1
|
Van Der Lee SJ, Hulsman M, Van Spaendonk R, Van Der Schaar J, Dijkstra J, Tesi N, van Ruissen F, Elting M, Reinders M, De Rojas I, Verschuuren-Bemelmans CC, Van Der Flier WM, van Haelst MM, de Geus C, Pijnenburg Y, Holstege H. Prevalence of Pathogenic Variants and Eligibility Criteria for Genetic Testing in Patients Who Visit a Memory Clinic. Neurology 2025; 104:e210273. [PMID: 39869842 PMCID: PMC11776143 DOI: 10.1212/wnl.0000000000210273] [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: 06/24/2024] [Accepted: 11/27/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Identifying genetic causes of dementia in patients visiting memory clinics is important for patient care and family planning. Traditional clinical selection criteria for genetic testing may miss carriers of pathogenic variants in dementia-related genes. This study aimed identify how many carriers we are missing and to optimize criteria for selecting patients for genetic counseling in memory clinics. METHODS In this clinical cohort study, we retrospectively genetically tested patients during 2.5 years (2010-2012) visiting the Alzheimer Center Amsterdam, a specialized memory clinic. Genetic tests consisted of a 54-gene dementia panel, focusing on Class IV/V variants per American College of Medical Genetics and Genomics guidelines, including APP duplications and the C9ORF72 repeat expansion. We determined the prevalence of pathogenic variants and propose new eligibility criteria for genetic testing in memory clinics. The eligibility criteria were prospectively applied for 1 year (2021-2022), and results were compared with the retrospective cohort. RESULTS Genetic tests were retrospectively performed in in 1,022 of 1,138 patients (90%) who consecutively visited the memory clinic. Among these, 1,022 patients analyzed (mean age 62.1 ± 8.9 years; 40.4% were female), 34 pathogenic variant carriers were identified (3.3%), with 24 being symptomatic. Previous clinical criteria would have identified only 15 carriers (44% of all carriers, 65% of symptomatic carriers). The proposed criteria increased identification to 22 carriers (62.5% of all carriers, 91% of symptomatic carriers). In the prospective cohort, 148 (28.7%) of 515 patients were eligible for testing under the new criteria. Of the 90 eligible patients who consented to testing, 13 pathogenic carriers were identified, representing a 73% increase compared with the previous criteria. DISCUSSION We found that patients who visit a memory clinic and carry a pathogenic genetic variant are often not eligible for genetic testing. The proposed new criteria improve the identification of patients with a genetic cause for their cognitive complaints. In systems without practical or financial barriers to genetic testing, the new criteria can enhance personalized care. In other countries where the health care systems differs and in other genetic ancestry groups, the performance of the criteria may be different.
Collapse
Affiliation(s)
- Sven J Van Der Lee
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, the Netherlands
| | - Marc Hulsman
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, the Netherlands
| | - Rosalina Van Spaendonk
- Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Jetske Van Der Schaar
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, the Netherlands
| | - Janna Dijkstra
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, the Netherlands
| | - Niccoló Tesi
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
| | - Fred van Ruissen
- Department of Human Genetics, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, the Netherlands
| | - Mariet Elting
- Clinical Genetics, Dept. Human Genetics, Amsterdam UMC, the Netherlands
| | - Marcel Reinders
- Delft Bioinformatics Lab, Delft University of Technology, Delft, the Netherlands
| | - Itziar De Rojas
- Research Center and Memory Clinic, Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- CIBERNED, Network Center for Biomedical Research in Neurodegenerative Diseases, National Institute of Health Carlos III, Madrid, Spain
| | | | - Wiesje M Van Der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, the Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
| | - Mieke M van Haelst
- Clinical Genetics, Dept. Human Genetics, Amsterdam UMC, the Netherlands
- Amsterdam Reproduction and Development, Amsterdam UMC, the Netherlands; and
- Emma Center for Personalized Medicine, Amsterdam UMC, the Netherlands
| | - Christa de Geus
- Clinical Genetics, Dept. Human Genetics, Amsterdam UMC, the Netherlands
| | - Yolande Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, the Netherlands
| | - Henne Holstege
- Genomics of Neurodegenerative Diseases and Aging, Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, the Netherlands
- Amsterdam Neuroscience, Neurodegeneration, the Netherlands
| |
Collapse
|
2
|
Langheinrich TC, Thompson JC, Jones M, Richardson AMT, Mann DMA, Snowden JS. Apraxia phenotypes and frontotemporal lobar degeneration. J Neurol 2024; 271:7471-7488. [PMID: 39387948 PMCID: PMC11588949 DOI: 10.1007/s00415-024-12706-5] [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: 05/02/2024] [Revised: 08/13/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Apraxia has been identified in all clinical forms of frontotemporal lobar degeneration (FTLD). The characteristics of apraxia symptoms and their underlying cognitive/motor basis are not fully understood. This study investigated apraxia in pathological subtypes of FTLD. METHODS The study constituted a retrospective review of 115 pathologically confirmed cases of FTLD from a single cognitive neurology centre. Patients in whom apraxia had been documented as a notable clinical characteristic were identified. Apraxia features, demographic, cognitive, neurological, and imaging findings were recorded. RESULTS Eighteen patients were identified: 12 with FTLD-tau pathology (7 corticobasal degeneration (CBD), four Pick type and one progressive supranuclear palsy (PSP)) and six with FTLD-TDP pathology, all type A and four linked to progranulin gene mutations. Apraxia as a dominant presenting feature was typically associated with tau pathologies, whereas it emerged in the context of aphasia in TDP pathology. Apraxia typically predominated in one body part (face or limb) in tau but not TDP pathology. Relatively preserved activities in daily life were associated with TDP. Apraxia of speech was associated with tau pathology. Pick-type pathology was linked to symmetrical atrophy and late development of limb rigidity. CONCLUSION Apraxia in FTLD subtypes has variable characteristics. Apraxia associated with CBD pathology conformed to criteria for probable corticobasal syndrome (CBS), whereas apraxia with Pick-type pathology did not. Apraxia in patients with TDP-A pathology was interpreted as one manifestation of their generalised communication disorder. Apraxia in FTLD may have distinct cognitive and motor substrates that require prospective investigation.
Collapse
Affiliation(s)
- Tobias C Langheinrich
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Northern Care AllianceNHS Foundation Trust, Salford, UK.
- Division of Psychology, Communication & Human Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Jennifer C Thompson
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Northern Care AllianceNHS Foundation Trust, Salford, UK
- Division of Psychology, Communication & Human Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Matthew Jones
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Northern Care AllianceNHS Foundation Trust, Salford, UK
- Division of Medical Education, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Anna M T Richardson
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Northern Care AllianceNHS Foundation Trust, Salford, UK
- Division of Medical Education, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - David M A Mann
- Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Julie S Snowden
- Cerebral Function Unit, Manchester Centre for Clinical Neurosciences, Northern Care AllianceNHS Foundation Trust, Salford, UK
- Division of Neuroscience, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
3
|
Ward M, Carter LP, Huang JY, Maslyar D, Budda B, Paul R, Rosenthal A. Phase 1 study of latozinemab in progranulin-associated frontotemporal dementia. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12452. [PMID: 38356474 PMCID: PMC10865485 DOI: 10.1002/trc2.12452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 02/16/2024]
Abstract
INTRODUCTION Heterozygous mutations in the GRN gene lead to reduced progranulin (PGRN) levels in plasma and cerebrospinal fluid (CSF) and are causative of frontotemporal dementia (FTD) with > 90% penetrance. Latozinemab is a human monoclonal immunoglobulin G1 antibody that is being developed to increase PGRN levels in individuals with FTD caused by heterozygous loss-of-function GRN mutations. METHODS A first-in-human phase 1 study was conducted to evaluate the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of multiple-dose intravenous administration of latozinemab in eight symptomatic participants with FTD caused by a heterozygous loss-of-function GRN mutation (FTD-GRN). RESULTS Latozinemab demonstrated favorable safety and PK/PD profiles. Multiple-dose administration of latozinemab increased plasma and CSF PGRN levels in participants with FTD-GRN to levels that approximated those seen in healthy volunteers. DISCUSSION Data from the first-in-human phase 1 study support further development of latozinemab for the treatment of FTD-GRN. Highlights GRN mutations decrease progranulin (PGRN) and cause frontotemporal dementia (FTD).Latozinemab is being developed as a PGRN-elevating therapy.Latozinemab demonstrated a favorable safety profile in a phase 1 clinical trial.Latozinemab increased PGRN levels in the CNS of symptomatic FTD-GRN participants.
