1
|
Kakeda S, Miki Y, Kudo K, Mori H, Tokumaru AM, Abe O, Aoki S. Practical brain MRI guidelines for anti-Aβ antibody treatment in early symptomatic Alzheimer's disease. Jpn J Radiol 2025:10.1007/s11604-025-01773-x. [PMID: 40266549 DOI: 10.1007/s11604-025-01773-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE These guidelines aim to support magnetic resonance imaging (MRI) diagnosis in patients receiving anti-amyloid β (Aβ) antibody treatment without restricting treatment eligibility. MATERIALS AND METHODS These guidelines were collaboratively established by Japan Radiological Society, The Japanese Society of Neuroradiology, and Japanese Society for Magnetic Resonance in Medicine by reviewing existing literature and the results of clinical trials. RESULTS Facility standards should comply with the "Optimal Use Promotion Guidelines" of Japan, and physicians should possess comprehensive knowledge of amyloid-related imaging abnormalities (ARIA) and expertise in brain MRI interpretation. The acquisition of knowledge regarding amyloid-related imaging abnormalities, brain MRI, anti-Aβ antibody introduction, and post-treatment diagnosis are also recommended. CONCLUSION These guidelines facilitate the accurate diagnosis and effective management of ARIA; ensure the safe administration of anti-Aβ drugs; and provide a framework for MRI facilities, includes staffing requirements and the use of MRI management systems.
Collapse
Affiliation(s)
- Shingo Kakeda
- Department of Radiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, 036-8562, Japan.
| | - Yukio Miki
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Kohsuke Kudo
- Department of Diagnostic Imaging, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Harushi Mori
- Department of Radiology, School of Medicine, Jichi Medical University, Tochigi, Japan
| | - Aya M Tokumaru
- Department of Diagnostic Radiology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shigeki Aoki
- Department of Neurophysiology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Forrest SL, Kovacs GG. Current concepts and molecular pathology of neurodegenerative diseases. Pathology 2025; 57:178-190. [PMID: 39672768 DOI: 10.1016/j.pathol.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 10/27/2024] [Indexed: 12/15/2024]
Abstract
Neurodegenerative diseases are a pathologically, clinically and genetically diverse group of diseases characterised by selective dysfunction, loss of synaptic connectivity and neurodegeneration, and are associated with the deposition of misfolded proteins in neurons and/or glia. Molecular studies have highlighted the role of conformationally altered proteins in the pathogenesis of neurodegenerative diseases and have paved the way for developing disease-specific biomarkers that capture and differentiate the main type/s of protein abnormality responsible for neurodegenerative diseases, some of which are currently used in clinical practice. These proteins follow sequential patterns of anatomical involvement and disease spread in the brain and may also be detected in peripheral organs. Recent studies suggest that glia are likely to have an important role in pathological spread throughout the brain and even follow distinct progression patterns from neurons. In addition to morphological and molecular approaches to the classification of these disorders, a further new stratification level incorporates the structure of protein filaments detected by cryogenic electron microscopy. Rather than occurring in isolation, combined deposition of tau, amyloid-β, α-synuclein and TDP-43 are frequently observed in neurodegenerative diseases and in the ageing brain. These can be overlooked, and their clinicopathological relevance is difficult to interpret. This review provides an overview of disease pathogenesis and diagnostic implications, recent molecular and ultrastructural classification of neurodegenerative diseases, how to approach ageing-related and mixed pathologies, and the importance of the protein-based classification system for practising neuropathologists and clinicians. This review also informs general pathologists about the relevance of ongoing full body autopsy studies to understand the spectrum and pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Shelley L Forrest
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada; Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada; Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Gabor G Kovacs
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada; Laboratory Medicine Program and Krembil Brain Institute, University Health Network, Toronto, ON, Canada; Dementia Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia; Edmond J. Safra Program in Parkinson's Disease, Rossy PSP Centre and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology and Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Rambarack N, Fodder K, Murthy M, Toomey C, de Silva R, Heutink P, Humphrey J, Raj T, Lashley T, Bettencourt C. DNA methylation as a contributor to dysregulation of STX6 and other frontotemporal lobar degeneration genetic risk-associated loci. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634065. [PMID: 39975316 PMCID: PMC11838521 DOI: 10.1101/2025.01.21.634065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Frontotemporal Lobar Degeneration (FTLD) represents a spectrum of clinically, genetically, and pathologically heterogeneous neurodegenerative disorders characterised by progressive atrophy of the frontal and temporal lobes of the brain. The two major FTLD pathological subgroups are FTLD-TDP and FTLD-tau. While the majority of FTLD cases are sporadic, heterogeneity also exists within the familial cases, typically involving mutations in MAPT, GRN or C9orf72, which is not fully explained by known genetic mechanisms. We sought to address this gap by investigating the effect of epigenetic modifications, specifically DNA methylation variation, on genes associated with FTLD genetic risk in different FTLD subtypes. We compiled a list of genes associated with genetic risk of FTLD using text-mining databases and literature searches. Frontal cortex DNA methylation profiles were derived from three FTLD datasets containing different subgroups of FTLD-TDP and FTLD-tau: FTLD1m (N = 23) containing FTLD-TDP type A C9orf72 mutation carriers and TDP Type C sporadic cases, FTLD2m (N = 48) containing FTLD-Tau MAPT mutation carriers, FTLD-TDP Type A GRN mutation carriers, and FTLD-TDP Type B C9orf72 mutation carriers and FTLD3m (N = 163) progressive supranuclear palsy (PSP) cases, and corresponding controls. To investigate the downstream effects of DNA methylation further, we then leveraged transcriptomic and proteomic datasets for FTLD cases and controls to examine gene and protein expression levels. Our analysis revealed shared promoter region hypomethylation in STX6 across FTLD-TDP and FTLD-tau subtypes, though the largest effect size was observed in the PSP cases compared to controls (delta-beta = -32%, adjusted-p value=0.002). We also observed dysregulation of the STX6 gene and protein expression across FTLD subtypes. Additionally, we performed a detailed examination of MAPT, GRN and C9orf72 in subtypes with and without the presence of the genetic mutations and observed nominally significant differentially methylated CpGs in variable positions across the genes, often with unique patterns and downstream consequences in gene/protein expression in mutation carriers. We highlight the contribution of DNA methylation at different gene regions in regulating the expression of genes previously associated with genetic risk of FTLD, including STX6. We analysed the relationship of subtypes and presence of mutations with this epigenetic mechanism to increase our understanding of how these mechanisms interact in FTLD.
Collapse
Affiliation(s)
- Naiomi Rambarack
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Katherine Fodder
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Megha Murthy
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Christina Toomey
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Rohan de Silva
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Peter Heutink
- German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Jack Humphrey
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Towfique Raj
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Conceição Bettencourt
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
4
|
Zeng X, Hua L, Ma G, Zhao Z, Yuan Z. Dysregulated neurofluid coupling as a new noninvasive biomarker for primary progressive aphasia. Neuroimage 2024; 303:120924. [PMID: 39547457 DOI: 10.1016/j.neuroimage.2024.120924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/15/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024] Open
Abstract
Accumulation of pathological tau is one of the primary causes of Primary Progressive Aphasia (PPA). The glymphatic system is crucial for removing metabolite waste from the brain whereas impairments in glymphatic clearance in PPA are poorly understood. Thus, this study aims to investigate the role of dysregulated macroscopic cerebrospinal fluid (CSF) movement in PPA. Fifty-six PPA individuals and ninety-four healthy controls were included in our analysis after excluding those with excessive head motions during the scan. The coupling strength between blood-oxygen-level-dependent (BOLD) signals in the gray matter and CSF flow was calculated using Pearson correlation and compared between the groups. Its associations with clinical characteristics including scores from Clinical Dementia Rating (CDR), Mini-Mental State Exam, Geriatric Depression Scale and with morphological measures in the hippocampus and entorhinal cortex were examined. PPA subjects exhibited weaker global BOLD-CSF coupling compared to HCs, indicating impairments in glymphatic function in the patients (p = 0.01). In the PPA but not HC group, global BOLD-CSF coupling correlated with the CDR scores (p = 0.04) and hippocampal volume (p = 0.009). The observed decoupling between global brain activity and CSF flow and its association with symptomatology and brain structural changes in PPA converges with previous reports on the same measure in other neurodegenerative diseases. These findings support the potential role of global BOLD-CSF coupling as a noninvasive marker for glymphatic dysregulation in PPA.
Collapse
Affiliation(s)
- Xinglin Zeng
- Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Cognitive and Brain Sciences, University of Macau, Macau SAR, China
| | - Lin Hua
- Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Cognitive and Brain Sciences, University of Macau, Macau SAR, China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing, PR China
| | - Zhiying Zhao
- Centre for Cognitive and Brain Sciences, University of Macau, Macau SAR, China.
| | - Zhen Yuan
- Faculty of Health Sciences, University of Macau, Macau SAR, China; Centre for Cognitive and Brain Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
5
|
Lee JJ, Keener LC, Phan TX, Reeder JE, Wang S, Considine CM, Darby RR. Distinct Patterns of Socioemotional Dysfunction Relate to Aggressive Versus Nonaggressive Rule-breaking Antisocial Behaviors in Behavioral Variant Frontotemporal Dementia. Cogn Behav Neurol 2024; 37:194-204. [PMID: 39618111 PMCID: PMC11614378 DOI: 10.1097/wnn.0000000000000381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/25/2024] [Indexed: 12/06/2024]
Abstract
BACKGROUND Antisocial behaviors occur in up to 91% of individuals with behavioral variant frontotemporal dementia (bvFTD). Prior work has shown that antisocial behaviors can be differentiated into aggressive and nonaggressive rule-breaking behavioral subtypes. Socioemotional dysfunction is common in bvFTD and unique compared to other types of dementia. OBJECTIVE To determine whether socioemotional dysfunction relates to general antisocial behaviors in individuals with bvFTD, or whether different types of socioemotional dysfunction relate to aggressive versus rule-breaking behaviors. METHODS Informants for 28 participants with bvFTD and 21 participants with Alzheimer disease (AD) completed the Social Behavior Questionnaire (SBQ) and the Interpersonal Reactivity Index (IRI). The SBQ measures the presence and severity of 26 antisocial behaviors, including subscales for aggressive behaviors (SBQ-AGG) and nonaggressive rule-breaking behaviors (SBQ-RB). The IRI measures cognitive and emotional empathy capabilities, including subscales for Empathic Concern (IRI-EC) and Perspective-taking (IRI-PT). RESULTS As expected, participants with bvFTD had higher scores on the SBQ in total than participants with AD, as well as on the SBQ-AGG and SBQ-RB separately. Participants with bvFTD had lower scores on the IRI-EC and IRI-PT than participants with AD (P < 0.0001 for all measures). Lower scores on the IRI-PT correlated with higher scores on the SBQ-AGG-but not with higher scores on the SBQ-RB-across the combined group of participants (P = 0.007), and within participants in the bvFTD group (P = 0.01) specifically, after controlling for covariates of age, sex, dementia severity, and IRI-EC scores. Lower scores on the IRI-EC correlated with higher scores on the SBQ-AGG-but not with higher scores on the SBQ-RB-across the combined group of participants (P = 0.02) after controlling for covariates of age, sex, dementia severity, and IRI-PT scores. CONCLUSION Our results suggest that socioemotional dysfunction relates to antisocial behaviors in individuals with bvFTD, but that the mechanisms leading to aggressive and rule-breaking behaviors are differentiable, providing meaningful implications for distinct approaches to treatment and prevention.
Collapse
Affiliation(s)
- Jayden J. Lee
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lindsey C. Keener
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Tony X. Phan
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jerica E. Reeder
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Siyi Wang
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ciaran M. Considine
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - R. Ryan Darby
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
6
|
Hok-A-Hin YS, Vermunt L, Peeters CF, van der Ende EL, de Boer SC, Meeter LH, van Swieten JC, Hu WT, Lleó A, Alcolea D, Engelborghs S, Sieben A, Chen-Plotkin A, Irwin DJ, van der Flier WM, Pijnenburg YA, Teunissen CE, del Campo M. Large-scale CSF proteome profiling identifies biomarkers for accurate diagnosis of Frontotemporal Dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.19.24312100. [PMID: 39228745 PMCID: PMC11370532 DOI: 10.1101/2024.08.19.24312100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Diagnosis of Frontotemporal dementia (FTD) and the specific underlying neuropathologies (frontotemporal lobar degeneration; FTLD- Tau and FTLD-TDP) is challenging, and thus fluid biomarkers are needed to improve diagnostic accuracy. We used proximity extension assays to analyze 665 proteins in cerebrospinal fluid (CSF) samples from a multicenter cohort including patients with FTD (n = 189), Alzheimer's Disease dementia (AD; n = 232), and cognitively unimpaired individuals (n = 196). In a subset, FTLD neuropathology was determined based on phenotype or genotype (FTLD-Tau = 87 and FTLD-TDP = 68). Forty three proteins were differentially regulated in FTD compared to controls and AD, reflecting axon development, regulation of synapse assembly, and cell-cell adhesion mediator activity pathways. Classification analysis identified a 14- and 13-CSF protein panel that discriminated FTD from controls (AUC: 0.96) or AD (AUC: 0.91). Custom multiplex panels confirmed the highly accurate discrimination between FTD and controls (AUCs > 0.96) or AD (AUCs > 0.88) in three validation cohorts, including one with autopsy confirmation (AUCs > 0.90). Six proteins were differentially regulated between FTLD-TDP and FTLD-Tau, but no reproducible classification model could be generated (AUC: 0.80). Overall, this study introduces novel FTD-specific biomarker panels with potential use in diagnostic setting.
Collapse
Affiliation(s)
- Yanaika S. Hok-A-Hin
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Carel F.W. Peeters
- Mathematical & Statistical Methods group – Biometris, Wageningen University & Research, Wageningen, The Netherlands
| | - Emma L. van der Ende
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Sterre C.M. de Boer
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, Australia
| | - Lieke H. Meeter
- Alzheimer center and department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - John C. van Swieten
- Alzheimer center and department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - William T. Hu
- Department of Neurology, Center for Neurodegenerative Diseases Research, Emory University School of Medicine, Atlanta, USA
| | - Alberto Lleó
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB SANT PAU) - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Daniel Alcolea
- Department of Neurology, Institut d’Investigacions Biomèdiques Sant Pau (IIB SANT PAU) - Hospital de Sant Pau, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Vrije Universiteit Brussel, Center for Neurosciences (C4N), Neuroprotection and Neuromodulation Research Group (NEUR), Brussels, Belgium
- Universitair Ziekenhuis Brussel, Department of Neurology, Brussels, Belgium
| | - Anne Sieben
- Lab of neuropathology, Neurobiobank, Institute Born-Bunge, Antwerp University, Edegem, Belgium
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David J. Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wiesje M. van der Flier
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Yolande A.L. Pijnenburg
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Alzheimer Center and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Marta del Campo
- Neurochemistry Laboratory, Department of Laboratory Medicine, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, The Netherlands
- Barcelonaßeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San PabloCEU, CEU Universities, Madrid, Spain
| |
Collapse
|
7
|
Panahi S, Mayo J, Kennedy E, Christensen L, Kamineni S, Sagiraju HKR, Cooper T, Tate DF, Rupper R, Pugh MJ. Identifying clinical phenotypes of frontotemporal dementia in post-9/11 era veterans using natural language processing. Front Neurol 2024; 15:1270688. [PMID: 38426171 PMCID: PMC10902457 DOI: 10.3389/fneur.2024.1270688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 01/09/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Frontotemporal dementia (FTD) encompasses a clinically and pathologically diverse group of neurodegenerative disorders, yet little work has quantified the unique phenotypic clinical presentations of FTD among post-9/11 era veterans. To identify phenotypes of FTD using natural language processing (NLP) aided medical chart reviews of post-9/11 era U.S. military Veterans diagnosed with FTD in Veterans Health Administration care. Methods A medical record chart review of clinician/provider notes was conducted using a Natural Language Processing (NLP) tool, which extracted features related to cognitive dysfunction. NLP features were further organized into seven Research Domain Criteria Initiative (RDoC) domains, which were clustered to identify distinct phenotypes. Results Veterans with FTD were more likely to have notes that reflected the RDoC domains, with cognitive and positive valence domains showing the greatest difference across groups. Clustering of domains identified three symptom phenotypes agnostic to time of an individual having FTD, categorized as Low (16.4%), Moderate (69.2%), and High (14.5%) distress. Comparison across distress groups showed significant differences in physical and psychological characteristics, particularly prior history of head injury, insomnia, cardiac issues, anxiety, and alcohol misuse. The clustering result within the FTD group demonstrated a phenotype variant that exhibited a combination of language and behavioral symptoms. This phenotype presented with manifestations indicative of both language-related impairments and behavioral changes, showcasing the coexistence of features from both domains within the same individual. Discussion This study suggests FTD also presents across a continuum of severity and symptom distress, both within and across variants. The intensity of distress evident in clinical notes tends to cluster with more co-occurring conditions. This examination of phenotypic heterogeneity in clinical notes indicates that sensitivity to FTD diagnosis may be correlated to overall symptom distress, and future work incorporating NLP and phenotyping may help promote strategies for early detection of FTD.
