 0.001 for comparison with 'possible' and 'probable' criteria). Patients who failed to meet revised criteria were significantly older and most had atypical presentations with marked memory impairment. In conclusion, the revised criteria for behavioural variant frontotemporal dementia improve diagnostic accuracy compared with previously established criteria in a sample with known frontotemporal lobar degeneration. Greater sensitivity of the proposed criteria may reflect the optimized diagnostic features, less restrictive exclusion features and a flexible structure that accommodates different initial clinical presentations. Future studies will be needed to establish the reliability and specificity of these revised diagnostic guidelines.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS.

            Several families have been reported with autosomal-dominant frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), genetically linked to chromosome 9p21. Here, we report an expansion of a noncoding GGGGCC hexanucleotide repeat in the gene C9ORF72 that is strongly associated with disease in a large FTD/ALS kindred, previously reported to be conclusively linked to chromosome 9p. This same repeat expansion was identified in the majority of our families with a combined FTD/ALS phenotype and TDP-43-based pathology. Analysis of extended clinical series found the C9ORF72 repeat expansion to be the most common genetic abnormality in both familial FTD (11.7%) and familial ALS (23.5%). The repeat expansion leads to the loss of one alternatively spliced C9ORF72 transcript and to formation of nuclear RNA foci, suggesting multiple disease mechanisms. Our findings indicate that repeat expansion in C9ORF72 is a major cause of both FTD and ALS. Copyright © 2011 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Association of missense and 5'-splice-site mutations in tau with the inherited dementia FTDP-17.

              Thirteen families have been described with an autosomal dominantly inherited dementia named frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17), historically termed Pick's disease. Most FTDP-17 cases show neuronal and/or glial inclusions that stain positively with antibodies raised against the microtubule-associated protein Tau, although the Tau pathology varies considerably in both its quantity (or severity) and characteristics. Previous studies have mapped the FTDP-17 locus to a 2-centimorgan region on chromosome 17q21.11; the tau gene also lies within this region. We have now sequenced tau in FTDP-17 families and identified three missense mutations (G272V, P301L and R406W) and three mutations in the 5' splice site of exon 10. The splice-site mutations all destabilize a potential stem-loop structure which is probably involved in regulating the alternative splicing of exon10. This causes more frequent usage of the 5' splice site and an increased proportion of tau transcripts that include exon 10. The increase in exon 10+ messenger RNA will increase the proportion of Tau containing four microtubule-binding repeats, which is consistent with the neuropathology described in several families with FTDP-17.
                Bookmark

                Author and article information

                Contributors
                j.rohrer@ucl.ac.uk
                Journal
                Ann Clin Transl Neurol
                Ann Clin Transl Neurol
                10.1002/(ISSN)2328-9503
                ACN3
                Annals of Clinical and Translational Neurology
                John Wiley and Sons Inc. (Hoboken )
                2328-9503
                26 March 2022
                May 2022
                : 9
                : 5 ( doiID: 10.1002/acn3.v9.5 )
                : 644-658
                Affiliations
                [ 1 ] Dementia Research Centre, Department of Neurodegenerative Disease UCL Queen Square Institute of Neurology London UK
                [ 2 ] Department of Medical Statistics London School of Hygiene and Tropical Medicine London UK
                [ 3 ] Department of Neurology Erasmus Medical Centre Rotterdam Netherlands
                [ 4 ] Cognitive Disorders Unit, Department of Neurology Donostia Universitary Hospital San Sebastian Spain
                [ 5 ] Neuroscience Area Biodonostia Health Research Institute San Sebastian Gipuzkoa Spain
                [ 6 ] Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer University of Barcelona Barcelona Spain
                [ 7 ] Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine Université Laval Québec Canada
                [ 8 ] Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society Bioclinicum, Karolinska Institutet Solna Sweden
                [ 9 ] Unit for Hereditary Dementias, Theme Aging Karolinska University Hospital Solna Sweden
                [ 10 ] Sunnybrook Health Sciences Centre, Sunnybrook Research Institute University of Toronto Toronto Canada
                [ 11 ] Tanz Centre for Research in Neurodegenerative Diseases University of Toronto Toronto Ontario Canada
                [ 12 ] Department of Clinical Neurosciences University of Cambridge Cambridge UK
                [ 13 ] Neurology Unit, Department of Clinical and Experimental Sciences University of Brescia Brescia Italy
                [ 14 ] Department of Clinical Neurological Sciences University of Western Ontario London Ontario Canada
                [ 15 ] Department of Neurodegenerative Diseases, Hertie‐Institute for Clinical Brain Research and Center of Neurology University of Tübingen Tübingen Germany
                [ 16 ] Center for Neurodegenerative Diseases (DZNE Tübingen Germany
                [ 17 ] Fondazione Ca’ Granda, IRCCS Ospedale Policlinico Milan Italy
                [ 18 ] University of Milan, Centro Dino Ferrari Milan Italy
                [ 19 ] Laboratory for Cognitive Neurology, Department of Neurosciences KU Leuven Leuven Belgium
                [ 20 ] Neurology Service, University Hospitals Leuven Leuven Belgium
                [ 21 ] Leuven Brain Institute, KU Leuven Leuven Belgium
                [ 22 ] Faculty of Medicine, University of Lisbon Lisbon Portugal
                [ 23 ] Nuffield Department of Clinical Neurosciences Medical Sciences Division, University of Oxford Oxford UK
                [ 24 ] Department of Brain Sciences Imperial College London London UK
                [ 25 ] Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre University of Manchester Manchester UK
                [ 26 ] Departments of Geriatric Medicine and Nuclear Medicine University of Duisburg‐Essen Duisburg Germany
                [ 27 ] Department of Psychiatry, McGill University Health Centre McGill University Montreal QC Canada
                [ 28 ] McConnell Brain Imaging Centre, Montreal Neurological Institute McGill University Montreal QC Canada
                [ 29 ] Sorbonne Université, Paris Brain Institute – Institut du Cerveau – ICM Inserm U1127, CNRS UMR 7225, AP‐HP – Hôpital Pitié‐Salpêtrière Paris France
                [ 30 ] Département de Neurologie Centre de référence des démences rares ou précoces, IM2A, AP‐HP – Hôpital Pitié‐Salpêtrière Paris France
                [ 31 ] Département de Neurologie AP‐HP – Hôpital Pitié‐Salpêtrière Paris France
                [ 32 ] Reference Network for Rare Neurological Diseases (ERN‐RND), European Union
                [ 33 ] University Hospital of Coimbra (HUC), Neurology Service, Faculty of Medicine University of Coimbra Coimbra Portugal
                [ 34 ] Center for Neuroscience and Cell Biology, Faculty of Medicine University of Coimbra Coimbra Portugal
                [ 35 ] Univ Lille Lille France
                [ 36 ] Inserm 1172 Lille France
                [ 37 ] CHU, CNR‐MAJ, Labex Distalz LiCEND Lille Lille France
                [ 38 ] Department of Neurology Ludwig‐Maximilians Universität München Munich Germany
                [ 39 ] German Center for Neurodegenerative Diseases (DZNE) Munich Germany
                [ 40 ] Munich Cluster of Systems Neurology (SyNergy) Munich Germany
                [ 41 ] Department of Neurology University of Ulm Ulm Germany
                [ 42 ] Department of Neurofarba University of Florence Florence Italy
                [ 43 ] IRCCS Fondazione Don Carlo Gnocchi Florence Italy
                Author notes
                [*] [* ] Correspondence