Collapse
Affiliation(s)
| | | | | | | | | | - Robert Paul
- Alector Inc.South San FranciscoCaliforniaUSA
- Present address:
Nine Square Therapeutics, Inc.South San FranciscoCalifornia94080USA
| | | |
Collapse
|
4
|
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
|
5
|
Kurnellas M, Mitra A, Schwabe T, Paul R, Arrant AE, Roberson ED, Ward M, Yeh F, Long H, Rosenthal A. Latozinemab, a novel progranulin-elevating therapy for frontotemporal dementia. J Transl Med 2023; 21:387. [PMID: 37322482 PMCID: PMC10268535 DOI: 10.1186/s12967-023-04251-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 06/08/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Heterozygous loss-of-function mutations in the progranulin (PGRN) gene (GRN) cause a reduction in PGRN and lead to the development of frontotemporal dementia (FTD-GRN). PGRN is a secreted lysosomal chaperone, immune regulator, and neuronal survival factor that is shuttled to the lysosome through multiple receptors, including sortilin. Here, we report the characterization of latozinemab, a human monoclonal antibody that decreases the levels of sortilin, which is expressed on myeloid and neuronal cells and shuttles PGRN to the lysosome for degradation, and blocks its interaction with PGRN. METHODS In vitro characterization studies were first performed to assess the mechanism of action of latozinemab. After the in vitro studies, a series of in vivo studies were performed to assess the efficacy of a mouse-cross reactive anti-sortilin antibody and the pharmacokinetics, pharmacodynamics, and safety of latozinemab in nonhuman primates and humans. RESULTS In a mouse model of FTD-GRN, the rodent cross-reactive anti-sortilin antibody, S15JG, decreased total sortilin levels in white blood cell (WBC) lysates, restored PGRN to normal levels in plasma, and rescued a behavioral deficit. In cynomolgus monkeys, latozinemab decreased sortilin levels in WBCs and concomitantly increased plasma and cerebrospinal fluid (CSF) PGRN by 2- to threefold. Finally, in a first-in-human phase 1 clinical trial, a single infusion of latozinemab caused a reduction in WBC sortilin, tripled plasma PGRN and doubled CSF PGRN in healthy volunteers, and restored PGRN to physiological levels in asymptomatic GRN mutation carriers. CONCLUSIONS These findings support the development of latozinemab for the treatment of FTD-GRN and other neurodegenerative diseases where elevation of PGRN may be beneficial. Trial registration ClinicalTrials.gov, NCT03636204. Registered on 17 August 2018, https://clinicaltrials.gov/ct2/show/NCT03636204 .
Collapse
Affiliation(s)
- Michael Kurnellas
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA.
- Neuron23, South San Francisco, CA, 94080, USA.
| | - Ananya Mitra
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| | - Tina Schwabe
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, 94080, USA
| | - Robert Paul
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
- Nine Square Therapeutics, Inc., South San Francisco, CA, 94080, USA
| | - Andrew E Arrant
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, and Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, and Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael Ward
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| | - Felix Yeh
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
- Genentech, South San Francisco, CA, 94080, USA
| | - Hua Long
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| | - Arnon Rosenthal
- Alector, Inc., 131 Oyster Point Blvd, #600, South San Francisco, CA, 94080, USA
| |
Collapse
|
6
|
Zecca C, Tortelli R, Carrera P, Dell'Abate MT, Logroscino G, Ferrari M. Genotype-phenotype correlation in the spectrum of frontotemporal dementia-parkinsonian syndromes and advanced diagnostic approaches. Crit Rev Clin Lab Sci 2022; 60:171-188. [PMID: 36510705 DOI: 10.1080/10408363.2022.2150833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The term frontotemporal dementia (FTD) refers to a group of progressive neurodegenerative disorders characterized mainly by atrophy of the frontal and anterior temporal lobes. Based on clinical presentation, three main clinical syndromes have traditionally been described: behavioral variant frontotemporal dementia (bvFTD), non-fluent/agrammatic primary progressive aphasia (nfPPA), and semantic variant PPA (svPPA). However, over the last 20 years, it has been recognized that cognitive phenotypes often overlap with motor phenotypes, either motor neuron diseases or parkinsonian signs and/or syndromes like progressive supranuclear palsy (PSP) and cortico-basal syndrome (CBS). Furthermore, FTD-related genes are characterized by genetic pleiotropy and can cause, even in the same family, pure motor phenotypes, findings that underlie the clinical continuum of the spectrum, which has pure cognitive and pure motor phenotypes as the extremes. The genotype-phenotype correlation of the spectrum, FTD-motor neuron disease, has been well defined and extensively investigated, while the continuum, FTD-parkinsonism, lacks a comprehensive review. In this narrative review, we describe the current knowledge about the genotype-phenotype correlation of the spectrum, FTD-parkinsonism, focusing on the phenotypes that are less frequent than bvFTD, namely nfPPA, svPPA, PSP, CBS, and cognitive-motor overlapping phenotypes (i.e. PPA + PSP). From a pathological point of view, they are characterized mainly by the presence of phosphorylated-tau inclusions, either 4 R or 3 R. The genetic correlate of the spectrum can be heterogeneous, although some variants seem to lead preferentially to specific clinical syndromes. Furthermore, we critically review the contribution of genome-wide association studies (GWAS) and next-generation sequencing (NGS) in disentangling the complex heritability of the FTD-parkinsonism spectrum and in defining the genotype-phenotype correlation of the entire clinical scenario, owing to the ability of these techniques to test multiple genes, and so to allow detailed investigations of the overlapping phenotypes. Finally, we conclude with the importance of a detailed genetic characterization and we offer to patients and families the chance to be included in future randomized clinical trials focused on autosomal dominant forms of FTLD.
Collapse
Affiliation(s)
- Chiara Zecca
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy
| | - Rosanna Tortelli
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis and Clinical Molecular Biology Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Teresa Dell'Abate
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy
| | - Giancarlo Logroscino
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy.,Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | | |
Collapse
|
7
|
Borghesani V, DeLeon J, Gorno-Tempini ML. Frontotemporal dementia: A unique window on the functional role of the temporal lobes. HANDBOOK OF CLINICAL NEUROLOGY 2022; 187:429-448. [PMID: 35964986 PMCID: PMC9793689 DOI: 10.1016/b978-0-12-823493-8.00011-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Frontotemporal dementia (FTD) is an umbrella term covering a plethora of progressive changes in executive functions, motor abilities, behavior, and/or language. Different clinical syndromes have been described in relation to localized atrophy, informing on the functional networks that underlie these specific cognitive, emotional, and behavioral processes. These functional declines are linked with the underlying neurodegeneration of frontal and/or temporal lobes due to diverse molecular pathologies. Initially, the accumulation of misfolded proteins targets specifically susceptible cell assemblies, leading to relatively focal neurodegeneration that later spreads throughout large-scale cortical networks. Here, we discuss the most recent clinical, neuropathological, imaging, and genetics findings in FTD-spectrum syndromes affecting the temporal lobe. We focus on the semantic variant of primary progressive aphasia and its mirror image, the right temporal variant of FTD. Incipient focal atrophy of the left anterior temporal lobe (ATL) manifests with predominant naming, word comprehension, reading, and object semantic deficits, while cases of predominantly right ATL atrophy present with impairments of socioemotional, nonverbal semantic, and person-specific knowledge. Overall, the observations in FTD allow for crucial clinical-anatomic inferences, shedding light on the role of the temporal lobes in both cognition and complex behaviors. The concerted activity of both ATLs is critical to ensure that percepts are translated into concepts, yet important hemispheric differences should be acknowledged. On one hand, the left ATL attributes meaning to linguistic, external stimuli, thus supporting goal-oriented, action-related behaviors (e.g., integrating sounds and letters into words). On the other hand, the right ATL assigns meaning to emotional, visceral stimuli, thus guiding socially relevant behaviors (e.g., integrating body sensations into feelings of familiarity).