Collapse
Affiliation(s)
- Samin Panahi
- VA Salt Lake City Health Care System, Informatics, Decision-Enhancement and Analytic Sciences Center, Salt Lake City, UT, United States
- Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Jamie Mayo
- VA Salt Lake City Health Care System, Informatics, Decision-Enhancement and Analytic Sciences Center, Salt Lake City, UT, United States
- Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Eamonn Kennedy
- VA Salt Lake City Health Care System, Informatics, Decision-Enhancement and Analytic Sciences Center, Salt Lake City, UT, United States
- Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Lee Christensen
- VA Salt Lake City Health Care System, Informatics, Decision-Enhancement and Analytic Sciences Center, Salt Lake City, UT, United States
- Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Sreekanth Kamineni
- VA Salt Lake City Health Care System, Informatics, Decision-Enhancement and Analytic Sciences Center, Salt Lake City, UT, United States
- Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | | | - Tyler Cooper
- VA Salt Lake City Health Care System, Informatics, Decision-Enhancement and Analytic Sciences Center, Salt Lake City, UT, United States
- Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - David F. Tate
- VA Salt Lake City Health Care System, Informatics, Decision-Enhancement and Analytic Sciences Center, Salt Lake City, UT, United States
- Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Randall Rupper
- VA Salt Lake City Health Care System, Geriatric Research, Education and Clinical Center, Salt Lake City, UT, United States
| | - Mary Jo Pugh
- VA Salt Lake City Health Care System, Informatics, Decision-Enhancement and Analytic Sciences Center, Salt Lake City, UT, United States
- Division of Epidemiology, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
8
|
Das S, van Engelen MPE, Goossens J, Jacobs D, Bongers B, Fieldhouse JLP, Pijnenburg YAL, Teunissen CE, Vanmechelen E, Verberk IMW. The use of synaptic biomarkers in cerebrospinal fluid to differentiate behavioral variant of frontotemporal dementia from primary psychiatric disorders and Alzheimer's disease. Alzheimers Res Ther 2024; 16:34. [PMID: 38355535 PMCID: PMC10865562 DOI: 10.1186/s13195-024-01409-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/04/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Lack of early molecular biomarkers in sporadic behavioral variants of frontotemporal dementia (bvFTD) and its clinical overlap with primary psychiatric disorders (PPD) hampers its diagnostic distinction. Synaptic dysfunction is an early feature in bvFTD and identification of specific biomarkers might improve its diagnostic accuracy. Our goal was to understand the differential diagnostic potential of cerebrospinal fluid (CSF) synaptic biomarkers in bvFTD versus PPD and their specificity towards bvFTD compared with Alzheimer's disease (AD) and controls. Additionally, we explored the association of CSF synaptic biomarkers with social cognition, cognitive performance, and disease severity in these clinical groups. METHODS Participants with probable bvFTD (n = 57), PPD (n = 71), AD (n = 60), and cognitively normal controls (n = 39) with available CSF, cognitive tests, and disease severity as frontotemporal lobar degeneration-modified clinical dementia rating scale (FTLD-CDR) were included. In a subset of bvFTD and PPD cases, Ekman 60 faces test scores for social cognition were available. CSF synaptosomal-associated protein 25 (SNAP25), neurogranin (Ng), neuronal pentraxin 2 (NPTX2), and glutamate receptor 4 (GluR4) were measured, along with neurofilament light (NfL), and compared between groups using analysis of covariance (ANCOVA) and logistic regression. Diagnostic accuracy was assessed using ROC analyses, and biomarker panels were selected using Wald's backward selection. Correlations with cognitive measures were performed using Pearson's partial correlation analysis. RESULTS NPTX2 concentrations were lower in the bvFTD group compared with PPD (p < 0.001) and controls (p = 0.003) but not compared with AD. Concentrations of SNAP25 (p < 0.001) and Ng (p < 0.001) were elevated in patients with AD versus those with bvFTD and controls. The modeled panel for differential diagnosis of bvFTD versus PPD consisted of NfL and NPTX2 (AUC = 0.96, CI: 0.93-0.99, p < 0.001). In bvFTD versus AD, the modeled panel consisted of NfL, SNAP25, Ng, and GluR4 (AUC = 0.86, CI: 0.79-0.92, p < 0.001). In bvFTD, lower NPTX2 (Pearson's r = 0.29, p = 0.036) and GluR4 (Pearson's r = 0.34, p = 0.014) concentrations were weakly associated with worse performance of total cognitive score. Lower GluR4 concentrations were also associated with worse MMSE scores (Pearson's r = 0.41, p = 0.002) as well as with worse executive functioning (Pearson's r = 0.36, p = 0.011) in bvFTD. There were no associations between synaptic markers and social cognition or disease severity in bvFTD. CONCLUSION Our findings of involvement of NTPX2 in bvFTD but not PPD contribute towards better understanding of bvFTD disease pathology.
Collapse
Affiliation(s)
- Shreyasee Das
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam, UMC location VrijeUniversiteit Amsterdam, Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- ADx NeuroSciences, Technologiepark-Zwijnaarde 6, 9052, Gent, Belgium
| | - Marie-Paule E van Engelen
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Julie Goossens
- ADx NeuroSciences, Technologiepark-Zwijnaarde 6, 9052, Gent, Belgium
| | - Dirk Jacobs
- ADx NeuroSciences, Technologiepark-Zwijnaarde 6, 9052, Gent, Belgium
| | - Bram Bongers
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam, UMC location VrijeUniversiteit Amsterdam, Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
| | - Jay L P Fieldhouse
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Yolande A L Pijnenburg
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Charlotte E Teunissen
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam, UMC location VrijeUniversiteit Amsterdam, Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | | | - Inge M W Verberk
- Department of Laboratory Medicine, Neurochemistry Laboratory, Amsterdam, UMC location VrijeUniversiteit Amsterdam, Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands.
- Neurology, Amsterdam UMC location VUmc, Alzheimer Center Amsterdam, VrijeUniversiteit Amsterdam, De Boelelaan 1118, Amsterdam, 1081 HZ, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands.
| |
Collapse
|
9
|
Matsuura S, Tatebe H, Higuchi M, Tokuda T. Validation of a newly developed immunoassay for TDP-43 in human plasma. Heliyon 2024; 10:e24672. [PMID: 38304795 PMCID: PMC10831789 DOI: 10.1016/j.heliyon.2024.e24672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/27/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
The level of TAR DNA-binding protein 43 (TDP-43) in human blood was reported to have potential for use as a specific fluid biomarker, which represents disease-specific pathologies, for TDP-43 proteinopathies, including amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), which involves the aggregation and deposition of TDP-43 in the nervous system. However, at present, no reliable immunoassay can precisely quantify TDP-43 in human plasma and detect the difference in plasma TDP-43 levels between patients with ALS and controls. We recently developed a novel ultrasensitive immunoassay to quantify TDP-43 in human plasma, and in this study, we analytically validated this assay for application as a diagnostic biomarker for TDP-43 proteinopathies. The novel TDP-43 assay was assessed for the limit of detection, lower limit of quantification, intra- and interassay variation, linearity, parallelism, and analytical spike recoveries. Additionally, 17 pilot plasma samples obtained from patients with ALS and age-matched controls were analyzed using the assay. Our novel TDP-43 assay showed sufficient analytical performance to quantify TDP-43 in human plasma, with high sensitivity (LOD and LLOQ of 0.109 and 0.759 pg/mL, respectively) and high intra- and interassay precision (%CV) below 15 %. The experimental results for spike recovery, parallelism, and dilution linearity were also acceptable. In addition, despite a small sample size, significant differences in the plasma levels of TDP-43 were found between patients with ALS and controls (ALS, 66.63 ± 20.52 pg/mL; control, 42.70 ± 23.06 pg/mL, p = 0.0330). These results support that our novel TDP-43 assay is a reliable and innovative method for the quantification of TDP-43 in human plasma and can be a potential blood-based biomarker for the diagnosis of TDP-43 proteinopathies. Further large-scale studies are warranted to validate its usefulness.
Collapse
Affiliation(s)
- Sayo Matsuura
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Harutsugu Tatebe
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| | - Takahiko Tokuda
- Department of Functional Brain Imaging, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
| |
Collapse
|
10
|
Tetter S, Arseni D, Murzin AG, Buhidma Y, Peak-Chew SY, Garringer HJ, Newell KL, Vidal R, Apostolova LG, Lashley T, Ghetti B, Ryskeldi-Falcon B. TAF15 amyloid filaments in frontotemporal lobar degeneration. Nature 2024; 625:345-351. [PMID: 38057661 PMCID: PMC10781619 DOI: 10.1038/s41586-023-06801-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 10/30/2023] [Indexed: 12/08/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) causes frontotemporal dementia (FTD), the most common form of dementia after Alzheimer's disease, and is often also associated with motor disorders1. The pathological hallmarks of FTLD are neuronal inclusions of specific, abnormally assembled proteins2. In the majority of cases the inclusions contain amyloid filament assemblies of TAR DNA-binding protein 43 (TDP-43) or tau, with distinct filament structures characterizing different FTLD subtypes3,4. The presence of amyloid filaments and their identities and structures in the remaining approximately 10% of FTLD cases are unknown but are widely believed to be composed of the protein fused in sarcoma (FUS, also known as translocated in liposarcoma). As such, these cases are commonly referred to as FTLD-FUS. Here we used cryogenic electron microscopy (cryo-EM) to determine the structures of amyloid filaments extracted from the prefrontal and temporal cortices of four individuals with FTLD-FUS. Surprisingly, we found abundant amyloid filaments of the FUS homologue TATA-binding protein-associated factor 15 (TAF15, also known as TATA-binding protein-associated factor 2N) rather than of FUS itself. The filament fold is formed from residues 7-99 in the low-complexity domain (LCD) of TAF15 and was identical between individuals. Furthermore, we found TAF15 filaments with the same fold in the motor cortex and brainstem of two of the individuals, both showing upper and lower motor neuron pathology. The formation of TAF15 amyloid filaments with a characteristic fold in FTLD establishes TAF15 proteinopathy in neurodegenerative disease. The structure of TAF15 amyloid filaments provides a basis for the development of model systems of neurodegenerative disease, as well as for the design of diagnostic and therapeutic tools targeting TAF15 proteinopathy.
Collapse
Affiliation(s)
| | - Diana Arseni
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | - Yazead Buhidma
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, London, UK
| | | | - Holly J Garringer
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kathy L Newell
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ruben Vidal
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liana G Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Tammaryn Lashley
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, London, UK
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, London, UK
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
11
|
Gentleman SM, Liu AKL. Neuropathological Assessment as an Endpoint in Clinical Trial Design. Methods Mol Biol 2024; 2785:261-270. [PMID: 38427198 DOI: 10.1007/978-1-0716-3774-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Different neurodegenerative conditions can have complex, overlapping clinical presentations that make accurate diagnosis during life very challenging. For this reason, confirmation of the clinical diagnosis still requires postmortem verification. This is particularly relevant for clinical trials of novel therapeutics where it is important to ascertain what disease- and/or pathology-modifying effects the therapeutics have had. Furthermore, it is important to confirm that patients in the trial had the correct clinical diagnosis as this will have a major bearing on the interpretation of trial results. Here we present a simple protocol for pathological assessment of neurodegenerative changes.
Collapse
Affiliation(s)
| | - Alan King Lun Liu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
12
|
de Boer SC, Riedl L, Fenoglio C, Rue I, Landin-Romero R, Matis S, Chatterton Z, Galimberti D, Halliday G, Diehl-Schmid J, Piguet O, Pijnenburg YA, Ducharme S. Rationale and Design of the "DIagnostic and Prognostic Precision Algorithm for behavioral variant Frontotemporal Dementia" (DIPPA-FTD) Study: A Study Aiming to Distinguish Early Stage Sporadic FTD from Late-Onset Primary Psychiatric Disorders. J Alzheimers Dis 2024; 97:963-973. [PMID: 38143357 PMCID: PMC10836537 DOI: 10.3233/jad-230829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 12/26/2023]
Abstract
BACKGROUND The behavioral variant of frontotemporal dementia (bvFTD) is very heterogeneous in pathology, genetics, and disease course. Unlike Alzheimer's disease, reliable biomarkers are lacking and sporadic bvFTD is often misdiagnosed as a primary psychiatric disorder (PPD) due to overlapping clinical features. Current efforts to characterize and improve diagnostics are centered on the minority of genetic cases. OBJECTIVE The multi-center study DIPPA-FTD aims to develop diagnostic and prognostic algorithms to help distinguish sporadic bvFTD from late-onset PPD in its earliest stages. METHODS The prospective DIPPA-FTD study recruits participants with late-life behavioral changes, suspect for bvFTD or late-onset PPD diagnosis with a negative family history for FTD and/or amyotrophic lateral sclerosis. Subjects are invited to participate after diagnostic screening at participating memory clinics or recruited by referrals from psychiatric departments. At baseline visit, participants undergo neurological and psychiatric examination, questionnaires, neuropsychological tests, and brain imaging. Blood is obtained to investigate biomarkers. Patients are informed about brain donation programs. Follow-up takes place 10-14 months after baseline visit where all examinations are repeated. Results from the DIPPA-FTD study will be integrated in a data-driven approach to develop diagnostic and prognostic models. CONCLUSIONS DIPPA-FTD will make an important contribution to early sporadic bvFTD identification. By recruiting subjects with ambiguous or prodromal diagnoses, our research strategy will allow the characterization of early disease stages that are not covered in current sporadic FTD research. Results will hopefully increase the ability to diagnose sporadic bvFTD in the early stage and predict progression rate, which is pivotal for patient stratification and trial design.
Collapse
Affiliation(s)
- Sterre C.M. de Boer
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, Australia
| | - Lina Riedl
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Chiara Fenoglio
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Ishana Rue
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada
| | - Ramon Landin-Romero
- Faculty of Medicine and Health, School of Health Sciences & Brain and Mind Sciences, The University of Sydney, Sydney, Australia
| | - Sophie Matis
- Faculty of Medicine and Health, School of Health Sciences & Brain and Mind Sciences, The University of Sydney, Sydney, Australia
| | - Zac Chatterton
- Brain and Mind Centre and Faculty of Medicine and Health School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Daniela Galimberti
- University of Milan, Milan, Italy
- Fondazione Ca’ Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Glenda Halliday
- School of Medical Sciences & Brain and Mind Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
- kbo-Inn-Salzach-Klinikum, Clinical Center for Psychiatry, Psychotherapy, Psychosomatic Medicine, Geriatrics and Neurology, Wasserburg/Inn, Germany
| | - Olivier Piguet
- School of Psychology and Brain & Mind Centre, The University of Sydney, Sydney, Australia
| | - Yolande A.L. Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Simon Ducharme
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada
| |
Collapse
|
13
|
van der Ende EL, In ‘t Veld SGJG, Hanskamp I, van der Lee S, Dijkstra JIR, Hok-A-Hin YS, Blujdea ER, van Swieten JC, Irwin DJ, Chen-Plotkin A, Hu WT, Lemstra AW, Pijnenburg YAL, van der Flier WM, del Campo M, Teunissen CE, Vermunt L. CSF proteomics in autosomal dominant Alzheimer's disease highlights parallels with sporadic disease. Brain 2023; 146:4495-4507. [PMID: 37348871 PMCID: PMC10629764 DOI: 10.1093/brain/awad213] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 06/24/2023] Open
Abstract
Autosomal dominant Alzheimer's disease (ADAD) offers a unique opportunity to study pathophysiological changes in a relatively young population with few comorbidities. A comprehensive investigation of proteome changes occurring in ADAD could provide valuable insights into AD-related biological mechanisms and uncover novel biomarkers and therapeutic targets. Furthermore, ADAD might serve as a model for sporadic AD, but in-depth proteome comparisons are lacking. We aimed to identify dysregulated CSF proteins in ADAD and determine the degree of overlap with sporadic AD. We measured 1472 proteins in CSF of PSEN1 or APP mutation carriers (n = 22) and age- and sex-matched controls (n = 20) from the Amsterdam Dementia Cohort using proximity extension-based immunoassays (PEA). We compared protein abundance between groups with two-sided t-tests and identified enriched biological pathways. Using the same protein panels in paired plasma samples, we investigated correlations between CSF proteins and their plasma counterparts. Finally, we compared our results with recently published PEA data from an international cohort of sporadic AD (n = 230) and non-AD dementias (n = 301). All statistical analyses were false discovery rate-corrected. We detected 66 differentially abundant CSF proteins (65 increased, 1 decreased) in ADAD compared to controls (q < 0.05). The most strongly upregulated proteins (fold change >1.8) were related to immunity (CHIT1, ITGB2, SMOC2), cytoskeletal structure (MAPT, NEFL) and tissue remodelling (TMSB10, MMP-10). Significant CSF-plasma correlations were found for the upregulated proteins SMOC2 and LILR1B. Of the 66 differentially expressed proteins, 36 had been measured previously in the sporadic dementias cohort, 34 of which (94%) were also significantly upregulated in sporadic AD, with a strong correlation between the fold changes of these proteins in both cohorts (rs = 0.730, P < 0.001). Twenty-nine of the 36 proteins (81%) were also upregulated among non-AD patients with suspected AD co-pathology. This CSF proteomics study demonstrates substantial biochemical similarities between ADAD and sporadic AD, suggesting involvement of the same biological processes. Besides known AD-related proteins, we identified several relatively novel proteins, such as TMSB10, MMP-10 and SMOC2, which have potential as novel biomarkers. With shared pathophysiological CSF changes, ADAD study findings might be translatable to sporadic AD, which could greatly expedite therapy development.