                Jonathan Rohrer, Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK. Tel: +44 0845 155 5000; E‐mail: j.rohrer@ 123456ucl.ac.uk

                [†]

                List of consortium authors in Appendix.

                Author information
                https://orcid.org/0000-0001-5023-5893
                https://orcid.org/0000-0003-1814-5024
                https://orcid.org/0000-0001-7750-896X
                https://orcid.org/0000-0002-2031-490X
                https://orcid.org/0000-0001-9340-9814
                https://orcid.org/0000-0003-4461-7427
                https://orcid.org/0000-0002-2280-7273
                https://orcid.org/0000-0002-9284-5953
                https://orcid.org/0000-0002-7309-1113
                https://orcid.org/0000-0002-2508-5181
                https://orcid.org/0000-0002-6155-8417
                Article
                ACN351544 ACN3-2022-01-0023.R1
                10.1002/acn3.51544
                9082390
                f696e910-fd2d-4b2e-803f-ffc7d8c155a8
                © 2022 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 23 February 2022
                : 17 January 2022
                : 25 February 2022
                Page count
                Figures: 3, Tables: 1, Pages: 658, Words: 9130
                Funding
                Funded by: Alzheimer's Society , doi 10.13039/501100000320;
                Award ID: AS‐JF‐19a‐004‐517
                Funded by: EU Joint Programme ‐ Neurodegenerative Disease Research
                Award ID: 2019‐02248
                Award ID: 739510
                Funded by: Medical Research Council , doi 10.13039/501100000265;
                Award ID: MR/M008525/1
                Award ID: MR/M023664/1
                Funded by: The National Brain Appeal
                Award ID: RCN 290173
                Funded by: JPND GENFI‐PROX
                Award ID: 2019‐02248
                Funded by: NIHR Rare Disease Translational Research Collaboration
                Award ID: BRC149/NS/MH
                Funded by: UK Dementia Research Institute
                Funded by: NIHR UCL/H Biomedical Research Centre
                Funded by: The Wolfson Foundation
                Funded by: Brain Research UK , doi 10.13039/100013790;
                Funded by: Alzheimer’s Research UK , doi 10.13039/501100002283;
                This work was funded by Alzheimer's Society , doi 10.13039/501100000320; grant AS‐JF‐19a‐004‐517; EU Joint Programme ‐ Neurodegenerative Disease Research grants 2019‐02248 and 739510; Medical Research Council , doi 10.13039/501100000265; grants MR/M008525/1 and MR/M023664/1; The National Brain Appeal grant RCN 290173; JPND GENFI‐PROX grant 2019‐02248; NIHR Rare Disease Translational Research Collaboration grant BRC149/NS/MH; UK Dementia Research Institute; NIHR UCL/H Biomedical Research Centre; The Wolfson Foundation; Brain Research UK , doi 10.13039/100013790; ; Alzheimer’s Research UK , doi 10.13039/501100002283; .
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                May 2022
                Converter:WILEY_ML3GV2_TO_JATSPMC version:6.1.5 mode:remove_FC converted:09.05.2022

                Comments

                Comment on this article