Collapse
Affiliation(s)
- Valentina Borghesani
- Centre de recherche de l'Institut universitaire de gériatrie de Montréal, Montréal, QC, Canada; Department of Psychology, Université de Montréal, Montréal, QC, Canada.
| | - Jessica DeLeon
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, United States; Department of Neurology, Dyslexia Center, University of California, San Francisco, CA, United States
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, United States; Department of Neurology, Dyslexia Center, University of California, San Francisco, CA, United States
| |
Collapse
|
8
|
Zampatti S, Ragazzo M, Peconi C, Luciano S, Gambardella S, Caputo V, Strafella C, Cascella R, Caltagirone C, Giardina E. Genetic Counselling Improves the Molecular Characterisation of Dementing Disorders. J Pers Med 2021; 11:474. [PMID: 34073306 PMCID: PMC8227097 DOI: 10.3390/jpm11060474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/31/2022] Open
Abstract
Dementing disorders are a complex group of neurodegenerative diseases characterised by different, but often overlapping, pathological pathways. Genetics have been largely associated with the development or the risk to develop dementing diseases. Recent advances in molecular technologies permit analyzing of several genes in a small time, but the interpretation analysis is complicated by several factors: the clinical complexity of neurodegenerative disorders, the frequency of co-morbidities, and the high phenotypic heterogeneity of genetic diseases. Genetic counselling supports the diagnostic path, providing an accurate familial and phenotypic characterisation of patients. In this review, we summarise neurodegenerative dementing disorders and their genetic determinants. Genetic variants and associated phenotypes will be divided into high and low impact, in order to reflect the pathologic continuum between multifactorial and mendelian genetic factors. Moreover, we report a molecular characterisation of genes associated with neurodegenerative disorders with cognitive impairment. In particular, the high frequency of rare coding genetic variants in dementing genes strongly supports the role of geneticists in both, clinical phenotype characterisation and interpretation of genotypic data. The smart application of exome analysis to dementia patients, with a pre-analytical selection on familial, clinical, and instrumental features, improves the diagnostic yield of genetic test, reduces time for diagnosis, and allows a rapid and personalised management of disease.
Collapse
Affiliation(s)
- Stefania Zampatti
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (S.Z.); (C.P.); (S.L.); (C.S.); (R.C.)
| | - Michele Ragazzo
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy; (M.R.); (V.C.)
| | - Cristina Peconi
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (S.Z.); (C.P.); (S.L.); (C.S.); (R.C.)
| | - Serena Luciano
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (S.Z.); (C.P.); (S.L.); (C.S.); (R.C.)
| | - Stefano Gambardella
- IRCCS Neuromed, 86077 Pozzilli, Italy;
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, 61029 Urbino, Italy
| | - Valerio Caputo
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy; (M.R.); (V.C.)
| | - Claudia Strafella
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (S.Z.); (C.P.); (S.L.); (C.S.); (R.C.)
| | - Raffaella Cascella
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (S.Z.); (C.P.); (S.L.); (C.S.); (R.C.)
- Department of Biomedical Sciences, Catholic University Our Lady of Good Counsel, 1000 Tirana, Albania
| | - Carlo Caltagirone
- Department of Clinical and Behavioral Neurology, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy;
| | - Emiliano Giardina
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (S.Z.); (C.P.); (S.L.); (C.S.); (R.C.)
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy; (M.R.); (V.C.)
| |
Collapse
|
9
|
Caso F, Agosta F, Magnani G, Cardamone R, Borghesani V, Miller Z, Riva N, La Joie R, Coppola G, Grinberg LT, Seeley WW, Miller BL, Gorno-Tempini ML, Filippi M. Temporal variant of frontotemporal dementia in C9orf72 repeat expansion carriers: two case studies. Brain Imaging Behav 2021; 14:336-345. [PMID: 32180125 DOI: 10.1007/s11682-019-00253-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The temporal variant of frontotemporal dementia (tv-FTD) is a progressive neurodegenerative disease with a complex clinical picture mainly characterized by behavioral and language disorders. In this work, we describe clinical, genetic, neuroanatomical and neuropathological (only in one case) features of two patients with tv-FTD carrying C9orf72 repeat expansion. The first patient (AB) presented with a 1-year disease duration showing focal right anterior temporal lobe (ATL) atrophy on magnetic resonance imaging (MRI). The second patient (BC) came to medical attention 13 years after disease onset and showed a prominent bilateral ATL involvement. Both patients showed naming deficits, impairment in identifying known faces and proper names, and personality changes with new onset behavioral rigidity, and progressing language difficulties to single-word and sentence comprehension difficulties. They were classified as tv-FTD. Clinical, cognitive and MRI follow-up were performed. As cognitive impairment progressed, MRI atrophy worsened in ATL and frontotemporal areas in both patients. Both cases had clear family histories of neurological and/or psychiatric disease. Genetic testing revealed a C9orf72 hexanucleotide repeat expansion in both cases. BC passed away after 15 years of disease and autopsy showed the expected TDP-type B pathology. These genetic cases of tv-FTD highlight the susceptibility of ATL to C9orf72-related pathology and emphasize the importance of genetical testing in FTD-spectrum disorders, regardless of the clinical phenotype.
Collapse
Affiliation(s)
- Francesca Caso
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina, 60, 20132, Milan, Italy.,Neurology Unit, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina, 60, 20132, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | | | | | | | - Zachary Miller
- Memory and Aging Center, University of California, San Francisco, CA, USA
| | - Nilo Riva
- Neuropathology Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Renaud La Joie
- Memory and Aging Center, University of California, San Francisco, CA, USA
| | - Giovanni Coppola
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA.,Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Lea T Grinberg
- Memory and Aging Center, University of California, San Francisco, CA, USA
| | - William W Seeley
- Memory and Aging Center, University of California, San Francisco, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, University of California, San Francisco, CA, USA
| | | | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina, 60, 20132, Milan, Italy. .,Neurology Unit, IRCCS Ospedale San Raffaele, Milan, Italy. .,Vita-Salute San Raffaele University, Milan, Italy. .,Neurophysiology Unit, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
10
|
Koriath CAM, Kenny J, Ryan NS, Rohrer JD, Schott JM, Houlden H, Fox NC, Tabrizi SJ, Mead S. Genetic testing in dementia - utility and clinical strategies. Nat Rev Neurol 2021; 17:23-36. [PMID: 33168964 DOI: 10.1038/s41582-020-00416-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2020] [Indexed: 02/07/2023]
Abstract
Techniques for clinical genetic testing in dementia disorders have advanced rapidly but remain to be more widely implemented in practice. A positive genetic test offers a precise molecular diagnosis, can help members of an affected family to determine personal risk, provides a basis for reproductive choices and can offer options for clinical trials. The likelihood of identifying a specific genetic cause of dementia depends on the clinical condition, the age at onset and family history. Attempts to match phenotypes to single genes are mostly inadvisable owing to clinical overlap between the dementias, genetic heterogeneity, pleiotropy and concurrent mutations. Currently, the appropriate genetic test in most cases of dementia is a next-generation sequencing gene panel, though some conditions necessitate specific types of test such as repeat expansion testing. Whole-exome and whole-genome sequencing are becoming financially feasible but raise or exacerbate complex issues such as variants of uncertain significance, secondary findings and the potential for re-analysis in light of new information. However, the capacity for data analysis and counselling is already restricting the provision of genetic testing. Patients and their relatives need to be given reliable information to enable them to make informed choices about tests, treatments and data sharing; the ability of patients with dementia to make decisions must be considered when providing this information.