Collapse
Affiliation(s)
- Emma L van der Ende
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sjors G J G In ‘t Veld
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Iris Hanskamp
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sven van der Lee
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Janna I R Dijkstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Elena R Blujdea
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - John C van Swieten
- Alzheimer Center and Department of Neurology, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - David J Irwin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alice Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Afina W Lemstra
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Epidemiology and Data Science, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marta del Campo
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, 28003 Madrid, Spain
- Barcelonabeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Lisa Vermunt
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
14
|
Nag S, Schneider JA. Limbic-predominant age-related TDP43 encephalopathy (LATE) neuropathological change in neurodegenerative diseases. Nat Rev Neurol 2023; 19:525-541. [PMID: 37563264 PMCID: PMC10964248 DOI: 10.1038/s41582-023-00846-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 08/12/2023]
Abstract
TAR DNA-binding protein 43 (TDP43) is a focus of research in late-onset dementias. TDP43 pathology in the brain was initially identified in amyotrophic lateral sclerosis and frontotemporal lobar degeneration, and later in Alzheimer disease (AD), other neurodegenerative diseases and ageing. Limbic-predominant age-related TDP43 encephalopathy (LATE), recognized as a clinical entity in 2019, is characterized by amnestic dementia resembling AD dementia and occurring most commonly in adults over 80 years of age. Neuropathological findings in LATE, referred to as LATE neuropathological change (LATE-NC), consist of neuronal and glial cytoplasmic TDP43 localized predominantly in limbic areas with or without coexisting hippocampal sclerosis and/or AD neuropathological change and without frontotemporal lobar degeneration or amyotrophic lateral sclerosis pathology. LATE-NC is frequently associated with one or more coexisting pathologies, mainly AD neuropathological change. The focus of this Review is the pathology, genetic risk factors and nature of the cognitive impairments and dementia in pure LATE-NC and in LATE-NC associated with coexisting pathologies. As the clinical and cognitive profile of LATE is currently not easily distinguishable from AD dementia, it is important to develop biomarkers to aid in the diagnosis of this condition in the clinic. The pathogenesis of LATE-NC should be a focus of future research to form the basis for the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Sukriti Nag
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
15
|
de Boer SCM, Gossink F, Krudop W, Vijverberg E, Schouws S, Reus LM, Pijnenburg YAL, Dols A. Diagnostic Instability Over Time in the Late-Onset Frontal Lobe Syndrome: When Can We Say it's FTD? Am J Geriatr Psychiatry 2023; 31:679-690. [PMID: 37028983 DOI: 10.1016/j.jagp.2023.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023]
Abstract
OBJECTIVES Distinguishing sporadic behavioral variant of frontotemporal dementia (bvFTD) from late-onset primary psychiatric disorders (PPD) remains challenging with the lack of robust biomarkers. An early bvFTD misdiagnosis in PPD cases and vice-versa is common. Little is known about diagnostic (in)stability over longer period of time. We investigated diagnostic instability in a neuropsychiatric cohort up to 8 years after baseline visit and identified which clinical hallmarks contribute to diagnostic instability. DESIGN Diagnoses of participants of the late-onset frontal lobe (LOF) study were collected from the baseline visit (T0) and the 2-year follow-up visit (T2). Clinical outcomes were retrieved 5-8 years after baseline visit (Tfinal). Endpoint diagnoses were categorized into bvFTD, PPD and other neurological disorders (OND). We calculated the total amount of participants that switched diagnosis between T0-T2 and T2-Tfinal. Clinical records of participants that switched diagnosis were assessed. RESULTS Of the 137 patients that were included in the study, the final diagnoses at Tfinal were bvFTD 24.1% (n = 33), PPD 39.4% (n = 54), OND 33.6% (n = 46) and unknown 2.9% (n = 4). Between T0 and T2, a total of 29 (21.2%) patients switched diagnosis. Between T2 and Tfinal, 8 (5.8%) patients switched diagnosis. Prolonged follow-up identified few cases with diagnostic instability. Major contributors to diagnostic instability where a nonconverting diagnosis of possible bvFTD and a probable bvFTD diagnosis based on informant-based history and an abnormal FDG-PET scan whilst having a normal MRI. CONCLUSION Considering these lessons, a FTD diagnosis remains stable enough to conclude that 2 years is sufficient to say if a patient with late-life behavioral disorder has FTD.
Collapse
Affiliation(s)
- Sterre C M de Boer
- Alzheimer Center Amsterdam, Neurology (SCDB, WK, EV, LMR, YALP), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands; Amsterdam Neuroscience (SCDB, WK, EV, LMR, YALP), Neurodegeneration, Amsterdam, The Netherlands.
| | - Flora Gossink
- Reinier van Arkel, Geriatric and Hospital Psychiatric Centre (COZ) (FG), Jeroen Bosch Hospital, Den Bosch, The Netherlands
| | - Welmoed Krudop
- Alzheimer Center Amsterdam, Neurology (SCDB, WK, EV, LMR, YALP), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands; Amsterdam Neuroscience (SCDB, WK, EV, LMR, YALP), Neurodegeneration, Amsterdam, The Netherlands; Department of Psychology and Psychiatry, Antonius Ziekenhuis Utrecht (WK), Utrecht, The Netherlands
| | - Everard Vijverberg
- Alzheimer Center Amsterdam, Neurology (SCDB, WK, EV, LMR, YALP), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands; Amsterdam Neuroscience (SCDB, WK, EV, LMR, YALP), Neurodegeneration, Amsterdam, The Netherlands
| | - Sigfried Schouws
- Department of Old Age Psychiatry (SS), GGZ inGeest Specialized Mental Health Care, Amsterdam, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Psychiatry (SS), Amsterdam Public Health research institute, Amsterdam, The Netherlands
| | - Lianne Maria Reus
- Alzheimer Center Amsterdam, Neurology (SCDB, WK, EV, LMR, YALP), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands; Amsterdam Neuroscience (SCDB, WK, EV, LMR, YALP), Neurodegeneration, Amsterdam, The Netherlands; Center for Neurobehavioral Genetics (LMR), University of California, Los Angeles, Los Angeles, CA
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology (SCDB, WK, EV, LMR, YALP), Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands; Amsterdam Neuroscience (SCDB, WK, EV, LMR, YALP), Neurodegeneration, Amsterdam, The Netherlands
| | - Annemiek Dols
- Department of Psychiatry (AD), UMC Utrecht Brain Center, University Utrecht, Utrecht, The Netherlands
| |
Collapse
|
16
|
Murthy M, Rizzu P, Heutink P, Mill J, Lashley T, Bettencourt C. Epigenetic Age Acceleration in Frontotemporal Lobar Degeneration: A Comprehensive Analysis in the Blood and Brain. Cells 2023; 12:1922. [PMID: 37508584 PMCID: PMC10378390 DOI: 10.3390/cells12141922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/22/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Frontotemporal lobar degeneration (FTLD) includes a heterogeneous group of disorders pathologically characterized by the degeneration of the frontal and temporal lobes. In addition to major genetic contributors of FTLD such as mutations in MAPT, GRN, and C9orf72, recent work has identified several epigenetic modifications including significant differential DNA methylation in DLX1, and OTUD4 loci. As aging remains one of the major risk factors for FTLD, we investigated the presence of accelerated epigenetic aging in FTLD compared to controls. We calculated epigenetic age in both peripheral blood and brain tissues of multiple FTLD subtypes using several DNA methylation clocks, i.e., DNAmClockMulti, DNAmClockHannum, DNAmClockCortical, GrimAge, and PhenoAge, and determined age acceleration and its association with different cellular proportions and clinical traits. Significant epigenetic age acceleration was observed in the peripheral blood of both frontotemporal dementia (FTD) and progressive supranuclear palsy (PSP) patients compared to controls with DNAmClockHannum, even after accounting for confounding factors. A similar trend was observed with both DNAmClockMulti and DNAmClockCortical in post-mortem frontal cortex tissue of PSP patients and in FTLD cases harboring GRN mutations. Our findings support that increased epigenetic age acceleration in the peripheral blood could be an indicator for PSP and to a smaller extent, FTD.
Collapse
Affiliation(s)
- Megha Murthy
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK (T.L.)
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| | - Patrizia Rizzu
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Alector, Inc., South San Francisco, CA 94080, USA
| | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX4 5DW, UK
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK (T.L.)
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| | - Conceição Bettencourt
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK (T.L.)
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
| |
Collapse
|
17
|
Benussi A, Borroni B. Advances in the treatment and management of frontotemporal dementia. Expert Rev Neurother 2023; 23:621-639. [PMID: 37357688 DOI: 10.1080/14737175.2023.2228491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/19/2023] [Indexed: 06/27/2023]
Abstract
INTRODUCTION Frontotemporal dementia (FTD) is a complex neurodegenerative disorder, characterized by a wide range of pathological conditions associated with the buildup of proteins such as tau and TDP-43. With a strong hereditary component, FTD often results from genetic variants in three genes - MAPT, GRN, and C9orf72. AREAS COVERED In this review, the authors explore abnormal protein accumulation in FTD and forthcoming treatments, providing a detailed analysis of new diagnostic advancements, including innovative markers. They analyze how these discoveries have influenced therapeutic strategies, particularly disease-modifying treatments, which could potentially transform FTD management. This comprehensive exploration of FTD from its molecular underpinnings to its therapeutic prospects offers a compelling overview of the current state of FTD research. EXPERT OPINION Notable challenges in FTD management involve identifying reliable biomarkers for early diagnosis and response monitoring. Genetic forms of FTD, particularly those linked to C9orf72 and GRN, show promise, with targeted therapies resulting in substantial progress in disease-modifying strategies. The potential of neuromodulation techniques, like tDCS and rTMS, is being explored, requiring further study. Ongoing trials and multi-disciplinary care highlight the continued push toward effective FTD treatments. With increasing understanding of FTD's molecular and clinical intricacies, the hope for developing effective interventions grows.
Collapse
Affiliation(s)
- Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Neurology Unit, Department of Neurological and Vision Sciences, ASST Spedali Civili di Brescia, Brescia, Italy
| |
Collapse
|
18
|
Fodder K, Murthy M, Rizzu P, Toomey CE, Hasan R, Humphrey J, Raj T, Lunnon K, Mill J, Heutink P, Lashley T, Bettencourt C. Brain DNA methylomic analysis of frontotemporal lobar degeneration reveals OTUD4 in shared dysregulated signatures across pathological subtypes. Acta Neuropathol 2023; 146:77-95. [PMID: 37149835 PMCID: PMC10261190 DOI: 10.1007/s00401-023-02583-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/08/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) is an umbrella term describing the neuropathology of a clinically, genetically and pathologically heterogeneous group of diseases, including frontotemporal dementia (FTD) and progressive supranuclear palsy (PSP). Among the major FTLD pathological subgroups, FTLD with TDP-43 positive inclusions (FTLD-TDP) and FTLD with tau-positive inclusions (FTLD-tau) are the most common, representing about 90% of the cases. Although alterations in DNA methylation have been consistently associated with neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease, little is known for FTLD and its heterogeneous subgroups and subtypes. The main goal of this study was to investigate DNA methylation variation in FTLD-TDP and FTLD-tau. We used frontal cortex genome-wide DNA methylation profiles from three FTLD cohorts (142 FTLD cases and 92 controls), generated using the Illumina 450K or EPIC microarrays. We performed epigenome-wide association studies (EWAS) for each cohort followed by meta-analysis to identify shared differentially methylated loci across FTLD subgroups/subtypes. In addition, we used weighted gene correlation network analysis to identify co-methylation signatures associated with FTLD and other disease-related traits. Wherever possible, we also incorporated relevant gene/protein expression data. After accounting for a conservative Bonferroni multiple testing correction, the EWAS meta-analysis revealed two differentially methylated loci in FTLD, one annotated to OTUD4 (5'UTR-shore) and the other to NFATC1 (gene body-island). Of these loci, OTUD4 showed consistent upregulation of mRNA and protein expression in FTLD. In addition, in the three independent co-methylation networks, OTUD4-containing modules were enriched for EWAS meta-analysis top loci and were strongly associated with the FTLD status. These co-methylation modules were enriched for genes implicated in the ubiquitin system, RNA/stress granule formation and glutamatergic synaptic signalling. Altogether, our findings identified novel FTLD-associated loci, and support a role for DNA methylation as a mechanism involved in the dysregulation of biological processes relevant to FTLD, highlighting novel potential avenues for therapeutic development.
Collapse
Affiliation(s)
- Katherine Fodder
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Megha Murthy
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Patrizia Rizzu
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Christina E Toomey
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, London, UK
| | - Rahat Hasan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jack Humphrey
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Towfique Raj
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katie Lunnon
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Alector, Inc., South San Francisco, CA, USA
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Conceição Bettencourt
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
19
|
Jiang X, Gatt A, Lashley T. HnRNP Pathologies in Frontotemporal Lobar Degeneration. Cells 2023; 12:1633. [PMID: 37371103 DOI: 10.3390/cells12121633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Frontotemporal dementia (FTD) is the second most common form of young-onset (<65 years) dementia. Clinically, it primarily manifests as a disorder of behavioural, executive, and/or language functions. Pathologically, frontotemporal lobar degeneration (FTLD) is the predominant cause of FTD. FTLD is a proteinopathy, and the main pathological proteins identified so far are tau, TAR DNA-binding protein 43 (TDP-43), and fused in sarcoma (FUS). As TDP-43 and FUS are members of the heterogeneous ribonucleic acid protein (hnRNP) family, many studies in recent years have expanded the research on the relationship between other hnRNPs and FTLD pathology. Indeed, these studies provide evidence for an association between hnRNP abnormalities and FTLD. In particular, several studies have shown that multiple hnRNPs may exhibit nuclear depletion and cytoplasmic mislocalisation within neurons in FTLD cases. However, due to the diversity and complex association of hnRNPs, most studies are still at the stage of histological discovery of different hnRNP abnormalities in FTLD. We herein review the latest studies relating hnRNPs to FTLD. Together, these studies outline an important role of multiple hnRNPs in the pathogenesis of FTLD and suggest that future research into FTLD should include the whole spectrum of this protein family.
Collapse
Affiliation(s)
- Xinwa Jiang
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Ariana Gatt
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| |
Collapse
|
20
|
Life B, Petkau TL, Cruz GNF, Navarro-Delgado EI, Shen N, Korthauer K, Leavitt BR. FTD-associated behavioural and transcriptomic abnormalities in 'humanized' progranulin-deficient mice: A novel model for progranulin-associated FTD. Neurobiol Dis 2023; 182:106138. [PMID: 37105261 DOI: 10.1016/j.nbd.2023.106138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023] Open
Abstract
Frontotemporal dementia (FTD) is an early onset dementia characterized by neuropathology and behavioural changes. A common genetic cause of FTD is haploinsufficiency of the gene progranulin (GRN). Mouse models of progranulin deficiency have provided insight into progranulin neurobiology, but the description of phenotypes with preclinical relevance has been limited in the currently available heterozygous progranulin-null mice. The identification of robust and reproducible FTD-associated behavioural, neuropathological, and biochemical phenotypes in progranulin deficient mice is a critical step in the preclinical development of therapies for FTD. In this work, we report the generation of a novel, 'humanized' mouse model of progranulin deficiency that expresses a single, targeted copy of human GRN in the absence of mouse progranulin. We also report the in-depth, longitudinal characterization of humanized progranulin-deficient mice and heterozygous progranulin-null mice over 18 months. Our analysis yielded several novel progranulin-dependent physiological and behavioural phenotypes, including increased marble burying, open field hyperactivity, and thalamic microgliosis in both models. RNAseq analysis of cortical tissue revealed an overlapping profile of transcriptomic dysfunction. Further transcriptomic analysis offers new insights into progranulin neurobiology. In sum, we have identified several consistent phenotypes in two independent mouse models of progranulin deficiency that are expected to be useful endpoints in the development of therapies for progranulin-deficient FTD. Furthermore, the presence of the human progranulin gene in the humanized progranulin-deficient mice will expedite the development of clinically translatable gene therapy strategies.
Collapse
Affiliation(s)
- Benjamin Life
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 0B3, Canada; BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Terri L Petkau
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 0B3, Canada; BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Giuliano N F Cruz
- BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Department of Statistics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Erick I Navarro-Delgado
- BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Department of Statistics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Ning Shen
- BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Department of Statistics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Keegan Korthauer
- BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Department of Statistics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 0B3, Canada; BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Division of Neurology, Department of Medicine, University of British Columbia Hospital, Vancouver, BC V6T 2B5, Canada; Center for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
21
|
Bandopadhyay R, Gatt A, Lashley T. Advances in the Understanding of Frontotemporal Dementia. Cells 2023; 12:cells12050781. [PMID: 36899917 PMCID: PMC10000525 DOI: 10.3390/cells12050781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Frontotemporal dementia (FTD) encompasses a group of clinically, genetically and pathologically heterogeneous neurodegenerative disorders that mainly affect people under the age of 64 years [...].
Collapse
Affiliation(s)
- Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, Department of Clinical and Movement Neuroscience, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, UK
- Correspondence:
| | - Ariana Gatt
- Queen Square Brain Bank, Department of Neurodegenerative Diseases, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, UK
| | - Tammaryn Lashley
- Queen Square Brain Bank, Department of Neurodegenerative Diseases, UCL Institute of Neurology, 1 Wakefield Street, London WC1N 1PJ, UK
| |
Collapse
|
22
|
Vignaroli F, Mele A, Tondo G, De Giorgis V, Manfredi M, Comi C, Mazzini L, De Marchi F. The Need for Biomarkers in the ALS-FTD Spectrum: A Clinical Point of View on the Role of Proteomics. Proteomes 2023; 11:proteomes11010001. [PMID: 36648959 PMCID: PMC9844364 DOI: 10.3390/proteomes11010001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are severely debilitating and progressive neurodegenerative disorders. A distinctive pathological feature of several neurodegenerative diseases, including ALS and FTD, is the deposition of aberrant protein inclusions in neuronal cells, which leads to cellular dysfunction and neuronal damage and loss. Despite this, to date, the biological process behind developing these protein inclusions must be better clarified, making the development of disease-modifying treatment impossible until this is done. Proteomics is a powerful tool to characterize the expression, structure, functions, interactions, and modifications of proteins of tissue and biological fluid, including plasma, serum, and cerebrospinal fluid. This protein-profiling characterization aims to identify disease-specific protein alteration or specific pathology-based mechanisms which may be used as markers of these conditions. Our narrative review aims to highlight the need for biomarkers and the potential use of proteomics in clinical practice for ALS-FTD spectrum disorders, considering the emerging rationale in proteomics for new drug development. Certainly, new data will emerge in the near future in this regard and support clinicians in the development of personalized medicine.