Collapse
Affiliation(s)
| | - Joanna Kenny
- South West Thames Regional Genetics Service, London, UK
| | - Natalie S Ryan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Jonathan M Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Henry Houlden
- Neurogenetics Laboratory, National Hospital for Neurology and Neurosurgery, London, UK
| | - Nick C Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Simon Mead
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London, UK.
| |
Collapse
|
11
|
Ganguly J, Jog M. Tauopathy and Movement Disorders-Unveiling the Chameleons and Mimics. Front Neurol 2020; 11:599384. [PMID: 33250855 PMCID: PMC7674803 DOI: 10.3389/fneur.2020.599384] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
The spectrum of tauopathy encompasses heterogenous group of neurodegenerative disorders characterized by neural or glial deposition of pathological protein tau. Clinically they can present as cognitive syndromes, movement disorders, motor neuron disease, or mixed. The heterogeneity in clinical presentation, genetic background, and underlying pathology make it difficult to classify and clinically approach tauopathy. In the literature, tauopathies are thus mostly highlighted from pathological perspective. From clinical standpoint, cognitive syndromes are often been focussed while reviewing tauopathies. However, the spectrum of tauopathy has also evolved significantly in the domain of movement disorders and has transgressed beyond the domain of primary tauopathies. Secondary tauopathies from neuroinflammation or autoimmune insults and some other "novel" tauopathies are increasingly being reported in the current literature, while some of them are geographically isolated. Because of the overlapping clinical phenotypes, it often becomes difficult for the clinician to diagnose them clinically and have to wait for the pathological confirmation by autopsy. However, each of these tauopathies has some clinical and radiological signatures those can help in clinical diagnosis and targeted genetic testing. In this review, we have exposed the heterogeneity of tauopathy from a movement disorder perspective and have provided a clinical approach to diagnose them ante mortem before confirmatory autopsy. Additionally, phenotypic variability of these disorders (chameleons) and the look-alikes (mimics) have been discussed with potential clinical pointers for each of them. The review provides a framework within which new and as yet undiscovered entities can be classified in the future.
Collapse
Affiliation(s)
| | - Mandar Jog
- Movement Disorder Centre, London Health Sciences Centre, University of Western Ontario, London, ON, Canada
| |
Collapse
|
12
|
Ducharme S, Dols A, Laforce R, Devenney E, Kumfor F, van den Stock J, Dallaire-Théroux C, Seelaar H, Gossink F, Vijverberg E, Huey E, Vandenbulcke M, Masellis M, Trieu C, Onyike C, Caramelli P, de Souza LC, Santillo A, Waldö ML, Landin-Romero R, Piguet O, Kelso W, Eratne D, Velakoulis D, Ikeda M, Perry D, Pressman P, Boeve B, Vandenberghe R, Mendez M, Azuar C, Levy R, Le Ber I, Baez S, Lerner A, Ellajosyula R, Pasquier F, Galimberti D, Scarpini E, van Swieten J, Hornberger M, Rosen H, Hodges J, Diehl-Schmid J, Pijnenburg Y. Recommendations to distinguish behavioural variant frontotemporal dementia from psychiatric disorders. Brain 2020; 143:1632-1650. [PMID: 32129844 PMCID: PMC7849953 DOI: 10.1093/brain/awaa018] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 11/27/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022] Open
Abstract
The behavioural variant of frontotemporal dementia (bvFTD) is a frequent cause of early-onset dementia. The diagnosis of bvFTD remains challenging because of the limited accuracy of neuroimaging in the early disease stages and the absence of molecular biomarkers, and therefore relies predominantly on clinical assessment. BvFTD shows significant symptomatic overlap with non-degenerative primary psychiatric disorders including major depressive disorder, bipolar disorder, schizophrenia, obsessive-compulsive disorder, autism spectrum disorders and even personality disorders. To date, ∼50% of patients with bvFTD receive a prior psychiatric diagnosis, and average diagnostic delay is up to 5-6 years from symptom onset. It is also not uncommon for patients with primary psychiatric disorders to be wrongly diagnosed with bvFTD. The Neuropsychiatric International Consortium for Frontotemporal Dementia was recently established to determine the current best clinical practice and set up an international collaboration to share a common dataset for future research. The goal of the present paper was to review the existing literature on the diagnosis of bvFTD and its differential diagnosis with primary psychiatric disorders to provide consensus recommendations on the clinical assessment. A systematic literature search with a narrative review was performed to determine all bvFTD-related diagnostic evidence for the following topics: bvFTD history taking, psychiatric assessment, clinical scales, physical and neurological examination, bedside cognitive tests, neuropsychological assessment, social cognition, structural neuroimaging, functional neuroimaging, CSF and genetic testing. For each topic, responsible team members proposed a set of minimal requirements, optimal clinical recommendations, and tools requiring further research or those that should be developed. Recommendations were listed if they reached a ≥ 85% expert consensus based on an online survey among all consortium participants. New recommendations include performing at least one formal social cognition test in the standard neuropsychological battery for bvFTD. We emphasize the importance of 3D-T1 brain MRI with a standardized review protocol including validated visual atrophy rating scales, and to consider volumetric analyses if available. We clarify the role of 18F-fluorodeoxyglucose PET for the exclusion of bvFTD when normal, whereas non-specific regional metabolism abnormalities should not be over-interpreted in the case of a psychiatric differential diagnosis. We highlight the potential role of serum or CSF neurofilament light chain to differentiate bvFTD from primary psychiatric disorders. Finally, based on the increasing literature and clinical experience, the consortium determined that screening for C9orf72 mutation should be performed in all possible/probable bvFTD cases or suspected cases with strong psychiatric features.
Collapse
Affiliation(s)
- Simon Ducharme
- Department of Psychiatry, McGill University Health Centre, McGill University, Montreal, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Str., Montreal, Quebec, H3A 2B4, Canada
| | - Annemiek Dols
- Department of Old Age Psychiatry, GGZ InGeest, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire (CIME), Laval University, Quebec, Canada
| | - Emma Devenney
- Brain and Mind Centre, University of Sydney, Sydney, Australia
| | - Fiona Kumfor
- Brain and Mind Centre, University of Sydney, Sydney, Australia
| | - Jan van den Stock
- Laboratory for Translational Neuropsychiatry, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | | | - Harro Seelaar
- Department of Neurology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Flora Gossink
- Department of Old Age Psychiatry, GGZ InGeest, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Everard Vijverberg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Edward Huey
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Department of Psychiatry, Colombia University, New York, USA
| | - Mathieu Vandenbulcke
- Department of Geriatric Psychiatry, University Hospitals Leuven, Leuven, Belgium
| | - Mario Masellis
- Department of Neurology, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Calvin Trieu
- Department of Old Age Psychiatry, GGZ InGeest, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Chiadi Onyike
- Division of Geriatric Psychiatry and Neuropsychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Paulo Caramelli
- Behavioral and Cognitive Neurology Research Group, Department of Internal Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo Cruz de Souza
- Behavioral and Cognitive Neurology Research Group, Department of Internal Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Maria Landqvist Waldö
- Division of Clinical Sciences Helsingborg, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Olivier Piguet
- Division of Clinical Sciences Helsingborg, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Wendy Kelso
- Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne, Australia
| | - Dhamidhu Eratne
- Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne, Australia
| | - Dennis Velakoulis
- Neuropsychiatry Unit, Royal Melbourne Hospital, Melbourne, Australia
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - David Perry
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, USA
| | - Peter Pressman
- Department of Neurology, University of Colorado Denver, Aurora, USA
| | - Bradley Boeve
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Rik Vandenberghe
- Department of Neurology, University Hospital Leuven, Leuven, Belgium
| | - Mario Mendez
- Department of Neurology, UCLA Medical Centre, University of California Los Angeles, Los Angeles, USA
| | - Carole Azuar
- Department of Neurology, Hôpital La Pitié Salpêtrière, Paris, France
| | - Richard Levy
- Department of Neurology, Hôpital La Pitié Salpêtrière, Paris, France
| | - Isabelle Le Ber
- Department of Neurology, Hôpital La Pitié Salpêtrière, Paris, France
| | - Sandra Baez
- Department of Psychology, Andes University, Bogota, Colombia
| | - Alan Lerner
- Department of Neurology, University Hospital Cleveland Medical Center, Cleveland, USA
| | - Ratnavalli Ellajosyula
- Department of Neurology, Manipal Hospital and Annasawmy Mudaliar Hospital, Bangalore, India
| | - Florence Pasquier
- Univ Lille, Inserm U1171, Memory Center, CHU Lille, DISTAlz, Lille, France
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Centro Dino Ferrari, Milan, Italy
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, Neurodegenerative Diseases Unit Milan, Italy
| | - Elio Scarpini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Centro Dino Ferrari, Milan, Italy
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, Neurodegenerative Diseases Unit Milan, Italy
| | - John van Swieten
- Department of Neurology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Howard Rosen
- Memory and Aging Center, University of California San Francisco, San Francisco, USA
| | - John Hodges
- Brain and Mind Centre, University of Sydney, Sydney, Australia
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Technical University of Munich, School of Medicine, Munich, Germany
| | - Yolande Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Heuer HW, Wang P, Rascovsky K, Wolf A, Appleby B, Bove J, Bordelon Y, Brannelly P, Brushaber DE, Caso C, Coppola G, Dickerson B, Dickinson S, Domoto-Reilly K, Faber K, Ferrall J, Fields J, Fishman A, Fong J, Foroud T, Forsberg LK, Gearhart D, Ghazanfari B, Ghoshal N, Goldman J, Graff-Radford J, Graff-Radford N, Grant I, Grossman M, Haley D, Hsiung GY, Huey E, Irwin D, Jones D, Kantarci K, Karydas A, Kaufer D, Kerwin D, Knopman D, Kornak J, Kramer JH, Kraft R, Kremers WK, Kukull W, Litvan I, Ljubenkov P, Mackenzie IR, Maldonado M, Manoochehri M, McGinnis S, McKinley E, Mendez MF, Miller BL, Onyike C, Pantelyat A, Pearlman R, Petrucelli L, Potter M, Rademakers R, Ramos EM, Rankin KP, Roberson ED, Rogalski E, Sengdy P, Shaw L, Syrjanen J, Tartaglia MC, Tatton N, Taylor J, Toga A, Trojanowski J, Weintraub S, Wong B, Wszolek Z, Boeve BF, Rosen HJ, Boxer AL. Comparison of sporadic and familial behavioral variant frontotemporal dementia (FTD) in a North American cohort. Alzheimers Dement 2020; 16:60-70. [PMID: 31914226 PMCID: PMC7192555 DOI: 10.1002/alz.12046] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Behavioral variant frontotemporal dementia (bvFTD) may present sporadically or due to an autosomal dominant mutation. Characterization of both forms will improve understanding of the generalizability of assessments and treatments. METHODS A total of 135 sporadic (s-bvFTD; mean age 63.3 years; 34% female) and 99 familial (f-bvFTD; mean age 59.9; 48% female) bvFTD participants were identified. f-bvFTD cases included 43 with known or presumed chromosome 9 open reading frame 72 (C9orf72) gene expansions, 28 with known or presumed microtubule-associated protein tau (MAPT) mutations, 14 with known progranulin (GRN) mutations, and 14 with a strong family history of FTD but no identified mutation. RESULTS Participants with f-bvFTD were younger and had earlier age at onset. s-bvFTD had higher total Neuropsychiatric Inventory Questionnaire (NPI-Q) scores due to more frequent endorsement of depression and irritability. DISCUSSION f-bvFTD and s-bvFTD cases are clinically similar, suggesting the generalizability of novel biomarkers, therapies, and clinical tools developed in either form to the other.
Collapse
Affiliation(s)
- Hilary W Heuer
- University of California, San Francisco, San Francisco, California
| | - P Wang
- University of California, San Francisco, San Francisco, California
| | - K Rascovsky
- University of Pennsylvania, Philadelphia, Pennsylvania
| | - A Wolf
- University of California, San Francisco, San Francisco, California
| | - B Appleby
- Case Western Reserve University, Cleveland, Ohio
| | - J Bove
- University of Pennsylvania, Philadelphia, Pennsylvania
| | - Y Bordelon
- University of California, Los Angeles, Los Angeles, California
| | - P Brannelly
- Tau Consortium, Rainwater Charitable Foundation, Fort Worth, Texas
| | | | - C Caso
- U Washington, Seattle, Washington
| | - G Coppola
- University of California, Los Angeles, Los Angeles, California
| | - B Dickerson
- Harvard University/MGH, Boston, Massachusetts
| | - S Dickinson
- Association for Frontotemporal Degeneration, Radnor, Pennsylvania
| | | | - K Faber
- National Centralized Repository for Alzheimer's Disease and Related Disorders (NCRAD), Indiana University, Indianapolis, Indiana
| | - J Ferrall
- University of North Carolina, Chapel Hill, North Carolina
| | - J Fields
- Mayo Clinic, Rochester, Minnesota
| | - A Fishman
- Johns Hopkins University, Baltimore, Maryland
| | - J Fong
- University of California, San Francisco, San Francisco, California
| | - T Foroud
- National Centralized Repository for Alzheimer's Disease and Related Disorders (NCRAD), Indiana University, Indianapolis, Indiana
| | | | | | | | - N Ghoshal
- Washington University, St. Louis, Missouri
| | - J Goldman
- Columbia University, New York, New York
| | | | | | - I Grant
- Northwestern University, Chicago, Illinois
| | - M Grossman
- University of Pennsylvania, Philadelphia, Pennsylvania
| | - D Haley
- Mayo Clinic, Jacksonville, Florida
| | - G-Y Hsiung
- University of British Columbia, Vancouver, British Columbia, Canada
| | - E Huey
- Columbia University, New York, New York
| | - D Irwin
- University of Pennsylvania, Philadelphia, Pennsylvania
| | - D Jones
- Mayo Clinic, Rochester, Minnesota
| | | | - A Karydas
- University of California, San Francisco, San Francisco, California
| | - D Kaufer
- University of North Carolina, Chapel Hill, North Carolina
| | - D Kerwin
- The University of Texas, Southwestern Medical Center at Dallas, Dallas, Texas
| | | | - J Kornak
- University of California, San Francisco, San Francisco, California
| | - J H Kramer
- University of California, San Francisco, San Francisco, California
| | - R Kraft
- Mayo Clinic, Rochester, Minnesota
| | | | - W Kukull
- National Alzheimer Coordinating Center (NACC), University of Washington, Seattle, Washington
| | - I Litvan
- University of California, San Diego, San Diego, California
| | - P Ljubenkov
- University of California, San Francisco, San Francisco, California
| | - I R Mackenzie
- University of British Columbia, Vancouver, British Columbia, Canada
| | - M Maldonado
- University of California, Los Angeles, Los Angeles, California
| | | | - S McGinnis
- Harvard University/MGH, Boston, Massachusetts
| | - E McKinley
- University of Alabama at Birmingham, Birmingham, Alabama
| | - M F Mendez
- University of California, Los Angeles, Los Angeles, California
| | - B L Miller
- University of California, San Francisco, San Francisco, California
| | - C Onyike
- Johns Hopkins University, Baltimore, Maryland
| | - A Pantelyat
- Johns Hopkins University, Baltimore, Maryland
| | - R Pearlman
- Bluefield Project, San Francisco, California
| | | | - M Potter
- National Centralized Repository for Alzheimer's Disease and Related Disorders (NCRAD), Indiana University, Indianapolis, Indiana
| | | | - E M Ramos
- University of California, Los Angeles, Los Angeles, California
| | - K P Rankin
- University of California, San Francisco, San Francisco, California
| | - E D Roberson
- University of Alabama at Birmingham, Birmingham, Alabama
| | - E Rogalski
- Northwestern University, Chicago, Illinois
| | - P Sengdy
- University of British Columbia, Vancouver, British Columbia, Canada
| | - L Shaw
- University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - N Tatton
- Association for Frontotemporal Degeneration, Radnor, Pennsylvania
| | - J Taylor
- University of California, San Francisco, San Francisco, California
| | - A Toga
- Laboratory of Neuroimaging (LONI), USC, Los Angeles, California
| | | | | | - B Wong
- Harvard University/MGH, Boston, Massachusetts
| | | | | | - H J Rosen
- University of California, San Francisco, San Francisco, California
| | - A L Boxer
- University of California, San Francisco, San Francisco, California
| |
Collapse
|
14
|
Fedorova YB. [Current approaches to the treatment of fronto-temporal dementia]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:18-24. [PMID: 31825385 DOI: 10.17116/jnevro201911909218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Frontotemporal dementias (FTD) are one of the prevalent forms of early neurodegenerative diseases. FTD are characterized by heterogeneous clinical manifestations and syndromes. The current methods of FTD treatment and the clinical trials of new methods of FTD treatment are considered in the article. Biomarkers and their relationships with the results of recently completed clinical trials, as well as future therapeutic perspectives, are reviewed.