Collapse
Affiliation(s)
| | - Angelica Mele
- Neurology Unit, Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Giacomo Tondo
- Department of Neurology, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy
| | - Veronica De Giorgis
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research and Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Center for Translational Research and Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Cristoforo Comi
- Department of Neurology, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Letizia Mazzini
- Neurology Unit, Maggiore della Carità Hospital, 28100 Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Fabiola De Marchi
- Neurology Unit, Maggiore della Carità Hospital, 28100 Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Correspondence: ; Tel.: +39-0321-3733962
| |
Collapse
|
23
|
Li T, Pappas C, Klinedinst B, Pollpeter A, Larsen B, Hoth N, Anton F, Wang Q, Willette AA. Associations Between Insulin-Like Growth Factor-1 and Resting-State Functional Connectivity in Cognitively Unimpaired Midlife Adults. J Alzheimers Dis 2023; 94:S309-S318. [PMID: 36710671 PMCID: PMC10473072 DOI: 10.3233/jad-220608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Insulin-like growth factor (IGF)-1 plays an important role in Alzheimer's disease (AD) pathogenesis and increases disease risk. However, prior research examining IGF-1 levels and brain neural network activity is mixed. OBJECTIVE The present study investigated the relationship between IGF-1 levels and 21 neural networks, as measured by functional magnetic resonance imaging (fMRI) in 13,235 UK Biobank participants. METHODS Linear mixed models were used to regress IGF-1 against the intrinsic functional connectivity (i.e., degree of network activity) for each neural network. Interactions between IGF-1 and AD risk factors such as Apolipoprotein E4 (APOE4) genotype, sex, AD family history, and age were also tested. RESULTS Higher IGF-1 was associated with more network activity in the right Executive Function neural network. IGF-1 interactions with APOE4 or sex implicated motor, primary/extrastriate visual, and executive function related neural networks. Neural network activity trends with increasing IGF-1 were different in different age groups. Higher IGF-1 levels relate to much more network activity in the Sensorimotor Network and Cerebellum Network in early-life participants (40-52 years old), compared with mid-life (52-59 years old) and late-life (59-70 years old) participants. CONCLUSION These findings suggest that sex and APOE4 genotype may modify the relationship between IGF-1 and brain network activities related to visual, motor, and cognitive processing. Additionally, IGF-1 may have an age-dependent effect on neural network connectivity.
Collapse
Affiliation(s)
- Tianqi Li
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
- Genetics and Genomics Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
| | - Colleen Pappas
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Brandon Klinedinst
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
- Neuroscience Interdepartmental Graduate Program Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
| | - Amy Pollpeter
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
- Bioinformatics and Computational Biology Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
| | - Brittany Larsen
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
- Neuroscience Interdepartmental Graduate Program Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
| | - Nathan Hoth
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Faith Anton
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Qian Wang
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
| | - Auriel A. Willette
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, USA
- Genetics and Genomics Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
- Neuroscience Interdepartmental Graduate Program Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
- Bioinformatics and Computational Biology Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
- Department of Neurology, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
24
|
Poos JM, Grandpierre LDM, van der Ende EL, Panman JL, Papma JM, Seelaar H, van den Berg E, van 't Klooster R, Bron E, Steketee R, Vernooij MW, Pijnenburg YAL, Rombouts SARB, van Swieten J, Jiskoot LC. Longitudinal Brain Atrophy Rates in Presymptomatic Carriers of Genetic Frontotemporal Dementia. Neurology 2022; 99:e2661-e2671. [PMID: 36288997 PMCID: PMC9757869 DOI: 10.1212/wnl.0000000000201292] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 08/10/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES It is important to identify at what age brain atrophy rates in genetic frontotemporal dementia (FTD) start to accelerate and deviate from normal aging effects to find the optimal starting point for treatment. We investigated longitudinal brain atrophy rates in the presymptomatic stage of genetic FTD using normative brain volumetry software. METHODS Presymptomatic GRN, MAPT, and C9orf72 pathogenic variant carriers underwent longitudinal volumetric T1-weighted magnetic resonance imaging of the brain as part of a prospective cohort study. Images were automatically analyzed with Quantib® ND, which consisted of volume measurements (CSF and sum of gray and white matter) of lobes, cerebellum, and hippocampus. All volumes were compared with reference centile curves based on a large population-derived sample of nondemented individuals (n = 4,951). Mixed-effects models were fitted to analyze atrophy rates of the different gene groups as a function of age. RESULTS Thirty-four GRN, 8 MAPT, and 14 C9orf72 pathogenic variant carriers were included (mean age = 52.1, standard deviation = 7.2; 66% female). The mean follow-up duration of the study was 64 ± 33 months (median = 52; range 13-108). GRN pathogenic variant carriers showed a faster decline than the reference centile curves for all brain areas, though relative volumes remained between the 5th and 75th percentiles between the ages of 45 and 70 years. In MAPT pathogenic variant carriers, frontal lobe volume was already at the 5th percentile at age 45 years and showed a further decline between the ages 50 and 60 years. Temporal lobe volume started in the 50th percentile at age 45 years but showed fastest decline over time compared with other brain structures. Frontal, temporal, parietal, and cerebellar volume already started below the 5th percentile compared with the reference centile curves at age 45 years for C9orf72 pathogenic variant carriers, but there was minimal decline over time until the age of 60 years. DISCUSSION We provide evidence for longitudinal brain atrophy in the presymptomatic stage of genetic FTD. The affected brain areas and the age after which atrophy rates start to accelerate and diverge from normal aging slopes differed between gene groups. These results highlight the value of normative volumetry software for disease tracking and staging biomarkers in genetic FTD. These techniques could help in identifying the optimal time window for starting treatment and monitoring treatment response.
Collapse
Affiliation(s)
- Jackie M Poos
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Leonie D M Grandpierre
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Emma L van der Ende
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Jessica L Panman
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Janne M Papma
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Harro Seelaar
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Esther van den Berg
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Ronald van 't Klooster
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Esther Bron
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Rebecca Steketee
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Meike W Vernooij
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Yolande A L Pijnenburg
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Serge A R B Rombouts
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - John van Swieten
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Lize C Jiskoot
- From the Department of Neurology and Alzheimer Center Erasmus MC (Jackie M. Poos, L.D.M.G., E.L.E., J.L.P., Janne M. Papma, H.S., Esther van den Berg, J.S., L.C.J.), Erasmus MC University Medical Center; Quantib B.V. (R.K.), Rotterdam; Departments of Radiology and Nuclear Medicine (Esther Bron, R.S., M.W.V.) and Epidemiology (M.W.V.), Erasmus MC University Medical Center Rotterdam; Department of Neurology (Y.A.L.P.), Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center; Department of Radiology (S.A.R.B.R.), Leiden University Medical Center; Institute of Psychology (S.A.R.B.R.) and Leiden Institute for Brain and Cognition (S.A.R.B.R.), Leiden University, The Netherlands; and Dementia Research Centre (L.C.J.), Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom.
| |
Collapse
|
25
|
van der Ende EL, Heller C, Sogorb-Esteve A, Swift IJ, McFall D, Peakman G, Bouzigues A, Poos JM, Jiskoot LC, Panman JL, Papma JM, Meeter LH, Dopper EGP, Bocchetta M, Todd E, Cash D, Graff C, Synofzik M, Moreno F, Finger E, Sanchez-Valle R, Vandenberghe R, Laforce R, Masellis M, Tartaglia MC, Rowe JB, Butler C, Ducharme S, Gerhard A, Danek A, Levin J, Pijnenburg YAL, Otto M, Borroni B, Tagliavini F, de Mendonça A, Santana I, Galimberti D, Sorbi S, Zetterberg H, Huang E, van Swieten JC, Rohrer JD, Seelaar H. Elevated CSF and plasma complement proteins in genetic frontotemporal dementia: results from the GENFI study. J Neuroinflammation 2022; 19:217. [PMID: 36064709 PMCID: PMC9446850 DOI: 10.1186/s12974-022-02573-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 08/19/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroinflammation is emerging as an important pathological process in frontotemporal dementia (FTD), but biomarkers are lacking. We aimed to determine the value of complement proteins, which are key components of innate immunity, as biomarkers in cerebrospinal fluid (CSF) and plasma of presymptomatic and symptomatic genetic FTD mutation carriers. METHODS We measured the complement proteins C1q and C3b in CSF by ELISAs in 224 presymptomatic and symptomatic GRN, C9orf72 or MAPT mutation carriers and non-carriers participating in the Genetic Frontotemporal Dementia Initiative (GENFI), a multicentre cohort study. Next, we used multiplex immunoassays to measure a panel of 14 complement proteins in plasma of 431 GENFI participants. We correlated complement protein levels with corresponding clinical and neuroimaging data, neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP). RESULTS CSF C1q and C3b, as well as plasma C2 and C3, were elevated in symptomatic mutation carriers compared to presymptomatic carriers and non-carriers. In genetic subgroup analyses, these differences remained statistically significant for C9orf72 mutation carriers. In presymptomatic carriers, several complement proteins correlated negatively with grey matter volume of FTD-related regions and positively with NfL and GFAP. In symptomatic carriers, correlations were additionally observed with disease duration and with Mini Mental State Examination and Clinical Dementia Rating scale® plus NACC Frontotemporal lobar degeneration sum of boxes scores. CONCLUSIONS Elevated levels of CSF C1q and C3b, as well as plasma C2 and C3, demonstrate the presence of complement activation in the symptomatic stage of genetic FTD. Intriguingly, correlations with several disease measures in presymptomatic carriers suggest that complement protein levels might increase before symptom onset. Although the overlap between groups precludes their use as diagnostic markers, further research is needed to determine their potential to monitor dysregulation of the complement system in FTD.
Collapse
Affiliation(s)
- Emma L. van der Ende
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Carolin Heller
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Aitana Sogorb-Esteve
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Imogen J. Swift
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, London, UK
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - David McFall
- Department of Pathology, University of California San Francisco, San Francisco, USA
| | - Georgia Peakman
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Arabella Bouzigues
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Jackie M. Poos
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Lize C. Jiskoot
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jessica L. Panman
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Janne M. Papma
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Lieke H. Meeter
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Elise G. P. Dopper
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Emily Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - David Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Hospital Universitario Donostia, San Sebastian, Gipuzkoa Spain
- Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Gipuzkoa Spain
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, ON Canada
| | - Raquel Sanchez-Valle
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Louvain, Belgium
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département Des Sciences Neurologiques, CHU de Québec, Université Laval, Québec, Canada
| | | | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON Canada
| | - James B. Rowe
- Cambridge University Centre for Frontotemporal Dementia, University of Cambridge, Cambridge, UK
| | - Chris Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Montreal Neurological Institute and McGill University Health Centre, McGill University, Montreal, Québec Canada
| | - Alexander Gerhard
- Department of Nuclear Medicine and Geriatric Medicine, University Hospital Essen, Essen, Germany
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Adrian Danek
- Neurologische Klinik Und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Johannes Levin
- Neurologische Klinik Und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Yolande A. L. Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Markus Otto
- Department of Neurology, Universität Ulm, Ulm, Germany
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | | | | - Isabel Santana
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Daniela Galimberti
- Fondazione IRCCS, Ospedale Maggiore Policlinico, Neurodegenerative Diseases Unit, Milan, Italy
- University of Milan, Centro Dino Ferrari, Milan, Italy
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, Florence, Italy
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, UCL Queen Square Institute of Neurology, London, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Eric Huang
- Department of Pathology, University of California San Francisco, San Francisco, USA
| | - John C. van Swieten
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jonathan D. Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Harro Seelaar
- Alzheimer Center Rotterdam and Department of Neurology, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
26
|
Ryan B, O’Mara Baker A, Ilse C, Brickell KL, Kersten HM, Williams JM, Addis DR, Tippett LJ, Curtis MA. The New Zealand Genetic Frontotemporal Dementia Study (FTDGeNZ): a longitudinal study of pre-symptomatic biomarkers. J R Soc N Z 2022; 53:511-531. [PMID: 39439966 PMCID: PMC11459842 DOI: 10.1080/03036758.2022.2101483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 07/08/2022] [Indexed: 10/16/2022]
Abstract
The New Zealand Genetic Frontotemporal Dementia Study (FTDGeNZ) is an emerging longitudinal study of a large New Zealand pedigree with genetic frontotemporal dementia (FTD). Natural history studies of genetic FTD cohorts provide a unique opportunity to identify biomarkers of pre-symptomatic dementia, as carriers can be identified and studied decades before expected symptom onset. FTDGeNZ was established in 2016 with the aim of identifying the earliest pre-symptomatic biomarkers of FTD, in collaboration with international multi-centre cohorts. We enrolled 25 participants from a single family between April 2016 and August 2018. Participants were genotyped to determine whether they were pre-symptomatic carriers of the mutation (MAPT IVS 10 + 16 C > T), or non-carrier controls. Participants have undergone clinical assessments including neuropsychological and mood assessment; olfactory testing; assessment of social cognition; and blood collection for analyses of microRNA and protein fluid biomarkers annually. We have also performed structural and functional MRI of the brain and assessment of autobiographical memory biennially, and retinal imaging at baseline. Here, we describe the full study protocol and the baseline demographic and clinical characteristics of the FTDGeNZ cohort, and we highlight the latest findings in the field.
Collapse
Affiliation(s)
- Brigid Ryan
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Ashleigh O’Mara Baker
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Christina Ilse
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Kiri L. Brickell
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Hannah M. Kersten
- School of Optometry and Vision Science, University of Auckland, Auckland, New Zealand
| | - Joanna M. Williams
- Department of Anatomy, University of Otago, Dunedin, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Donna Rose Addis
- School of Psychology, University of Auckland, Auckland, New Zealand
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
- Department of Psychology, University of Toronto, Toronto, Canada
| | - Lynette J. Tippett
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- School of Psychology, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| | - Maurice A. Curtis
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Brain Research New Zealand, Rangahau Roro Aotearoa, New Zealand
| |
Collapse
|
27
|
Poos JM, MacDougall A, van den Berg E, Jiskoot LC, Papma JM, van der Ende EL, Seelaar H, Russell LL, Peakman G, Convery R, Pijnenburg YAL, Moreno F, Sanchez-Valle R, Borroni B, Laforce R, Doré MC, Masellis M, Tartaglia MC, Graff C, Galimberti D, Rowe JB, Finger E, Synofzik M, Vandenberghe R, Mendonça A, Tiraboschi P, Santana I, Ducharme S, Butler C, Gerhard A, Levin J, Danek A, Otto M, Le Ber I, Pasquier F, van Swieten J, Rohrer JD. Longitudinal Cognitive Changes in Genetic Frontotemporal Dementia Within the GENFI Cohort. Neurology 2022; 99:e281-e295. [PMID: 35483895 PMCID: PMC9302936 DOI: 10.1212/wnl.0000000000200384] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/25/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Disease-modifying therapeutic trials for genetic frontotemporal dementia (FTD) are underway, but sensitive cognitive outcome measures are lacking. The aim of this study was to identify such cognitive tests in early stage FTD by investigating cognitive decline in a large cohort of genetic FTD pathogenic variant carriers and by investigating whether gene-specific differences are moderated by disease stage (asymptomatic, prodromal, and symptomatic). METHODS C9orf72, GRN, and MAPT pathogenic variant carriers as well as controls underwent a yearly neuropsychological assessment covering 8 cognitive domains as part of the Genetic FTD Initiative, a prospective multicenter cohort study. Pathogenic variant carriers were stratified according to disease stage using the global Clinical Dementia Rating (CDR) plus National Alzheimer's Coordinating Center (NACC) FTLD score (0, 0.5, or ≥1). Linear mixed-effects models were used to investigate differences between genetic groups and disease stages as well as the 3-way interaction between time, genetic group, and disease stage. RESULTS A total of 207 C9orf72, 206 GRN, and 86 MAPT pathogenic variant carriers and 255 controls were included. C9orf72 pathogenic variant carriers performed lower on attention, executive function, and verbal fluency from CDR plus NACC FTLD 0 onwards, with relatively minimal decline over time regardless of the CDR plus NACC FTLD score (i.e., disease progression). The cognitive profile in MAPT pathogenic variant carriers was characterized by lower memory performance at CDR plus NACC FTLD 0.5, with decline over time in language from the CDR plus NACC FTLD 0.5 stage onwards, and executive dysfunction rapidly developing at CDR plus NACC FTLD ≥1. GRN pathogenic variant carriers declined on verbal fluency and visuoconstruction in the CDR plus NACC FTLD 0.5 stage, with progressive decline in other cognitive domains starting at CDR plus NACC FTLD ≥1. DISCUSSION We confirmed cognitive decline in the asymptomatic and prodromal stage of genetic FTD. Specifically, tests for attention, executive function, language, and memory showed clear differences between genetic groups and controls at baseline, but the speed of change over time differed depending on genetic group and disease stage. This confirms the value of neuropsychological assessment in tracking clinical onset and progression and could inform clinical trials in selecting sensitive end points for measuring treatment effects as well as characterizing the best time window for starting treatment.
Collapse
Affiliation(s)
- Jackie M Poos
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Amy MacDougall
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Esther van den Berg
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Lize C Jiskoot
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Janne M Papma
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Emma L van der Ende
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Harro Seelaar
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Lucy L Russell
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Georgia Peakman
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Rhian Convery
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Yolande A L Pijnenburg
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Fermin Moreno
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Raquel Sanchez-Valle
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Barbara Borroni
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Robert Laforce
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Marie-Claire Doré
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Mario Masellis
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Maria Carmela Tartaglia
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Caroline Graff
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Daniela Galimberti
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - James B Rowe
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Elizabeth Finger
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Matthis Synofzik
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Rik Vandenberghe
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Alexandre Mendonça
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Pietro Tiraboschi
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Isabel Santana
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Simon Ducharme
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Christopher Butler
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Alexander Gerhard
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Johannes Levin
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Adrian Danek
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Markus Otto
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Isabelle Le Ber
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Florence Pasquier
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - John van Swieten
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - Jonathan D Rohrer
- From the Department of Neurology (J.M. Poos, E.v.d.B., L.C.J., J.M. Papma, E.L.v.d.E., H.S., J.v.S.), Erasmus MC University Medical Center, Rotterdam, the Netherlands; Dementia Research Centre (J.M. Poos, L.C.J., L.L.R., G.P., R.C., J.D.R.), Department of Neurodegenerative Disease, UCL Institute of Neurology; Department of Medical Statistics (A.M.), London School of Hygiene and Tropical Medicine, UK; Department of Neurology (Y.A.L.P.), Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, the Netherlands; Cognitive Disorders Unit (F.M.), Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I Sunyer, University of Barcelona, Spain; Centre for Neurodegenerative Disorders (B.B.), Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Italy; Clinique Interdisciplinaire de Mémoire (R.L., M.-C.D.), Département des Sciences Neurologiques, Université Laval, Québec; Sunnybrook Health Sciences Centre (M.M.), Sunnybrook Research Institute and Tanz Centre for Research in Neurodegenerative Diseases (M.C.T.), University of Toronto, Ontario, Canada; Department of Geriatric Medicine (C.G.), Karolinska University Hospital-Huddinge, Stockholm, Sweden; Centro Dino Ferrari (D.G.), University of Milan; Fondazione IRCCS Ca' Granda (D.G.), Ospedale Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; Department of Clinical Neurosciences (J.B.R.), University of Cambridge, UK; Department of Clinical Neurological Sciences (E.F.), University of Western Ontario, London, Canada; Department of Neurodegenerative Diseases (M.S.), Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen; German Center for Neurodegenerative Diseases (DZNE) (M.S.), Tübingen, Germany; Laboratory for Cognitive Neurology (R.V.), Department of Neurosciences, KU Leuven, Belgium; Faculty of Medicine (A.M.), University of Lisbon, Portugal; Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta (P.T.), Milan, Italy; Faculty of Medicine (I.S.), University of Coimbra, Portugal; Department of Psychiatry (S.D.), McGill University Health Centre, McGill University, Montreal, Québec, Canada; Department of Clinical Neurology (C.B.), University of Oxford; Divison of Neuroscience & Experimental Psychology (A.G.), Faculty of Medicine, Biology and Health, University of Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine (A.G.), Essen University Hospital, Germany; Department of Neurology (J.L., A.D.), Ludwig-Maximilians-University, Munich; German Center for Neurodegenerative Diseases (DZNE) (J.L.), Munich; Munich Cluster for Systems Neurology (SyNergy) (J.L.); Department of Neurology (M.O.), University of Ulm, Germany; Sorbonne Université (I.L.B.), Paris Brain Institute-Institut du Cerveau-ICM, Inserm U1127, CNRS UMR 7225, AP-HP-Hôpital Pitié-Salpêtrière; Centre de Référence des Démences Rares ou Précoces (I.L.B.), IM2A, Département de Neurologie, AP-HP-Hôpital Pitié-Salpêtrière; Univ Lille (F.P.); Inserm 1172 (F.P.); and CHU (F.P.), CNR-MAJ, Labex Distalz, LiCEND, Lille, France.
| | | |
Collapse
|
28
|
Hok-A-Hin YS, Dijkstra AA, Rábano A, Hoozemans JJ, Castillo L, Seelaar H, van Swieten JC, Pijnenburg YAL, Teunissen CE, Del Campo M. Apolipoprotein L1 is increased in frontotemporal lobar degeneration post-mortem brain but not in ante-mortem cerebrospinal fluid. Neurobiol Dis 2022; 172:105813. [PMID: 35820647 DOI: 10.1016/j.nbd.2022.105813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022] Open
Abstract
AIMS Frontotemporal Dementia (FTD) is caused by frontal-temporal lobar degeneration (FTLD), characterized mainly by brain protein aggregates of tau (FTLD-Tau) or TDP-43 (FTLD-TDP). The clinicopathological heterogeneity makes ante-mortem diagnosis of these pathological subtypes challenging. Our proteomics study showed increased Apolipoprotein L1 (APOL1) levels in CSF from FTD patients, which was prominently expressed in FTLD-Tau. We aimed to understand APOL1 expression in FTLD post-mortem brain tissue and to validate its potential as a CSF biomarker for FTD and its pathological subtypes. METHODS APOL1 levels were analyzed in the frontal cortex of FTLD (including FTLD-Tau and FTLD-TDP) and non-demented controls by immunohistochemistry (FTLD total = 18 (12 FTLD-Tau and 6 FTLD-TDP); controls = 9), western blot (WB), and a novel prototype ELISA (FTLD total = 44 (21 FTLD-Tau and 23 FTLD-TDP); controls = 9). The association of APOL1 immunoreactivity with phosphorylated Tau (pTau) and TDP-43 (pTDP-43) immunoreactivity was assessed. CSF APOL1 was analyzed in confirmed FTD patients (n = 27, including 12 FTLD-Tau and 15 FTLD-TDP) and controls (n = 15) using the same ELISA. RESULTS APOL1 levels were significantly increased in FTLD post-mortem tissue compared to controls as measured by immunohistochemistry, WB, and ELISA. However, no differences between the pathological subtypes were observed. APOL1 immunoreactivity was present in neuronal and glial cells but did not co-localize with pTau or pTDP-43. CSF APOL1 levels were comparable between FTD patients and controls and between pathological subtypes. CONCLUSION APOL1 is upregulated in FTLD pathology irrespective of the subtypes, indicating a role of this novel protein in FTD pathophysiology. The APOL1 levels detected in brain tissue were not mirrored in the CSF, limiting its potential as a specific FTD biofluid-based biomarker using our current immunoassay.
Collapse
Affiliation(s)
- Yanaika S Hok-A-Hin
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands.
| | - Anke A Dijkstra
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, the Netherlands
| | - Alberto Rábano
- CIEN Tissue Bank, Alzheimer's Centre Reina Sofía-CIEN Foundation, Madrid, Spain
| | - Jeroen J Hoozemans
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, the Netherlands
| | - Lucía Castillo
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands
| | - Harro Seelaar
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yolande A L Pijnenburg
- Alzheimer Centre Amsterdam and Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam UMC, the Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands
| | - Marta Del Campo
- Neurochemistry Laboratory, Clinical Chemistry Department, Amsterdam Neuroscience, VU University Medical Centers, the Netherlands; Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo- CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
29
|
van der Ende EL, Bron EE, Poos JM, Jiskoot LC, Panman JL, Papma JM, Meeter LH, Dopper EGP, Wilke C, Synofzik M, Heller C, Swift IJ, Sogorb-Esteve A, Bouzigues A, Borroni B, Sanchez-Valle R, Moreno F, Graff C, Laforce R, Galimberti D, Masellis M, Tartaglia MC, Finger E, Vandenberghe R, Rowe JB, de Mendonça A, Tagliavini F, Santana I, Ducharme S, Butler CR, Gerhard A, Levin J, Danek A, Otto M, Pijnenburg YAL, Sorbi S, Zetterberg H, Niessen WJ, Rohrer JD, Klein S, van Swieten JC, Venkatraghavan V, Seelaar H. A data-driven disease progression model of fluid biomarkers in genetic frontotemporal dementia. Brain 2022; 145:1805-1817. [PMID: 34633446 PMCID: PMC9166533 DOI: 10.1093/brain/awab382] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/22/2021] [Accepted: 09/09/2021] [Indexed: 11/17/2022] Open
Abstract
Several CSF and blood biomarkers for genetic frontotemporal dementia have been proposed, including those reflecting neuroaxonal loss (neurofilament light chain and phosphorylated neurofilament heavy chain), synapse dysfunction [neuronal pentraxin 2 (NPTX2)], astrogliosis (glial fibrillary acidic protein) and complement activation (C1q, C3b). Determining the sequence in which biomarkers become abnormal over the course of disease could facilitate disease staging and help identify mutation carriers with prodromal or early-stage frontotemporal dementia, which is especially important as pharmaceutical trials emerge. We aimed to model the sequence of biomarker abnormalities in presymptomatic and symptomatic genetic frontotemporal dementia using cross-sectional data from the Genetic Frontotemporal dementia Initiative (GENFI), a longitudinal cohort study. Two-hundred and seventy-five presymptomatic and 127 symptomatic carriers of mutations in GRN, C9orf72 or MAPT, as well as 247 non-carriers, were selected from the GENFI cohort based on availability of one or more of the aforementioned biomarkers. Nine presymptomatic carriers developed symptoms within 18 months of sample collection ('converters'). Sequences of biomarker abnormalities were modelled for the entire group using discriminative event-based modelling (DEBM) and for each genetic subgroup using co-initialized DEBM. These models estimate probabilistic biomarker abnormalities in a data-driven way and do not rely on previous diagnostic information or biomarker cut-off points. Using cross-validation, subjects were subsequently assigned a disease stage based on their position along the disease progression timeline. CSF NPTX2 was the first biomarker to become abnormal, followed by blood and CSF neurofilament light chain, blood phosphorylated neurofilament heavy chain, blood glial fibrillary acidic protein and finally CSF C3b and C1q. Biomarker orderings did not differ significantly between genetic subgroups, but more uncertainty was noted in the C9orf72 and MAPT groups than for GRN. Estimated disease stages could distinguish symptomatic from presymptomatic carriers and non-carriers with areas under the curve of 0.84 (95% confidence interval 0.80-0.89) and 0.90 (0.86-0.94) respectively. The areas under the curve to distinguish converters from non-converting presymptomatic carriers was 0.85 (0.75-0.95). Our data-driven model of genetic frontotemporal dementia revealed that NPTX2 and neurofilament light chain are the earliest to change among the selected biomarkers. Further research should investigate their utility as candidate selection tools for pharmaceutical trials. The model's ability to accurately estimate individual disease stages could improve patient stratification and track the efficacy of therapeutic interventions.
Collapse
Affiliation(s)
- Emma L van der Ende
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Esther E Bron
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Jackie M Poos
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Lize C Jiskoot
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Jessica L Panman
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Janne M Papma
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Lieke H Meeter
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Elise G P Dopper
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Carlo Wilke
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, 72076 Tübingen, Germany
| | - Carolin Heller
- UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Imogen J Swift
- UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Aitana Sogorb-Esteve
- UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Arabella Bouzigues
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, 25121 Brescia, Italy
| | - Raquel Sanchez-Valle
- Alzheimer’s Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, 20014 Gipuzkoa, Spain
- Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Gipuzkoa, Spain
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, 17176 Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, 17176 Solna, Sweden
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, Université Laval, G1Z 1J4 Québec, Canada
| | - Daniela Galimberti
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy
- Neurodegenerative Diseases Unit, Fondazione IRCCS, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, ON M4N 3M5 Toronto, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, M5S 1A8 Toronto, Canada
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, ON N6A 3K7 London, Ontario, Canada
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - James B Rowe
- Cambridge University Centre for Frontotemporal Dementia, University of Cambridge, CB2 0SZ Cambridge, UK
| | | | | | - Isabel Santana
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Montreal Neurological Institute and McGill University Health Centre, McGill University, 3801 Montreal, Québec, Canada
| | - Christopher R Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, OX3 9DU Oxford, UK
- Department of Brain Sciences, Imperial College London, SW7 2AZ London, UK
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, M20 3LJ Manchester, UK
- Department of Nuclear Medicine and Geriatric Medicine, University Hospital Essen, 45 147 Essen, Germany
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
- German Center for Neurodegenerative Diseases, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, 89081 Ulm, Germany
| | - Yolande A L Pijnenburg
- Department of Neurology, Alzheimer Center, Location VU University Medical Center Amsterdam Neuroscience, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, 50139 Florence, Italy
| | - Henrik Zetterberg
- UK Dementia Research Institute at University College London, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, 405 30 Mölndal, Sweden
| | - Wiro J Niessen
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Jonathan D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, WC1N 3BG London, UK
| | - Stefan Klein
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Vikram Venkatraghavan
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Harro Seelaar
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
30
|
Foster PH, Russell LL, Peakman G, Convery RS, Bouzigues A, Greaves CV, Bocchetta M, Cash DM, van Swieten JC, Jiskoot LC, Moreno F, Sanchez-Valle R, Laforce R, Graff C, Masellis M, Tartaglia C, Rowe JB, Borroni B, Finger E, Synofzik M, Galimberti D, Vandenberghe R, de Mendonça A, Butler CR, Gerhard A, Ducharme S, Le Ber I, Tagliavini F, Santana I, Pasquier F, Levin J, Danek A, Otto M, Sorbi S, Rohrer JD. Examining empathy deficits across familial forms of frontotemporal dementia within the GENFI cohort. Cortex 2022; 150:12-28. [PMID: 35325762 PMCID: PMC9067453 DOI: 10.1016/j.cortex.2022.01.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 12/14/2021] [Accepted: 01/09/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Reduced empathy is a common symptom in frontotemporal dementia (FTD). Although empathy deficits have been extensively researched in sporadic cases, few studies have explored the differences in familial forms of FTD. METHODS Empathy was examined using a modified version of the Interpersonal Reactivity Index (mIRI) in 676 participants from the Genetic FTD Initiative: 216 mutation-negative controls, 192 C9orf72 expansion carriers, 193 GRN mutation carriers and 75 MAPT mutation carriers. Using global scores from the CDR® plus NACC FTLD, mutation carriers were divided into three groups, asymptomatic (0), very mildly symptomatic/prodromal (.5), or fully symptomatic (1 or more). The mIRI Total score, as well as the subscores of Empathic Concern (EC) and Perspective Taking (PT) were assessed. Linear regression models with bootstrapping were used to assess empathy ratings across genetic groups, as well as across phenotypes in the symptomatic carriers. Neural correlates of empathy deficits were examined using a voxel-based morphometry (VBM) analysis. RESULTS All fully symptomatic groups scored lower on the mIRI Total, EC, and PT when compared to controls and their asymptomatic or prodromal counterparts (all p < .001). Prodromal C9orf72 expansion carriers also scored significantly lower than controls on the mIRI Total score (p = .046). In the phenotype analysis, all groups (behavioural variant FTD, primary progressive aphasia and FTD with amyotrophic lateral sclerosis) scored significantly lower than controls (all p < .007). VBM revealed an overlapping neural correlate of the mIRI Total score across genetic groups in the orbitofrontal lobe but with additional involvement in the temporal lobe, insula and basal ganglia in both the GRN and MAPT groups, and uniquely more posterior regions such as the parietal lobe and thalamus in the GRN group, and medial temporal structures in the MAPT group. CONCLUSIONS Significant empathy deficits present in genetic FTD, particularly in symptomatic individuals and those with a bvFTD phenotype, while prodromal deficits are only seen using the mIRI in C9orf72 expansion carriers.
Collapse
Affiliation(s)
- Phoebe H Foster
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Lucy L Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Georgia Peakman
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Rhian S Convery
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Arabella Bouzigues
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Caroline V Greaves
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK; Centre for Medical Image Computing, University College London, London, UK
| | | | - Lize C Jiskoot
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia Universitary Hospital, San Sebastian, Spain; Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Gipuzkoa, Spain
| | - Raquel Sanchez-Valle
- 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
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, QC, Canada
| | - Caroline Graff
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Bioclinicum, Karolinska Institutet, Solna, Sweden; Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - James B Rowe
- University of Cambridge Department of Clinical Neurosciences, and University of Cambridge Hospitals NHS Trust, University of Cambridge, UK
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, ON, Canada
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany; Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Daniela Galimberti
- Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy; University of Milan, Centro Dino Ferrari, Milan, Italy
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium; Neurology Service, University Hospitals Leuven, Leuven, Belgium; Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | | | - Chris R Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, UK; Department of Brain Sciences, Imperial College London, UK
| | - Alex Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK; Departments of Geriatric Medicine and Nuclear Medicine, University of Duisburg-Essen, Germany
| | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, Québec, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Québec, Canada
| | - Isabelle Le Ber
- Sorbonne Université, Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France; Reference Network for Rare Neurological Diseases (ERN-RND)
| | | | - Isabel Santana
- University Hospital of Coimbra (HUC), Neurology Service, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Florence Pasquier
- Univ Lille, France; Inserm 1172, Lille, France; CHU, CNR-MAJ, Labex Distalz, LiCEND Lille, France
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians Universität München, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians Universität München, Munich, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Germany
| | - Sandro Sorbi
- Department of Neurofarba, University of Florence, Italy; IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
31
|
Amyloid fibrils in FTLD-TDP are composed of TMEM106B and not TDP-43. Nature 2022; 605:304-309. [PMID: 35344984 PMCID: PMC9844993 DOI: 10.1038/s41586-022-04670-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/22/2022] [Indexed: 01/19/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) is the third most common neurodegenerative condition after Alzheimer's and Parkinson's diseases1. FTLD typically presents in 45 to 64 year olds with behavioural changes or progressive decline of language skills2. The subtype FTLD-TDP is characterized by certain clinical symptoms and pathological neuronal inclusions with TAR DNA-binding protein (TDP-43) immunoreactivity3. Here we extracted amyloid fibrils from brains of four patients representing four of the five FTLD-TDP subclasses, and determined their structures by cryo-electron microscopy. Unexpectedly, all amyloid fibrils examined were composed of a 135-residue carboxy-terminal fragment of transmembrane protein 106B (TMEM106B), a lysosomal membrane protein previously implicated as a genetic risk factor for FTLD-TDP4. In addition to TMEM106B fibrils, we detected abundant non-fibrillar aggregated TDP-43 by immunogold labelling. Our observations confirm that FTLD-TDP is associated with amyloid fibrils, and that the fibrils are formed by TMEM106B rather than TDP-43.
Collapse
|
32
|
Poos JM, van den Berg E, Papma JM, van der Tholen FC, Seelaar H, Donker Kaat L, Kievit JA, Tibben A, van Swieten JC, Jiskoot LC. Mindfulness-Based Stress Reduction in Pre-symptomatic Genetic Frontotemporal Dementia: A Pilot Study. Front Psychiatry 2022; 13:864391. [PMID: 35573340 PMCID: PMC9091907 DOI: 10.3389/fpsyt.2022.864391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/28/2022] [Indexed: 11/16/2022] Open
Abstract
Pre-symptomatic frontotemporal dementia (FTD) mutation carriers and first-degree family members that are 50% at-risk for FTD may experience symptoms of anxiety and depression as a result of the ambiguity of when or if symptoms of the disease will manifest. We conducted a pilot study to investigate the use of an online mindfulness-based stress reduction (MBSR) course to reduce symptoms of anxiety and depression in presymptomatic frontotemporal dementia (FTD) mutation carriers and individuals 50% at-risk. Seven known mutation carriers and six individuals 50% at-risk completed a standardized 8-week MBSR course, and filled out pre- and post and two-month follow-up questionnaires. The primary outcome measure was the Hospital Anxiety and Depression Scale (HADS). Measures of psychological distress (SCL-90-R), coping style (UCL), quality of life (SF-36) and mindfulness skills (FFMQ) were administered as secondary outcome. Group effects were analyzed with repeated measures ANOVA or Friedman's test, and the individual reliability change index (RCI) was calculated per participant for each outcome measure. Semi-quantitative data included an evaluation and process measure post-intervention. Significant decline was found on the HADS-A post-intervention and after 2 months (p = 0.01), with 54% and 62% of participants demonstrating a clinically significant RCI, respectively. On the HADS-D, significant decline was found 2 months post-intervention (p = 0.04), which was driven by 23% of participants whom had a clinically significant RCI. Additional changes were found between baseline and post-intervention on the seeking distraction and reassuring thoughts subscales of the UCL, the depression and interpersonal sensitivity subscales of the SCL, the observe subscale of the FFMQ, and on physical role limitations of the SF-36 (all p < 0.05). The process evaluation form indicated that the course was found beneficial by participants, and that they applied it in a wide range of everyday situations. This exploratory pilot study indicates the feasibility of MBSR in reducing anxiety and depression in presymptomatic FTD mutation carriers and 50% at-risk individuals. A randomized controlled trial is necessary to replicate these results.