Collapse
|
15
|
Gossye H, Van Broeckhoven C, Engelborghs S. The Use of Biomarkers and Genetic Screening to Diagnose Frontotemporal Dementia: Evidence and Clinical Implications. Front Neurosci 2019; 13:757. [PMID: 31447625 PMCID: PMC6691066 DOI: 10.3389/fnins.2019.00757] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
Within the wide range of neurodegenerative brain diseases, the differential diagnosis of frontotemporal dementia (FTD) frequently poses a challenge. Often, signs and symptoms are not characteristic of the disease and may instead reflect atypical presentations. Consequently, the use of disease biomarkers is of importance to correctly identify the patients. Here, we describe how neuropsychological characteristics, neuroimaging and neurochemical biomarkers and screening for causal gene mutations can be used to differentiate FTD from other neurodegenerative diseases as well as to distinguish between FTD subtypes. Summarizing current evidence, we propose a stepwise approach in the diagnostic evaluation. Clinical consensus criteria that take into account a full neuropsychological examination have relatively good accuracy (sensitivity [se] 75–95%, specificity [sp] 82–95%) to diagnose FTD, although misdiagnosis (mostly AD) is common. Structural brain MRI (se 70–94%, sp 89–99%) and FDG PET (se 47–90%, sp 68–98%) or SPECT (se 36–100%, sp 41–100%) brain scans greatly increase diagnostic accuracy, showing greater involvement of frontal and anterior temporal lobes, with sparing of hippocampi and medial temporal lobes. If these results are inconclusive, we suggest detecting amyloid and tau cerebrospinal fluid (CSF) biomarkers that can indicate the presence of AD with good accuracy (se 74–100%, sp 82–97%). The use of P-tau181 and the Aβ1–42/Aβ1–40 ratio significantly increases the accuracy of correctly identifying FTD vs. AD. Alternatively, an amyloid brain PET scan can be performed to differentiate FTD from AD. When autosomal dominant inheritance is suspected, or in early onset dementia, mutation screening of causal genes is indicated and may also be offered to at-risk family members. We have summarized genotype–phenotype correlations for several genes that are known to cause familial frontotemporal lobar degeneration, which is the neuropathological substrate of FTD. The genes most commonly associated with this disease (C9orf72, MAPT, GRN, TBK1) are discussed, as well as some less frequent ones (CHMP2B, VCP). Several other techniques, such as diffusion tensor imaging, tau PET imaging and measuring serum neurofilament levels, show promise for future implementation as diagnostic biomarkers.
Collapse
Affiliation(s)
- Helena Gossye
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium.,Institute Born - Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Center for Neurosciences, UZ Brussel and Vrije Universiteit Brussel, Brussels, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Antwerp, Belgium.,Institute Born - Bunge, University of Antwerp, Antwerp, Belgium
| | - Sebastiaan Engelborghs
- Institute Born - Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Center for Neurosciences, UZ Brussel and Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
16
|
O'Malley M, Parkes J, Stamou V, LaFontaine J, Oyebode J, Carter J. Young-onset dementia: scoping review of key pointers to diagnostic accuracy. BJPsych Open 2019; 5:e48. [PMID: 31530311 PMCID: PMC6582217 DOI: 10.1192/bjo.2019.36] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/30/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Routine psychiatric assessments tailored to older patients are often insufficient to identify the complexity of presentation in younger patients with dementia. Significant overlap between psychiatric disorders and neurodegenerative disease means that high rates of prior incorrect psychiatric diagnosis are common. Long delays to diagnosis, misdiagnosis and lack of knowledge from professionals are key concerns. No specific practice guidelines exist for diagnosis of young-onset dementia (YOD). AIMS The review evaluates the current evidence about best practice in diagnosis to guide thorough assessment of the complex presentations of YOD with a view to upskilling professionals in the field. METHOD A comprehensive search of the literature adopting a scoping review methodology was conducted regarding essential elements of diagnosis in YOD, over and above those in current diagnostic criteria for disease subtypes. This methodology was chosen because research in this area is sparse and not amenable to a traditional systematic review. RESULTS The quality of evidence identified is variable with the majority provided from expert opinion and evidence is lacking on some topics. Evidence appears weighted towards diagnosis in frontotemporal dementia and its subtypes and young-onset Alzheimer's disease. CONCLUSIONS The literature demonstrates that a clinically rigorous and systematic approach is necessary in order to avoid mis- or underdiagnosis for younger people. The advent of new disease-modifying treatments necessitates clinicians in the field to improve knowledge of new imaging techniques and genetics, with the goal of improving training and practice, and highlights the need for quality indicators and alignment of diagnostic procedures across clinical settings. DECLARATION OF INTEREST None.
Collapse
Affiliation(s)
- Mary O'Malley
- Research Assistant, Faculty of Health and Society, University of Northampton, UK
| | - Jacqueline Parkes
- Professor, Faculty of Health and Society, University of Northampton, UK
| | - Vasileios Stamou
- Research Assistant, Centre for Applied Dementia Studies, University of Bradford, UK
| | - Jenny LaFontaine
- Research Fellow, Centre for Applied Dementia Studies, University of Bradford, UK
| | - Jan Oyebode
- Centre for Applied Dementia Studies, University of Bradford, UK
| | - Janet Carter
- Assistant Professor, Division of Psychiatry, University College London, UK
| |
Collapse
|
17
|
Genetic mimics of the non-genetic atypical parkinsonian disorders – the ‘atypical’ atypical. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 149:327-351. [DOI: 10.1016/bs.irn.2019.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
Borrego-Écija S, Morgado J, Palencia-Madrid L, Grau-Rivera O, Reñé R, Hernández I, Almenar C, Balasa M, Antonell A, Molinuevo JL, Lladó A, Martínez de Pancorbo M, Gelpi E, Sánchez-Valle R. Frontotemporal Dementia Caused by the P301L Mutation in the MAPT Gene: Clinicopathological Features of 13 Cases from the Same Geographical Origin in Barcelona, Spain. Dement Geriatr Cogn Disord 2018; 44:213-221. [PMID: 28934750 DOI: 10.1159/000480077] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/04/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/AIMS We identified and studied 13 patients carrying the P301L mutation in the MAPT gene from the same area (Baix Llobregat County) in Barcelona, Spain. METHODS The demographic and clinical features were reviewed retrospectively. Detailed neuropathological characterization was obtained in 9 subjects. To investigate the origin of the P301L mutation in these families, 20 single nucleotide polymorphisms (SNPs) in the MAPT gene were analyzed. RESULTS The mean age at disease onset was 51 years and the mean disease duration was 7 years. The most common initial symptoms were behavioral changes (54%), followed by language disturbances (31%) and memory loss (15%). 46% developed parkinsonism. Neuropathology showed an extensive neuronal and glial 4-repeat (4R) tauopathy with "mini-Pick"-like bodies in the dentate gyrus as the characteristic underlying pathology in all cases. In 1 subject, additional 4R globular glial inclusions were observed. All the mutation carriers showed the same haplotype for the SNPs analyzed, suggesting a common ancestor. CONCLUSION These findings suggest a relative homogeneous clinicopathological phenotype in P301L MAPT mutation carriers in our series. This phenotype might help in the differential diagnosis from other tauopathies and be a morphological hint for genetic testing. The haplotype analysis results suggest a founder effect of the P301L mutation in this area.