Collapse
Affiliation(s)
- Jackie M Poos
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Esther van den Berg
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Janne M Papma
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Fleur C van der Tholen
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Harro Seelaar
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Laura Donker Kaat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - J Anneke Kievit
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Aad Tibben
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - John C van Swieten
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Lize C Jiskoot
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Dementia Research Centre, University College London, London, United Kingdom
| |
Collapse
|
33
|
Cervantes González A, Irwin DJ, Alcolea D, McMillan CT, Chen-Plotkin A, Wolk D, Sirisi S, Dols-Icardo O, Querol-Vilaseca M, Illán-Gala I, Santos-Santos MA, Fortea J, Lee EB, Trojanowski JQ, Grossman M, Lleó A, Belbin O. Multimarker synaptic protein cerebrospinal fluid panels reflect TDP-43 pathology and cognitive performance in a pathological cohort of frontotemporal lobar degeneration. Mol Neurodegener 2022; 17:29. [PMID: 35395770 PMCID: PMC8991834 DOI: 10.1186/s13024-022-00534-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 03/30/2022] [Indexed: 12/13/2022] Open
Abstract
Background Synapse degeneration is an early event in pathological frontotemporal lobar degeneration (FTLD). Consequently, a surrogate marker of synapse loss could be used to monitor early pathologic changes in patients with underlying FTLD. The aim of this study was to evaluate the relationship of antemortem cerebrospinal fluid (CSF) levels of 8 synaptic proteins with postmortem global tau and TDP-43 burden and cognitive performance and to assess their diagnostic capacity in a neuropathological FTLD cohort. Methods We included patients with a neuropathological confirmation of FTLD-Tau (n = 24, mean age-at-CSF 67 years ± 11), FTLD-TDP (n = 25, 66 years ± 9) or AD (n = 25, 73 years ± 6) as well as cognitively normal controls (n = 35, 69 years ± 7) from the Penn FTD Center and ADRC. We used a semi-quantitative measure of tau and TDP-43 inclusions to quantify pathological burden across 16 brain regions. Statistical methods included Spearman rank correlations, one-way analysis of covariance, ordinal regression, step-wise multiple linear regression and receiver-operating characteristic curves. Result CSF calsyntenin-1 and neurexin-2a were correlated in all patient groups (rs = .55 to .88). In FTLD-TDP, we observed low antemortem CSF levels of calsyntenin-1 and neurexin-2a compared to AD (.72-fold, p = .001, .77-fold, p = .04, respectively) and controls (.80-fold, p = .02, .78-fold, p = .02, respectively), which were inversely associated with post-mortem global TDP-43 burden (regression r2 = .56, p = .007 and r2 = .57, p = .006, respectively). A multimarker panel including calsyntenin-1 was associated with TDP-43 burden (r2 = .69, p = .003) and MMSE score (r2 = .19, p = .03) in FTLD. A second multimarker synaptic panel, also including calsyntenin-1, was associated with MMSE score in FTLD-tau (r2 = .49, p = .04) and improved diagnostic performance to discriminate FTLD-Tau and FTLD-TDP neuropathologic subtypes (AUC = .83). Conclusion These synaptic panels have potential in the differential diagnosis of FTLD neuropathologic subtypes and as surrogate markers of cognitive performance in future clinical trials targeting TDP-43 or tau. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-022-00534-y.
Collapse
Affiliation(s)
- Alba Cervantes González
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain
| | - David J Irwin
- Penn FTD Center, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Alcolea
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain
| | - Corey T McMillan
- Penn FTD Center, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Alice Chen-Plotkin
- Penn Alzheimer's Disease Research Center, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - David Wolk
- Penn Alzheimer's Disease Research Center, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sònia Sirisi
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain
| | - Oriol Dols-Icardo
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain
| | - Marta Querol-Vilaseca
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain
| | - Ignacio Illán-Gala
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain
| | - Miguel Angel Santos-Santos
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain
| | - Juan Fortea
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain
| | - Edward B Lee
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Murray Grossman
- Penn FTD Center, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Alberto Lleó
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain.,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain
| | - Olivia Belbin
- Hospital de La Santa Creu I Sant Pau, Universitat Autonoma de Barcelona, Barcelona, Spain. .,Centre of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain. .,Memory Unit and Biomedical Research Institute, IIB Sant Pau, c/Sant Quintí 77, 08041, Barcelona, Spain.
| |
Collapse
|
34
|
McCarthy J, Borroni B, Sanchez‐Valle R, Moreno F, Laforce R, Graff C, Synofzik M, Galimberti D, Rowe JB, Masellis M, Tartaglia MC, Finger E, Vandenberghe R, de Mendonça A, Tagliavini F, Santana I, Butler C, Gerhard A, Danek A, Levin J, Otto M, Frisoni G, Ghidoni R, Sorbi S, Jiskoot LC, Seelaar H, van Swieten JC, Rohrer JD, Iturria‐Medina Y, Ducharme S. Data-driven staging of genetic frontotemporal dementia using multi-modal MRI. Hum Brain Mapp 2022; 43:1821-1835. [PMID: 35118777 PMCID: PMC8933323 DOI: 10.1002/hbm.25727] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 12/01/2022] Open
Abstract
Frontotemporal dementia in genetic forms is highly heterogeneous and begins many years to prior symptom onset, complicating disease understanding and treatment development. Unifying methods to stage the disease during both the presymptomatic and symptomatic phases are needed for the development of clinical trials outcomes. Here we used the contrastive trajectory inference (cTI), an unsupervised machine learning algorithm that analyzes temporal patterns in high‐dimensional large‐scale population datasets to obtain individual scores of disease stage. We used cross‐sectional MRI data (gray matter density, T1/T2 ratio as a proxy for myelin content, resting‐state functional amplitude, gray matter fractional anisotropy, and mean diffusivity) from 383 gene carriers (269 presymptomatic and 115 symptomatic) and a control group of 253 noncarriers in the Genetic Frontotemporal Dementia Initiative. We compared the cTI‐obtained disease scores to the estimated years to onset (age—mean age of onset in relatives), clinical, and neuropsychological test scores. The cTI based disease scores were correlated with all clinical and neuropsychological tests (measuring behavioral symptoms, attention, memory, language, and executive functions), with the highest contribution coming from mean diffusivity. Mean cTI scores were higher in the presymptomatic carriers than controls, indicating that the method may capture subtle pre‐dementia cerebral changes, although this change was not replicated in a subset of subjects with complete data. This study provides a proof of concept that cTI can identify data‐driven disease stages in a heterogeneous sample combining different mutations and disease stages of genetic FTD using only MRI metrics.
Collapse
Affiliation(s)
- Jillian McCarthy
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Raquel Sanchez‐Valle
- Alzheimer's disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacións Biomèdiques August Pi I SunyerUniversity of BarcelonaBarcelonaSpain
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of NeurologyDonostia University HospitalSan SebastianGipuzkoaSpain
- Neuroscience AreaBiodonostia Health Research InstituteSan SebastianGipuzkoaSpain
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de MédecineUniversité LavalQuebecQuebecCanada
| | - Caroline Graff
- Department of Geriatric MedicineKarolinska University Hospital‐HuddingeStockholmSweden
- Unit for Hereditary DementiasTheme Aging, Karolinska University HospitalSolnaSweden
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie‐Institute for Clinical Brain Research and Center of NeurologyUniversity of TübingenTübingenGermany
- Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Daniela Galimberti
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoNeurodegenerative Diseases UnitMilanItaly
- Department of Biomedical, Surgical, and Dental SciencesUniversity of Milan, Dino Ferrari CenterMilanItaly
| | - James B. Rowe
- University of Cambridge Department of Clinical NeurosciencesCambridge University Hospitals NHS Trust, and RC Cognition and Brain Sciences UnitCambridgeUK
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research InstituteUniversity of TorontoTorontoOntarioCanada
| | - Maria Carmela Tartaglia
- Toronto Western HospitalTanz Centre for Research in Neurodegenerative DiseaseTorontoOntarioCanada
| | - Elizabeth Finger
- Department of Clinical Neurological SciencesUniversity of Western OntarioLondonOntarioCanada
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of NeurosciencesKU LeuvenLeuvenBelgium
- Neurology ServiceUniversity Hospitals LeuvenBelgium
- Leuven Brain InstituteKU LeuvenLeuvenBelgium
| | | | - Fabrizio Tagliavini
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologico Carlo BestaMilanItaly
| | - Isabel Santana
- Neurology DepartmentCentro Hospitalar e Universitário de CoimbraCoimbraPortugal
- Center for Neuroscience and Cell Biology, Faculty of MedicineUniversity of CoimbraCoimbraPortugal
| | - Chris Butler
- Department of Clinical NeurologyUniversity of OxfordOxfordUK
- Department of Brain SciencesImperial College LondonUK
| | - Alex Gerhard
- Division of Neuroscience & Experimental Psychology, Faculty of Medicine, Biology, and HealthUniversity of ManchesterManchesterUK
- Departments of Geriatric Medicine and Nuclear MedicineEssen University HospitalEssenGermany
| | - Adrian Danek
- Ludwig‐Maximilians‐Universität MünchenMunichGermany
| | - Johannes Levin
- Ludwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster of Systems Neurology (SyNergy)MunichGermany
| | - Markus Otto
- Department of NeurologyUniversity Hospital UlmUlmGermany
| | - Giovanni Frisoni
- LANE ‐ Laboratory of Alzheimer's Neuroimaging and EpidemiologyIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
- Memory Clinic and LANVIE‐Laboratory of Neuroimaging of AgingUniversity Hospitals and University of GenevaGenevaSwitzerland
| | - Roberta Ghidoni
- Molecular Markers LaboratoryIRCCS Istituto Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Sandro Sorbi
- Department of NeurofarbaUniversity of FlorenceItaly
- IRCCS Fondazione Don Carlo GnocchiFlorenceItaly
| | - Lize C. Jiskoot
- Department of NeurologyErasmus University Medical CentreRotterdamNetherlands
| | - Harro Seelaar
- Department of NeurologyErasmus University Medical CentreRotterdamNetherlands
| | - John C. van Swieten
- Department of NeurologyErasmus University Medical CentreRotterdamNetherlands
| | - Jonathan D. Rohrer
- Department of Neurodegenerative Disease, Dementia Research CentreUCL Institute of NeurologyLondonUK
| | - Yasser Iturria‐Medina
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Neurology and Neurosurgery Department, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Ludmer Centre for Neuroinformatics & Mental HealthMcGill UniversityMontrealCanada
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Montreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University Institute, Department of PsychiatryMcGill UniversityMontrealCanada
| | | |
Collapse
|
35
|
Poos JM, Moore KM, Nicholas J, Russell LL, Peakman G, Convery RS, Jiskoot LC, van der Ende E, van den Berg E, Papma JM, Seelaar H, Pijnenburg YAL, Moreno F, Sanchez-Valle R, Borroni B, Laforce R, Masellis M, Tartaglia C, Graff C, Galimberti D, Rowe JB, Finger E, Synofzik M, Vandenberghe R, de Mendonça A, Tiraboschi P, Santana I, Ducharme S, Butler C, Gerhard A, Levin J, Danek A, Otto M, Le Ber I, Pasquier F, van Swieten JC, Rohrer JD. Cognitive composites for genetic frontotemporal dementia: GENFI-Cog. Alzheimers Res Ther 2022; 14:10. [PMID: 35045872 PMCID: PMC8772227 DOI: 10.1186/s13195-022-00958-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/28/2021] [Indexed: 11/18/2022]
Abstract
Background Clinical endpoints for upcoming therapeutic trials in frontotemporal dementia (FTD) are increasingly urgent. Cognitive composite scores are often used as endpoints but are lacking in genetic FTD. We aimed to create cognitive composite scores for genetic frontotemporal dementia (FTD) as well as recommendations for recruitment and duration in clinical trial design. Methods A standardized neuropsychological test battery covering six cognitive domains was completed by 69 C9orf72, 41 GRN, and 28 MAPT mutation carriers with CDR® plus NACC-FTLD ≥ 0.5 and 275 controls. Logistic regression was used to identify the combination of tests that distinguished best between each mutation carrier group and controls. The composite scores were calculated from the weighted averages of test scores in the models based on the regression coefficients. Sample size estimates were calculated for individual cognitive tests and composites in a theoretical trial aimed at preventing progression from a prodromal stage (CDR® plus NACC-FTLD 0.5) to a fully symptomatic stage (CDR® plus NACC-FTLD ≥ 1). Time-to-event analysis was performed to determine how quickly mutation carriers progressed from CDR® plus NACC-FTLD = 0.5 to ≥ 1 (and therefore how long a trial would need to be). Results The results from the logistic regression analyses resulted in different composite scores for each mutation carrier group (i.e. C9orf72, GRN, and MAPT). The estimated sample size to detect a treatment effect was lower for composite scores than for most individual tests. A Kaplan-Meier curve showed that after 3 years, ~ 50% of individuals had converted from CDR® plus NACC-FTLD 0.5 to ≥ 1, which means that the estimated effect size needs to be halved in sample size calculations as only half of the mutation carriers would be expected to progress from CDR® plus NACC FTLD 0.5 to ≥ 1 without treatment over that time period. Discussion We created gene-specific cognitive composite scores for C9orf72, GRN, and MAPT mutation carriers, which resulted in substantially lower estimated sample sizes to detect a treatment effect than the individual cognitive tests. The GENFI-Cog composites have potential as cognitive endpoints for upcoming clinical trials. The results from this study provide recommendations for estimating sample size and trial duration. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-00958-0.
Collapse
Affiliation(s)
- Jackie M Poos
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Dementia Research Centre, Department of Neurodegenerative Disease, National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, 8-11 Queen Square, Box 16, London, WC1N 3BG, UK
| | - Katrina M Moore
- Dementia Research Centre, Department of Neurodegenerative Disease, National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, 8-11 Queen Square, Box 16, London, WC1N 3BG, UK
| | - Jennifer Nicholas
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Lucy L Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, 8-11 Queen Square, Box 16, London, WC1N 3BG, UK
| | - Georgia Peakman
- Dementia Research Centre, Department of Neurodegenerative Disease, National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, 8-11 Queen Square, Box 16, London, WC1N 3BG, UK
| | - Rhian S Convery
- Dementia Research Centre, Department of Neurodegenerative Disease, National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, 8-11 Queen Square, Box 16, London, WC1N 3BG, UK
| | - Lize C Jiskoot
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Dementia Research Centre, Department of Neurodegenerative Disease, National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, 8-11 Queen Square, Box 16, London, WC1N 3BG, UK
| | - Emma van der Ende
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Esther van den Berg
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Janne M Papma
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Harro Seelaar
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Yolande A L Pijnenburg
- Department of Neurology, Alzheimer Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa, Spain
| | - Raquel Sanchez-Valle
- 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
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, Université Laval, Québec, Canada
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Caroline Graff
- Department of Geriatric Medicine, Karolinska University Hospital-Huddinge, Stockholm, Sweden
| | - Daniela Galimberti
- University of Milan, Centro Dino Ferrari, Milan, Italy.,Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Policlinico, Milan, Italy
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | | | - Pietro Tiraboschi
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Neurologica Carlo Besta, Milan, Italy
| | - Isabel Santana
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, Québec, Canada
| | - Chris Butler
- Department of Clinical Neurology, University of Oxford, Oxford, UK
| | - Alexander Gerhard
- Faculty of Medical and Human Sciences, Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Johannes Levin
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Isabel Le Ber
- Paris Brain Institute - Institut du Cerveau - ICM, Inserm U1127, CNRS UMR 7225, AP-HP - Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, France.,Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France.,Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Florence Pasquier
- University of Lille, Lille, France.,Inserm 1172, Lille, France.,CHU, CNR-MAJ, Labex Distalz, LiCEND, Lille, France
| | - John C van Swieten
- Department of Neurology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, 8-11 Queen Square, Box 16, London, WC1N 3BG, UK.
| | | |
Collapse
|
36
|
Poos JM, van den Berg E, Visch-Brink E, Eikelboom WS, Franzen S, van Hemmen J, Pijnenburg YAL, Satoer D, Dopper EGP, van Swieten JC, Papma JM, Seelaar H, Jiskoot LC. Exploring Abstract Semantic Associations in the Frontotemporal Dementia Spectrum in a Dutch Population. Arch Clin Neuropsychol 2022; 37:104-116. [PMID: 33856423 PMCID: PMC8763124 DOI: 10.1093/arclin/acab022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To investigate the differential ability of the "Test Relaties Abstracte Concepten" (TRACE), a Dutch test for abstract semantic knowledge, in frontotemporal dementia (FTD). METHODS The TRACE was administered in patients with behavioral variant FTD (bvFTD; n = 16), nonfluent variant (nfvPPA; n = 10), logopenic variant (lvPPA; n = 10), and semantic variant primary progressive aphasia (svPPA; n = 9), and controls (n = 59). We examined group differences, performed correlational analyses with other neuropsychological tests and investigated discriminative ability. We compared the TRACE with a semantic association test for concrete stimuli (SAT). RESULTS All patient groups, except nfvPPA, performed worse on the TRACE than controls (p < .01). svPPA patients performed worse than the other patient groups (p < .05). The TRACE discriminated well between patient groups, except nfvPPA, versus controls (all p < .01) and between svPPA versus other patient groups with high sensitivity (75-100%) and specificity (86%-92%). In bvFTD and nfvPPA the TRACE correlated with language tests (ρ > 0.6), whereas in svPPA the concrete task correlated (ρ ≥ 0.75) with language tests. Patients with bvFTD, nfvPPA and lvPPA performed lower on the TRACE than the SAT (p < .05), whereas patients with svPPA were equally impaired on both tasks (p = .2). DISCUSSION We demonstrated impaired abstract semantic knowledge in patients with bvFTD, lvPPA, and svPPA, but not nfvPPA, with svPPA patients performing worse than the other subtypes. The TRACE was a good classifier between each patient group versus controls and between svPPA versus other patient groups. This highlights the value of incorporating semantic tests with abstract stimuli into standard neuropsychological assessment for early differential diagnosis of FTD subtypes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - L C Jiskoot
- Corresponding author at: Dr. Molewaterplein 40, Room: Nf-331, 3015 GD Rotterdam, the Netherlands. Tel.: 0031650031894. E-mail address:
| |
Collapse
|
37
|
Benson BC, Shaw PJ, Azzouz M, Highley JR, Hautbergue GM. Proteinopathies as Hallmarks of Impaired Gene Expression, Proteostasis and Mitochondrial Function in Amyotrophic Lateral Sclerosis. Front Neurosci 2022; 15:783624. [PMID: 35002606 PMCID: PMC8733206 DOI: 10.3389/fnins.2021.783624] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/26/2021] [Indexed: 01/15/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal adult-onset neurodegenerative disease characterized by progressive degeneration of upper and lower motor neurons. As with the majority of neurodegenerative diseases, the pathological hallmarks of ALS involve proteinopathies which lead to the formation of various polyubiquitylated protein aggregates in neurons and glia. ALS is a highly heterogeneous disease, with both familial and sporadic forms arising from the convergence of multiple disease mechanisms, many of which remain elusive. There has been considerable research effort invested into exploring these disease mechanisms and in recent years dysregulation of RNA metabolism and mitochondrial function have emerged as of crucial importance to the onset and development of ALS proteinopathies. Widespread alterations of the RNA metabolism and post-translational processing of proteins lead to the disruption of multiple biological pathways. Abnormal mitochondrial structure, impaired ATP production, dysregulation of energy metabolism and calcium homeostasis as well as apoptosis have been implicated in the neurodegenerative process. Dysfunctional mitochondria further accumulate in ALS motor neurons and reflect a wider failure of cellular quality control systems, including mitophagy and other autophagic processes. Here, we review the evidence for RNA and mitochondrial dysfunction as some of the earliest critical pathophysiological events leading to the development of ALS proteinopathies, explore their relative pathological contributions and their points of convergence with other key disease mechanisms. This review will focus primarily on mutations in genes causing four major types of ALS (C9ORF72, SOD1, TARDBP/TDP-43, and FUS) and in protein homeostasis genes (SQSTM1, OPTN, VCP, and UBQLN2) as well as sporadic forms of the disease. Finally, we will look to the future of ALS research and how an improved understanding of central mechanisms underpinning proteinopathies might inform research directions and have implications for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Bridget C Benson
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom
| | - Pamela J Shaw
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.,Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom
| | - Mimoun Azzouz
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.,Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom.,Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, United Kingdom
| | - J Robin Highley
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.,Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom.,Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, United Kingdom
| | - Guillaume M Hautbergue
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.,Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom.,Healthy Lifespan Institute (HELSI), University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
38
|
Shapiro NL, Todd EG, Billot B, Cash DM, Iglesias JE, Warren JD, Rohrer JD, Bocchetta M. In vivo hypothalamic regional volumetry across the frontotemporal dementia spectrum. Neuroimage Clin 2022; 35:103084. [PMID: 35717886 PMCID: PMC9218583 DOI: 10.1016/j.nicl.2022.103084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Frontotemporal dementia (FTD) is a spectrum of diseases characterised by language, behavioural and motor symptoms. Among the different subcortical regions implicated in the FTD symptomatology, the hypothalamus regulates various bodily functions, including eating behaviours which are commonly present across the FTD spectrum. The pattern of specific hypothalamic involvement across the clinical, pathological, and genetic forms of FTD has yet to be fully investigated, and its possible associations with abnormal eating behaviours have yet to be fully explored. METHODS Using an automated segmentation tool for volumetric T1-weighted MR images, we measured hypothalamic regional volumes in a cohort of 439 patients with FTD (197 behavioural variant FTD [bvFTD]; 7 FTD with associated motor neurone disease [FTD-MND]; 99 semantic variant primary progressive aphasia [svPPA]; 117 non-fluent variant PPA [nfvPPA]; 19 PPA not otherwise specified [PPA-NOS]) and 118 age-matched controls. We compared volumes across the clinical, genetic (29 MAPT, 32 C9orf72, 23 GRN), and pathological diagnoses (61 tauopathy, 40 TDP-43opathy, 4 FUSopathy). We correlated the volumes with presence of abnormal eating behaviours assessed with the revised version of the Cambridge Behavioural Inventory (CBI-R). RESULTS On average, FTD patients showed 14% smaller hypothalamic volumes than controls. The groups with the smallest hypothalamic regions were FTD-MND (20%), MAPT (25%) and FUS (33%), with differences mainly localised in the anterior and posterior regions. The inferior tuberal region was only significantly smaller in tauopathies (MAPT and Pick's disease) and in TDP-43 type C compared to controls and was the only regions that did not correlate with eating symptoms. PPA-NOS and nfvPPA were the groups with the least frequent eating behaviours and the least hypothalamic involvement. CONCLUSIONS Abnormal hypothalamic volumes are present in all the FTD forms, but different hypothalamic regions might play a different role in the development of abnormal eating behavioural and metabolic symptoms. These findings might therefore help in the identification of different underlying pathological mechanisms, suggesting the potential use of hypothalamic imaging biomarkers and the research of potential therapeutic targets within the hypothalamic neuropeptides.
Collapse
Affiliation(s)
- Noah L Shapiro
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Benjamin Billot
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK; Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, UK; UK Dementia Research Institute at UCL, UCL, London, UK
| | - Juan Eugenio Iglesias
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, UK; Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Boston, USA
| | - Jason D Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, UK.
| |
Collapse
|
39
|
De Rossi P, Lewis AJ, Furrer J, De Vos L, Demeter T, Zbinden A, Zhong W, Wiersma VI, Scialo C, Weber J, Guo Z, Scaramuzza S, Di Fabrizio M, Böing C, Castaño‐Díez D, Al‐Amoudi A, Pérez‐Berlanga M, Lashley T, Stahlberg H, Polymenidou M. FTLD-TDP assemblies seed neoaggregates with subtype-specific features via a prion-like cascade. EMBO Rep 2021; 22:e53877. [PMID: 34806807 PMCID: PMC8647015 DOI: 10.15252/embr.202153877] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022] Open
Abstract
Morphologically distinct TDP-43 aggregates occur in clinically different FTLD-TDP subtypes, yet the mechanism of their emergence and contribution to clinical heterogeneity are poorly understood. Several lines of evidence suggest that pathological TDP-43 follows a prion-like cascade, but the molecular determinants of this process remain unknown. We use advanced microscopy techniques to compare the seeding properties of pathological FTLD-TDP-A and FTLD-TDP-C aggregates. Upon inoculation of patient-derived aggregates in cells, FTLD-TDP-A seeds amplify in a template-dependent fashion, triggering neoaggregation more efficiently than those extracted from FTLD-TDP-C patients, correlating with the respective disease progression rates. Neoaggregates are sequentially phosphorylated with N-to-C directionality and with subtype-specific timelines. The resulting FTLD-TDP-A neoaggregates are large and contain densely packed fibrils, reminiscent of the pure compacted fibrils present within cytoplasmic inclusions in postmortem brains. In contrast, FTLD-TDP-C dystrophic neurites show less dense fibrils mixed with cellular components, and their respective neoaggregates are small, amorphous protein accumulations. These cellular seeding models replicate aspects of the patient pathological diversity and will be a useful tool in the quest for subtype-specific therapeutics.
Collapse
Affiliation(s)
- Pierre De Rossi
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Amanda J Lewis
- Laboratory of Biological Electron MicroscopyInstitute of Physics, SB, EPFLDepartment of Fundamental MicrobiologyFaculty of Biology and MedicineUNILLausanneSwitzerland
| | - Johanna Furrer
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Laura De Vos
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Tomas Demeter
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Aurélie Zbinden
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Weijia Zhong
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Vera I Wiersma
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Carlo Scialo
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Julien Weber
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Zhongning Guo
- Department of Quantitative BiomedicineUniversity of ZurichZurichSwitzerland
| | - Stefano Scaramuzza
- C‐CINABiozentrumUniversity of BaselBaselSwitzerland
- BioEM LabBiozentrumUniversity of BaselBaselSwitzerland
| | - Marta Di Fabrizio
- Laboratory of Biological Electron MicroscopyInstitute of Physics, SB, EPFLDepartment of Fundamental MicrobiologyFaculty of Biology and MedicineUNILLausanneSwitzerland
| | | | | | | | | | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological diseasesDepartment of Movement DisordersUCL Institute of NeurologyLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
| | - Henning Stahlberg
- Laboratory of Biological Electron MicroscopyInstitute of Physics, SB, EPFLDepartment of Fundamental MicrobiologyFaculty of Biology and MedicineUNILLausanneSwitzerland
| | | |
Collapse
|
40
|
Woo JA, Yan Y, Kee TR, Cazzaro S, McGill Percy KC, Wang X, Liu T, Liggett SB, Kang DE. β-arrestin1 promotes tauopathy by transducing GPCR signaling, disrupting microtubules and autophagy. Life Sci Alliance 2021; 5:5/3/e202101183. [PMID: 34862271 PMCID: PMC8675912 DOI: 10.26508/lsa.202101183] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 01/14/2023] Open
Abstract
GPCRs regulator, β-arrestin1, is increased in FTLD-tau patients, is required for β2-adrenergic receptor and metabotropic glutamate receptor 2-induced tau phosphorylation, promotes tau aggregation by impairing autophagy, and destabilizes microtubule dynamics, whereas genetic reduction in β-arrestin1 mitigates tauopathy and cognitive impairments. G protein–coupled receptors (GPCRs) have been shown to play integral roles in Alzheimer’s disease pathogenesis. However, it is unclear how diverse GPCRs similarly affect Aβ and tau pathogenesis. GPCRs share a common mechanism of action via the β-arrestin scaffolding signaling complexes, which not only serve to desensitize GPCRs by internalization, but also mediate multiple downstream signaling events. As signaling via the GPCRs, β2-adrenergic receptor (β2AR), and metabotropic glutamate receptor 2 (mGluR2) promotes hyperphosphorylation of tau, we hypothesized that β-arrestin1 represents a point of convergence for such pathogenic activities. Here, we report that β-arrestins are not only essential for β2AR and mGluR2-mediated increase in pathogenic tau but also show that β-arrestin1 levels are increased in brains of Frontotemporal lobar degeneration (FTLD-tau) patients. Increased β-arrestin1 in turn drives the accumulation of pathogenic tau, whereas reduced ARRB1 alleviates tauopathy and rescues impaired synaptic plasticity and cognitive impairments in PS19 mice. Biochemical and cellular studies show that β-arrestin1 drives tauopathy by destabilizing microtubules and impeding p62/SQSTM1 autophagy flux by interfering with p62 body formation, which promotes pathogenic tau accumulation.
Collapse
Affiliation(s)
- Jung-Aa Woo
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Yan Yan
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.,Department of Molecular Medicine, University of South Florida, College of Medicine, Tampa, FL, USA
| | - Teresa R Kee
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.,Department of Molecular Medicine, University of South Florida, College of Medicine, Tampa, FL, USA
| | - Sara Cazzaro
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.,Department of Molecular Medicine, University of South Florida, College of Medicine, Tampa, FL, USA
| | - Kyle C McGill Percy
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Xinming Wang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Tian Liu
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Stephen B Liggett
- Department of Molecular Pharmacology and Physiology, University of South Florida, College of Medicine, Tampa, FL, USA
| | - David E Kang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.,Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| |
Collapse
|
41
|
Bergström S, Öijerstedt L, Remnestål J, Olofsson J, Ullgren A, Seelaar H, van Swieten JC, Synofzik M, Sanchez-Valle R, Moreno F, Finger E, Masellis M, Tartaglia C, Vandenberghe R, Laforce R, Galimberti D, Borroni B, Butler CR, Gerhard A, Ducharme S, Rohrer JD, Månberg A, Graff C, Nilsson P. A panel of CSF proteins separates genetic frontotemporal dementia from presymptomatic mutation carriers: a GENFI study. Mol Neurodegener 2021; 16:79. [PMID: 34838088 PMCID: PMC8626910 DOI: 10.1186/s13024-021-00499-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 11/01/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND A detailed understanding of the pathological processes involved in genetic frontotemporal dementia is critical in order to provide the patients with an optimal future treatment. Protein levels in CSF have the potential to reflect different pathophysiological processes in the brain. We aimed to identify and evaluate panels of CSF proteins with potential to separate symptomatic individuals from individuals without clinical symptoms (unaffected), as well as presymptomatic individuals from mutation non-carriers. METHODS A multiplexed antibody-based suspension bead array was used to analyse levels of 111 proteins in CSF samples from 221 individuals from families with genetic frontotemporal dementia. The data was explored using LASSO and Random forest. RESULTS When comparing affected individuals with unaffected individuals, 14 proteins were identified as potentially important for the separation. Among these, four were identified as most important, namely neurofilament medium polypeptide (NEFM), neuronal pentraxin 2 (NPTX2), neurosecretory protein VGF (VGF) and aquaporin 4 (AQP4). The combined profile of these four proteins successfully separated the two groups, with higher levels of NEFM and AQP4 and lower levels of NPTX2 in affected compared to unaffected individuals. VGF contributed to the models, but the levels were not significantly lower in affected individuals. Next, when comparing presymptomatic GRN and C9orf72 mutation carriers in proximity to symptom onset with mutation non-carriers, six proteins were identified with a potential to contribute to a separation, including progranulin (GRN). CONCLUSION In conclusion, we have identified several proteins with the combined potential to separate affected individuals from unaffected individuals, as well as proteins with potential to contribute to the separation between presymptomatic individuals and mutation non-carriers. Further studies are needed to continue the investigation of these proteins and their potential association to the pathophysiological mechanisms in genetic FTD.
Collapse
Affiliation(s)
- Sofia Bergström
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
- Swedish FTD Initiative, Stockholm, Sweden
| | - Linn Öijerstedt
- Swedish FTD Initiative, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Unit of Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Julia Remnestål
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
- Swedish FTD Initiative, Stockholm, Sweden
| | - Jennie Olofsson
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
- Swedish FTD Initiative, Stockholm, Sweden
| | - Abbe Ullgren
- Swedish FTD Initiative, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Unit of Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Harro Seelaar
- Department of Neurology, Erasmus Medical Centre, Rotterdam, Netherlands
| | | | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany
- Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Raquel Sanchez-Valle
- 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
| | - Fermin Moreno
- Cognitive Disorders Unit, Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa Spain
- Neuroscience Area, Biodonostia Health Research Institute, San Sebastian, Gipuzkoa Spain
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario Canada
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
- Neurology Service, University Hospitals Leuven, Leuven, Belgium
- Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire, Département des Sciences Neurologiques, CHU de Québec, and Faculté de Médecine, Université Laval, QC, Canada
| | - Daniela Galimberti
- Fondazione IRCCS Ospedale Policlinico, Milan, Italy
- University of Milan, Centro Dino Ferrari, Milan, Italy
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Chris R. Butler
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Alexander Gerhard
- Division of Neuroscience and Experimental Psychology, Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
- Departments of Geriatric Medicine and Nuclear Medicine, University of Duisburg- Essen, Duisburg, Germany
| | - Simon Ducharme
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Québec Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Québec Canada
| | - Jonathan D. Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, UK
| | - Anna Månberg
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
- Swedish FTD Initiative, Stockholm, Sweden
| | - Caroline Graff
- Swedish FTD Initiative, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Unit of Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
- Unit for Hereditary Dementias, Theme Aging, Karolinska University Hospital, Solna, Sweden
| | - Peter Nilsson
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
- Swedish FTD Initiative, Stockholm, Sweden
| |
Collapse
|
42
|
de Boer SCM, Riedl L, van der Lee SJ, Otto M, Anderl-Straub S, Landin-Romero R, Sorrentino F, Fieldhouse JLP, Reus LM, Vacaflor B, Halliday G, Galimberti D, Diehl-Schmid J, Ducharme S, Piguet O, Pijnenburg YAL. Differences in Sex Distribution Between Genetic and Sporadic Frontotemporal Dementia. J Alzheimers Dis 2021; 84:1153-1161. [PMID: 34633319 PMCID: PMC8673542 DOI: 10.3233/jad-210688] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Reported sex distributions differ between frontotemporal dementia (FTD) cohorts. Possible explanations are the evolving clinical criteria of FTD and its subtypes and the discovery of FTD causal genetic mutations that has resulted in varying demographics. Objective: Our aim was to determine the sex distribution of sporadic and genetic FTD cases and its subtypes in an international cohort. Methods: We included 910 patients with behavioral variant frontotemporal dementia (bvFTD; n = 654), non-fluent variant primary progressive aphasia (nfvPPA; n = 99), semantic variant primary progressive aphasia (svPPA; n = 117), and right temporal variant frontotemporal dementia (rtvFTD; n = 40). We compared sex distribution between genetic and sporadic FTD using χ2-tests. Results: The genetic FTD group consisted of 51.2% males, which did not differ from sporadic FTD (57.8% male, p = 0.08). In the sporadic bvFTD subgroup, males were predominant in contrast to genetic bvFTD (61.6% versus 52.9% males, p = 0.04). In the other clinical FTD subgroups, genetic cases were underrepresented and within the sporadic cases the sex distribution was somewhat equal. Conclusion: The higher male prevalence in sporadic bvFTD may provide important clues for its differential pathogenesis and warrants further research.