Collapse
Affiliation(s)
- Sergi Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Department, Hospital Clínic-Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Mandić-Stojmenović G, Kostić V, Stefanova E. The clinical spectrum of frontotemporal dementia. MEDICINSKI PODMLADAK 2018. [DOI: 10.5937/mp69-16325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
20
|
Bocchetta M, Mega A, Bernardi L, Di Maria E, Benussi L, Binetti G, Borroni B, Colao R, Di Fede G, Fostinelli S, Galimberti D, Gennarelli M, Ghidoni R, Piaceri I, Pievani M, Porteri C, Redaelli V, Rossi G, Suardi S, Babiloni C, Scarpini E, Tagliavini F, Padovani A, Nacmias B, Sorbi S, Frisoni GB, Bruni AC. Genetic Counseling and Testing for Alzheimer's Disease and Frontotemporal Lobar Degeneration: An Italian Consensus Protocol. J Alzheimers Dis 2016; 51:277-91. [PMID: 26901402 DOI: 10.3233/jad-150849] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Genetic testing of familial Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD) is attracting interest thanks to innovative primary prevention clinical trials and increased request for information by at-risk individuals. However, ethical, social, and psychological implications are paramount and genetic testing must be supported by structured genetic counseling. In Italy, practice parameters and guidelines for genetic counseling in dementia are not available. OBJECTIVE To develop a nationally harmonized protocol for genetic counseling and testing of familial AD and FTLD. METHODS Activities were carried out in the context of the Italian Dominantly Inherited Alzheimer's and Frontotemporal Network (IT-DIAfN) project, a national network of centers of excellence with expertise in managing patients with familial AD and FTLD. A survey of the literature on genetic counseling protocols and guidelines was conducted. Local protocols for genetic counseling were surveyed. Differences and commonalities among protocols were identified and discussed among project partners. Consensus was reached following implicit aggregation methods. RESULTS Consensus was reached on a protocol for patients with clinically diagnosed familial AD or FTLD and a distinct protocol for their at-risk relatives. Genetic counseling should be provided by a multidisciplinary team including a geneticist, a neurologist/geriatrician, and a psychologist/psychiatrist, according to the following schedule: (i) initial consultation with tailored information on the genetics of the dementias; (ii) clinical, psychological, and cognitive assessment; if deemed appropriate (iii) genetic testing following a structured decision tree for gene mutation search; (iv) genetic testing result disclosure; (v) psychological support follow-up. CONCLUSION This genetic counseling protocol provides Italian centers with a line of shared practice for dealing with the requests for genetic testing for familial AD and FTLD from patients and at-risk relatives, who may also be eligible participants for novel prevention clinical trials.
Collapse
Affiliation(s)
- Martina Bocchetta
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Anna Mega
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Livia Bernardi
- Centro Regionale di Neurogenetica, ASP Catanzaro, Lamezia terme (CZ) Italy
| | - Emilio Di Maria
- Department of Health Sciences, University of Genova and Division of Medical Genetics, Galliera Hospital, Genova, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Giuliano Binetti
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Barbara Borroni
- University of Brescia and Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Brescia, Brescia, Italy
| | - Rosanna Colao
- Centro Regionale di Neurogenetica, ASP Catanzaro, Lamezia terme (CZ) Italy
| | | | - Silvia Fostinelli
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Daniela Galimberti
- University of Milan, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Gennarelli
- Genetic Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Irene Piaceri
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Michela Pievani
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Corinna Porteri
- Bioethics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | | | - Giacomina Rossi
- IRCCS Fondazione Istituto Neurologico Carlo Besta, Milan, Italy
| | - Silvia Suardi
- IRCCS Fondazione Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudio Babiloni
- Departiment of Physiology and Pharmacology, University of Rome "La Sapienza", Rome, Italy; IRCCS San Raffaele Pisana of Rome, Italy
| | - Elio Scarpini
- University of Milan, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Alessandro Padovani
- University of Brescia and Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Brescia, Brescia, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Giovanni B Frisoni
- Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Memory Clinic and LANVIE - Laboratory of Neuroimaging of Aging, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Amalia C Bruni
- Centro Regionale di Neurogenetica, ASP Catanzaro, Lamezia terme (CZ) Italy
| | | |
Collapse
|
21
|
Fernández Suarez M, Surace E, Harris P, Tapajoz F, Sevlever G, Allegri R, Russo GN. C9ORF72 G4C2-repeat expansion and frontotemporal dementia first reported case in Argentina. Neurocase 2016; 22:281-4. [PMID: 27327087 DOI: 10.1080/13554794.2016.1186700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We present a female patient aged 51 who developed behavioral disorders followed by cognitive impairment over 3 years. Neuropsychological, neuropsychiatric, and radiological features suggested a probable behavioral variant of frontotemporal dementia (bvFTD). A family history of amyotrophic lateral sclerosis and parkinsonism suggested the hexanucleotide repeat expansion G4C2 in C9ORF72 . We set up a two-step genotyping algorithm for the detection of the expansion using fragment-length analysis polymerase chain reaction (PCR) and repeat-primed PCR with fluorescent primers. We confirmed the presence of an expanded G4C2 allele in the patient. This represents the first documented case of bvFTD due to a C9ORF72 expansion in Argentina.
Collapse
Affiliation(s)
- M Fernández Suarez
- a Neurology and Neurosurgery Unit , Swiss Medical, Sanatorio de los Arcos , Buenos Aires , Argentina
| | - Ezequiel Surace
- b Neuropathology and Molecular Biology Department , Instituto de Investigaciones Neurológica "Raúl Carrea" (FLENI) , Buenos Aires , Argentina.,c Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Argentina
| | - P Harris
- c Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Argentina.,d Cognitive and Neuropsychology Unit , Instituto de Investigaciones Neurológica "Raúl Carrea" (FLENI) , Buenos Aires , Argentina
| | - F Tapajoz
- c Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Argentina.,d Cognitive and Neuropsychology Unit , Instituto de Investigaciones Neurológica "Raúl Carrea" (FLENI) , Buenos Aires , Argentina
| | - G Sevlever
- b Neuropathology and Molecular Biology Department , Instituto de Investigaciones Neurológica "Raúl Carrea" (FLENI) , Buenos Aires , Argentina
| | - R Allegri
- c Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) , Argentina.,d Cognitive and Neuropsychology Unit , Instituto de Investigaciones Neurológica "Raúl Carrea" (FLENI) , Buenos Aires , Argentina
| | - G N Russo
- a Neurology and Neurosurgery Unit , Swiss Medical, Sanatorio de los Arcos , Buenos Aires , Argentina
| |
Collapse
|
22
|
Karch CM, Ezerskiy L, Redaelli V, Giovagnoli AR, Tiraboschi P, Pelliccioni G, Pelliccioni P, Kapetis D, D'Amato I, Piccoli E, Ferretti MG, Tagliavini F, Rossi G. Missense mutations in progranulin gene associated with frontotemporal lobar degeneration: study of pathogenetic features. Neurobiol Aging 2016; 38:215.e1-215.e12. [PMID: 26652843 PMCID: PMC4738142 DOI: 10.1016/j.neurobiolaging.2015.10.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/18/2015] [Accepted: 10/28/2015] [Indexed: 10/22/2022]
Abstract
GRN, the gene coding for the progranulin (PGRN) protein, was recognized as a gene linked to frontotemporal lobar degeneration (FTLD). The first mutations identified were null mutations giving rise to haploinsufficiency. Missense mutations were subsequently detected, but only a small subset has been functionally investigated. We identified missense mutations (C105Y, A199V, and R298H) in FTLD cases with family history and/or with low plasma PGRN levels. The aim of this study was to determine their pathogenicity. We performed functional studies, analyzing PGRN expression, secretion, and cleavage by elastase. GRN C105Y affected both secretion and elastase cleavage, likely representing a pathogenic mutation. GRN A199V did not alter the physiological properties of PGRN and GRN R298H produced only moderate effects on PGRN secretion, indicating that their pathogenicity is uncertain. In the absence of strong segregation data and neuropathological examinations, genetic, biomarker, and functional studies can be applied to an algorithm to assess the likelihood of pathogenicity for a mutation. This information can improve our understanding of the complex mechanisms by which GRN mutations lead to FTLD.