Collapse
Affiliation(s)
- Sterre C M de Boer
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Lina Riedl
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Markus Otto
- Department of Neurology, University Clinic, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | | | - Ramon Landin-Romero
- School of Psychology, and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Federica Sorrentino
- Neurodegenerative Disease Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Jay L P Fieldhouse
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Lianne M Reus
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Blanca Vacaflor
- Department of Psychiatry, McGill University Health Center, Montreal, Quebec, QC, Canada
| | - Glenda Halliday
- School of Medical Sciences, and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Daniela Galimberti
- Neurodegenerative Disease Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, University of Milan, Milan, Italy
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, QC, Canada.,Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, Quebec, QC, Canada
| | - Olivier Piguet
- School of Psychology, and Brain & Mind Centre, The University of Sydney, Sydney, NSW, Australia
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Geser F, Mitrovics TCG, Haybaeck J, Yilmazer-Hanke D. Premorbid de novo artistic creativity in frontotemporal dementia (FTD) syndromes. J Neural Transm (Vienna) 2021; 128:1813-1833. [PMID: 34618237 DOI: 10.1007/s00702-021-02426-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/26/2021] [Indexed: 12/18/2022]
Abstract
The emergence of new artistic activities or shifts in artistic style in patients with frontotemporal dementia (FTD) syndromes is well documented at or after disease onset. However, a closer look in the literature reveals emerging artistic creativity also before FTD onset, although the significance and underlying pathology of such creative endeavors remain elusive. Here, we systematically review relevant studies and report an additional FTD case to elaborate on artistic activities that developed years before disease manifestation by paying particular attention to the sequence of events in individual patients' biography and clinical history. We further discuss the FTD patient's creative activities in the context of their life events, other initial or "premorbid" dementia symptoms or risk factors described in the literature such as mental illness and mild behavioral impairment (MBI), as well as changes in neuronal systems (i.e., neuroimaging and neuropathology). In addition to our FTD patient, we identified five published cases with an FTD syndrome, including three with FTD, one with primary progressive aphasia (PPA), and one with the behavioral variant of PPA (bvPPA). Premorbid novel creativity emerged across different domains (visual, musical, writing), with the FTD diagnosis ensuing artistic productivity by a median of 8 years. Data on late-life and pre-dementia life events were available in four cases. The late creative phase in our case was accompanied by personality changes, accentuation of personality traits, and cessation of painting activities occurred with the onset of memory complaints. Thus, premorbid personality changes in FTD patients can be associated with de novo creative activity. Stressful life events may also contribute to the burgeoning of creativity. Moreover, primary neocortical areas that are largely spared by pathology at early FTD stages may facilitate the engagement in artistic activities, offering a window of opportunity for art therapy and other therapeutic interventions during the MBI stage or even earlier.
Collapse
Affiliation(s)
- Felix Geser
- Department of Geriatric Psychiatry, Klinikum Christophsbad, Faurndauer Str. 6-28, 73035, Göppingen, Germany.
| | - Tibor C G Mitrovics
- Department of Radiology and Neuroradiology, Klinikum Christophsbad, Göppingen, Germany
| | - Johannes Haybaeck
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria.,Diagnostic & Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Deniz Yilmazer-Hanke
- Clinical Neuroanatomy, Department of Neurology, University Hospital, Ulm University, Ulm, Germany
| |
Collapse
|
44
|
A comparison of automated atrophy measures across the frontotemporal dementia spectrum: Implications for trials. NEUROIMAGE-CLINICAL 2021; 32:102842. [PMID: 34626889 PMCID: PMC8503665 DOI: 10.1016/j.nicl.2021.102842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/13/2021] [Accepted: 09/23/2021] [Indexed: 11/22/2022]
Abstract
Background Frontotemporal dementia (FTD) is a common cause of young onset dementia, and whilst there are currently no treatments, there are several promising candidates in development and early phase trials. Comprehensive investigations of neuroimaging markers of disease progression across the full spectrum of FTD disorders are lacking and urgently needed to facilitate these trials. Objective To investigate the comparative performance of multiple automated segmentation and registration pipelines used to quantify longitudinal whole-brain atrophy across the clinical, genetic and pathological subgroups of FTD, in order to inform upcoming trials about suitable neuroimaging-based endpoints. Methods Seventeen fully automated techniques for extracting whole-brain atrophy measures were applied and directly compared in a cohort of 226 participants who had undergone longitudinal structural 3D T1-weighted imaging. Clinical diagnoses were behavioural variant FTD (n = 56) and primary progressive aphasia (PPA, n = 104), comprising semantic variant PPA (n = 38), non-fluent variant PPA (n = 42), logopenic variant PPA (n = 18), and PPA-not otherwise specified (n = 6). 49 of these patients had either a known pathogenic mutation or postmortem confirmation of their underlying pathology. 66 healthy controls were included for comparison. Sample size estimates to detect a 30% reduction in atrophy (80% power; 0.05 significance) were computed to explore the relative feasibility of these brain measures as surrogate markers of disease progression and their ability to detect putative disease-modifying treatment effects. Results Multiple automated techniques showed great promise, detecting significantly increased rates of whole-brain atrophy (p<0.001) and requiring sample sizes of substantially less than 100 patients per treatment arm. Across the different FTD subgroups, direct measures of volume change consistently outperformed their indirect counterparts, irrespective of the initial segmentation quality. Significant differences in performance were found between both techniques and patient subgroups, highlighting the importance of informed biomarker choice based on the patient population of interest. Conclusion This work expands current knowledge and builds on the limited longitudinal investigations currently available in FTD, as well as providing valuable information about the potential of fully automated neuroimaging biomarkers for sporadic and genetic FTD trials.
Collapse
|
45
|
HnRNP K mislocalisation is a novel protein pathology of frontotemporal lobar degeneration and ageing and leads to cryptic splicing. Acta Neuropathol 2021; 142:609-627. [PMID: 34274995 PMCID: PMC8423707 DOI: 10.1007/s00401-021-02340-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 02/08/2023]
Abstract
Heterogeneous nuclear ribonucleoproteins (HnRNPs) are a group of ubiquitously expressed RNA-binding proteins implicated in the regulation of all aspects of nucleic acid metabolism. HnRNP K is a member of this highly versatile hnRNP family. Pathological redistribution of hnRNP K to the cytoplasm has been linked to the pathogenesis of several malignancies but, until now, has been underexplored in the context of neurodegenerative disease. Here we show hnRNP K mislocalisation in pyramidal neurons of the frontal cortex to be a novel neuropathological feature that is associated with both frontotemporal lobar degeneration and ageing. HnRNP K mislocalisation is mutually exclusive to TDP-43 and tau pathological inclusions in neurons and was not observed to colocalise with mitochondrial, autophagosomal or stress granule markers. De-repression of cryptic exons in RNA targets following TDP-43 nuclear depletion is an emerging mechanism of potential neurotoxicity in frontotemporal lobar degeneration and the mechanistically overlapping disorder amyotrophic lateral sclerosis. We silenced hnRNP K in neuronal cells to identify the transcriptomic consequences of hnRNP K nuclear depletion. Intriguingly, by performing RNA-seq analysis we find that depletion of hnRNP K induces 101 novel cryptic exon events. We validated cryptic exon inclusion in an SH-SY5Y hnRNP K knockdown and in FTLD brain exhibiting hnRNP K nuclear depletion. We, therefore, present evidence for hnRNP K mislocalisation to be associated with FTLD and for this to induce widespread changes in splicing.
Collapse
|
46
|
Leveille E, Ross OA, Gan-Or Z. Tau and MAPT genetics in tauopathies and synucleinopathies. Parkinsonism Relat Disord 2021; 90:142-154. [PMID: 34593302 PMCID: PMC9310195 DOI: 10.1016/j.parkreldis.2021.09.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/25/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
MAPT encodes the microtubule-associated protein tau, which is the main component of neurofibrillary tangles (NFTs) and found in other protein aggregates. These aggregates are among the pathological hallmarks of primary tauopathies such as frontotemporal dementia (FTD). Abnormal tau can also be observed in secondary tauopathies such as Alzheimer's disease (AD) and synucleinopathies such as Parkinson's disease (PD). On top of pathological findings, genetic data also links MAPT to these disorders. MAPT variations are a cause or risk factors for many tauopathies and synucleinopathies and are associated with certain clinical and pathological features in affected individuals. In addition to clinical, pathological, and genetic overlap, evidence also suggests that tau and alpha-synuclein may interact on the molecular level, and thus might collaborate in the neurodegenerative process. Understanding the role of MAPT variations in tauopathies and synucleinopathies is therefore essential to elucidate the role of tau in the pathogenesis and phenotype of those disorders, and ultimately to develop targeted therapies. In this review, we describe the role of MAPT genetic variations in tauopathies and synucleinopathies, several genotype-phenotype and pathological features, and discuss their implications for the classification and treatment of those disorders.
Collapse
Affiliation(s)
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA; Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Ziv Gan-Or
- The Neuro (Montreal Neurological Institute-hospital), McGill University, Montréal, QC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada.
| |
Collapse
|
47
|
Bocchetta M, Malpetti M, Todd EG, Rowe JB, Rohrer JD. Looking beneath the surface: the importance of subcortical structures in frontotemporal dementia. Brain Commun 2021; 3:fcab158. [PMID: 34458729 PMCID: PMC8390477 DOI: 10.1093/braincomms/fcab158] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
Whilst initial anatomical studies of frontotemporal dementia focussed on cortical involvement, the relevance of subcortical structures to the pathophysiology of frontotemporal dementia has been increasingly recognized over recent years. Key structures affected include the caudate, putamen, nucleus accumbens, and globus pallidus within the basal ganglia, the hippocampus and amygdala within the medial temporal lobe, the basal forebrain, and the diencephalon structures of the thalamus, hypothalamus and habenula. At the most posterior aspect of the brain, focal involvement of brainstem and cerebellum has recently also been shown in certain subtypes of frontotemporal dementia. Many of the neuroimaging studies on subcortical structures in frontotemporal dementia have been performed in clinically defined sporadic cases. However, investigations of genetically- and pathologically-confirmed forms of frontotemporal dementia are increasingly common and provide molecular specificity to the changes observed. Furthermore, detailed analyses of sub-nuclei and subregions within each subcortical structure are being added to the literature, allowing refinement of the patterns of subcortical involvement. This review focuses on the existing literature on structural imaging and neuropathological studies of subcortical anatomy across the spectrum of frontotemporal dementia, along with investigations of brain–behaviour correlates that examine the cognitive sequelae of specific subcortical involvement: it aims to ‘look beneath the surface’ and summarize the patterns of subcortical involvement have been described in frontotemporal dementia.
Collapse
Affiliation(s)
- Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Maura Malpetti
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Emily G Todd
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK.,Medical Research Council Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
48
|
Duran-Aniotz C, Orellana P, Leon Rodriguez T, Henriquez F, Cabello V, Aguirre-Pinto MF, Escobedo T, Takada LT, Pina-Escudero SD, Lopez O, Yokoyama JS, Ibanez A, Parra MA, Slachevsky A. Systematic Review: Genetic, Neuroimaging, and Fluids Biomarkers for Frontotemporal Dementia Across Latin America Countries. Front Neurol 2021; 12:663407. [PMID: 34248820 PMCID: PMC8263937 DOI: 10.3389/fneur.2021.663407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/27/2021] [Indexed: 11/13/2022] Open
Abstract
Frontotemporal dementia (FTD) includes a group of clinically, genetically, and pathologically heterogeneous neurodegenerative disorders, affecting the fronto-insular-temporal regions of the brain. Clinically, FTD is characterized by progressive deficits in behavior, executive function, and language and its diagnosis relies mainly on the clinical expertise of the physician/consensus group and the use of neuropsychological tests and/or structural/functional neuroimaging, depending on local availability. The modest correlation between clinical findings and FTD neuropathology makes the diagnosis difficult using clinical criteria and often leads to underdiagnosis or misdiagnosis, primarily due to lack of recognition or awareness of FTD as a disease and symptom overlap with psychiatric disorders. Despite advances in understanding the underlying neuropathology of FTD, accurate and sensitive diagnosis for this disease is still lacking. One of the major challenges is to improve diagnosis in FTD patients as early as possible. In this context, biomarkers have emerged as useful methods to provide and/or complement clinical diagnosis for this complex syndrome, although more evidence is needed to incorporate most of them into clinical practice. However, most biomarker studies have been performed using North American or European populations, with little representation of the Latin American and the Caribbean (LAC) region. In the LAC region, there are additional challenges, particularly the lack of awareness and knowledge about FTD, even in specialists. Also, LAC genetic heritage and cultures are complex, and both likely influence clinical presentations and may modify baseline biomarker levels. Even more, due to diagnostic delay, the clinical presentation might be further complicated by both neurological and psychiatric comorbidity, such as vascular brain damage, substance abuse, mood disorders, among others. This systematic review provides a brief update and an overview of the current knowledge on genetic, neuroimaging, and fluid biomarkers for FTD in LAC countries. Our review highlights the need for extensive research on biomarkers in FTD in LAC to contribute to a more comprehensive understanding of the disease and its associated biomarkers. Dementia research is certainly reduced in the LAC region, highlighting an urgent need for harmonized, innovative, and cross-regional studies with a global perspective across multiple areas of dementia knowledge.
Collapse
Affiliation(s)
- Claudia Duran-Aniotz
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Paulina Orellana
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Tomas Leon Rodriguez
- Trinity College, Global Brain Health Institute, Dublin, Ireland
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fernando Henriquez
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Victoria Cabello
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | | | - Tamara Escobedo
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
| | - Leonel T. Takada
- Cognitive and Behavioral Neurology Unit - Department of Neurology, University of São Paulo, São Paulo, Brazil
| | - Stefanie D. Pina-Escudero
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA, United States
- UCSF Department of Neurology, Memory and Aging Center, UCSF, San Francisco, CA, United States
| | - Oscar Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jennifer S. Yokoyama
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA, United States
- UCSF Department of Neurology, Memory and Aging Center, UCSF, San Francisco, CA, United States
| | - Agustin Ibanez
- Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibanez, Santiago, Chile
- Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile
- Trinity College, Global Brain Health Institute, Dublin, Ireland
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA, United States
- Cognitive Neuroscience Center (CNC), Universidad de San Andrés, & National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Mario A. Parra
- School of Psychological Sciences and Health, University of Strathclyde, Glasgow, United Kingdom
| | - Andrea Slachevsky
- Memory and Neuropsychiatric Clinic (CMYN) Neurology Department, Hospital del Salvador and Faculty of Medicine, University of Chile, Santiago, Chile
- Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department - Institute of Biomedical Sciences (ICBM), Neuroscience and East Neuroscience Departments, Faculty of Medicine, University of Chile, Santiago, Chile
- Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
- Cognitive and Behavioral Neurology Unit - Department of Neurology, University of São Paulo, São Paulo, Brazil
- Department of Neurology and Psychiatry, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
49
|
Gaetani L, Parnetti L, Calabresi P, Di Filippo M. Tracing Neurological Diseases in the Presymptomatic Phase: Insights From Neurofilament Light Chain. Front Neurosci 2021; 15:672954. [PMID: 34108859 PMCID: PMC8180886 DOI: 10.3389/fnins.2021.672954] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/16/2021] [Indexed: 11/13/2022] Open
Abstract
The identification of neurological diseases in their presymptomatic phase will be a fundamental aim in the coming years. This step is necessary both to optimize early diagnostics and to verify the effectiveness of experimental disease modifying drugs in the early stages of diseases. Among the biomarkers that can detect neurological diseases already in their preclinical phase, neurofilament light chain (NfL) has given the most promising results. Recently, its measurement in serum has enabled the identification of neurodegeneration in diseases such as multiple sclerosis (MS) and Alzheimer’s disease (AD) up to 6–10 years before the onset of symptoms. Similar results have been obtained in conditions such as frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS), up to 2 years before clinical onset. Study of the longitudinal dynamics of serum NfL has also revealed interesting aspects of the pathophysiology of these diseases in the preclinical phase. This review sought to discuss these very recent findings on serum NfL in the presymptomatic phase of neurological diseases.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Calabresi
- Neurology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.,Neuroscience Department, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
50
|
Poos JM, Russell LL, Peakman G, Bocchetta M, Greaves CV, Jiskoot LC, van der Ende EL, Seelaar H, Papma JM, van den Berg E, Pijnenburg YA, Borroni B, Sanchez‐Valle R, Moreno F, Laforce R, Graff C, Synofzik M, Galimberti D, Rowe JB, Masellis M, Tartaglia C, Finger E, Vandenberghe R, de Medonça A, Tagliavini F, Butler CR, Santana I, Ber IL, Gerhard A, Ducharme S, Levin J, Danek A, Otto M, Sorbi S, Pasquier F, van Swieten JC, Rohrer JD. Impairment of episodic memory in genetic frontotemporal dementia: A GENFI study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12185. [PMID: 34027016 PMCID: PMC8116844 DOI: 10.1002/dad2.12185] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/06/2021] [Accepted: 03/09/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION We aimed to assess episodic memory in genetic frontotemporal dementia (FTD) with the Free and Cued Selective Reminding Test (FCSRT). METHODS The FCSRT was administered in 417 presymptomatic and symptomatic mutation carriers (181 chromosome 9 open reading frame 72 [C9orf72], 163 progranulin [GRN], and 73 microtubule-associated protein tau [MAPT]) and 290 controls. Group differences and correlations with other neuropsychological tests were examined. We performed voxel-based morphometry to investigate the underlying neural substrates of the FCSRT. RESULTS All symptomatic mutation carrier groups and presymptomatic MAPT mutation carriers performed significantly worse on all FCSRT scores compared to controls. In the presymptomatic C9orf72 group, deficits were found on all scores except for the delayed total recall task, while no deficits were found in presymptomatic GRN mutation carriers. Performance on the FCSRT correlated with executive function, particularly in C9orf72 mutation carriers, but also with memory and naming tasks in the MAPT group. FCSRT performance also correlated with gray matter volumes of frontal, temporal, and subcortical regions in C9orf72 and GRN, but mainly temporal areas in MAPT mutation carriers. DISCUSSION The FCSRT detects presymptomatic deficits in C9orf72- and MAPT-associated FTD and provides important insight into the underlying cause of memory impairment in different forms of FTD.
Collapse
|