Collapse
Affiliation(s)
- Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA; Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University School of Medicine, St Louis, MO, USA
| | - Lubov Ezerskiy
- Department of Psychiatry, Washington University School of Medicine, St Louis, MO, USA
| | - Veronica Redaelli
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Anna Rita Giovagnoli
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Pietro Tiraboschi
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | | | - Paolo Pelliccioni
- Division of Neurology, Geriatric Hospital, INRCA-IRCCS, Ancona, Italy
| | - Dimos Kapetis
- Bioinformatics Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Ilaria D'Amato
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Elena Piccoli
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Maria Giulia Ferretti
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Fabrizio Tagliavini
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giacomina Rossi
- Division of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy.
| |
Collapse
|
23
|
Abstract
Frontotemporal dementia (FTD) was one of the lesser known dementias until the recent advancements revealing its genetic and pathological foundation. This common neurodegenerative disorder has three clinical subtypes- behavioral, semantic and progressive non fluent aphasia. The behavioral variant mostly exhibits personality changes, while the other two encompass various language deficits. This review discusses the basic pathology, genetics, clinical and histological presentation and the diagnosis of the 3 subtypes. It also deliberates the different therapeutic modalities currently available for frontotemporal dementia and the challenges faced by the caregivers. Lastly it explores the scope of further research into the diagnosis and management of FTD.
Collapse
Affiliation(s)
- Sayantani Ghosh
- Department of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Carol F Lippa
- Department of Neurology, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
24
|
Abstract
Frontotemporal dementia is an umbrella clinical term that encompasses a group of neurodegenerative diseases characterised by progressive deficits in behaviour, executive function, or language. Frontotemporal dementia is a common type of dementia, particularly in patients younger than 65 years. The disease can mimic many psychiatric disorders because of the prominent behavioural features. Various underlying neuropathological entities lead to the frontotemporal dementia clinical phenotype, all of which are characterised by the selective degeneration of the frontal and temporal cortices. Genetics is an important risk factor for frontotemporal dementia. Advances in clinical, imaging, and molecular characterisation have increased the accuracy of frontotemporal dementia diagnosis, thus allowing for the accurate differentiation of these syndromes from psychiatric disorders. As the understanding of the molecular basis for frontotemporal dementia improves, rational therapies are beginning to emerge.
Collapse
Affiliation(s)
- Jee Bang
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Salvatore Spina
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Memory and Aging Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
25
|
Jarmolowicz AI, Chen HY, Panegyres PK. The patterns of inheritance in early-onset dementia: Alzheimer's disease and frontotemporal dementia. Am J Alzheimers Dis Other Demen 2015; 30:299-306. [PMID: 25147204 PMCID: PMC10852564 DOI: 10.1177/1533317514545825] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AIM To investigate the patterns of inheritance and gene mutation status in early-onset dementia (EOD). METHODS Data were collected on 202 consecutive patients presenting to an EOD clinic. Early-onset Alzheimer's disease (EOAD, n = 120) and early-onset frontotemporal dementia (EOFTD, n = 82) were studied. RESULTS The majority of participants, 72.5% with EOAD and 74.4% with EOFTD, did not have a positive family history of dementia. An autosomal dominant pattern of inheritance was observed in 14.2% of patients with EOAD and 13.4% of patients with FTD. Of those with an autosomal dominant pattern of inheritance, 11.8% of EOAD and 45.5% of FTD probands had known pathogenic mutations. Only 1.6% of the total population of EOAD and 7.3% of EOFTD possessed known gene mutations. CONCLUSION Early-onset dementia does not appear to be a strongly inherited autosomal dominant condition. The majority of patients were sporadic. Known mutations were uncommon and do not explain the total autosomal dominant burden.
Collapse
Affiliation(s)
- Anna I Jarmolowicz
- Neurodegenerative Disorders Research Pty Ltd, Subiaco, West Perth, Australia
| | - Huei-Yang Chen
- Neurodegenerative Disorders Research Pty Ltd, Subiaco, West Perth, Australia
| | - Peter K Panegyres
- Neurodegenerative Disorders Research Pty Ltd, Subiaco, West Perth, Australia
| |
Collapse
|
26
|
Miller BL, Dickerson BC, Lucente DE, Larvie M, Frosch MP. Case records of the Massachusetts General Hospital. Case 9-2015. A 31-year-old man with personality changes and progressive neurologic decline. N Engl J Med 2015; 372:1151-62. [PMID: 25785973 PMCID: PMC4795456 DOI: 10.1056/nejmcpc1409839] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
27
|
Clerc MT, Deprez M, Leuba G, Lhermitte B, Lopez U, von Gunten A. Atypical association of semantic dementia, corticobasal syndrome, and 4R tauopathy. Neurocase 2015; 21:1-15. [PMID: 24156410 DOI: 10.1080/13554794.2013.841953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
A 57-year-old male with no family history was diagnosed with semantic dementia. He also showed some unusual cognitive features such as episodic memory and executive dysfunctions, spatial disorientation, and dyscalculia. Rapidly progressive cognitive and physical decline occurred. About 1.5 years later, he developed clinical features of a corticobasal syndrome. He died at the age of 60. Brain autopsy revealed numerous 4R-tau-positive lesions in the frontal, parietal and temporal lobes, basal ganglia, and brainstem. Neuronal loss was severe in the temporal cortex. Such association of semantic dementia with tauopathy and corticobasal syndrome is highly unusual. These findings are discussed in the light of current knowledge about frontotemporal lobar degeneration.
Collapse
Affiliation(s)
- Marie-Therese Clerc
- a Service Universitaire de Psychiatrie de l'âge avancé, Département de Psychiatrie , Centre Hospitalier Universitaire Vaudois , Lausanne , Switzerland
| | | | | | | | | | | |
Collapse
|
28
|
Siuda J, Fujioka S, Wszolek ZK. Parkinsonian syndrome in familial frontotemporal dementia. Parkinsonism Relat Disord 2014; 20:957-64. [PMID: 24998994 DOI: 10.1016/j.parkreldis.2014.06.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/27/2014] [Accepted: 06/06/2014] [Indexed: 12/12/2022]
Abstract
Parkinsonism in frontotemporal dementia (FTD) was first described in families with mutations in the microtubule-associated protein tau (MAPT) and progranulin (PRGN) genes. Since then, mutations in several other genes have been identified for FTD with parkinsonism, including chromosome 9 open reading frame 72 (C9ORF72), chromatin modifying protein 2B (CHMP2B), valosin-containing protein (VCP), fused in sarcoma (FUS) and transactive DNA-binding protein (TARDBP). The clinical presentation of patients with familial forms of FTD with parkinsonism is highly variable. The parkinsonism seen in FTD patients is usually characterized by akinetic-rigid syndrome and is mostly associated with the behavioral variant of FTD (bvFTD); however, some cases may present with classical Parkinson's disease. In other cases, atypical parkinsonism resembling progressive supranuclear palsy (PSP) or corticobasal syndrome (CBS) has also been described. Although rare, parkinsonism in FTD may coexist with motor neuron disease. Structural neuroimaging, which is crucial for the diagnosis of FTD, shows characteristic patterns of brain atrophy associated with specific mutations. Structural neuroimaging is not helpful in distinguishing among patients with parkinsonian features. Furthermore, dopaminergic imaging that shows nigrostriatal neurodegeneration in FTD with parkinsonism cannot discriminate parkinsonian syndromes that arise from different mutations. Generally, parkinsonism in FTD is levodopa unresponsive, but there have been cases where a temporary benefit has been reported, so dopaminergic treatment is worth trying, especially, when motor and non-motor manifestations can cause significant problems with daily functioning. In this review, we present an update on the clinical and genetic correlations of FTD with parkinsonism.
Collapse
Affiliation(s)
- Joanna Siuda
- Department of Neurology, Silesian Medical University, Katowice, Poland; Department of Neuroscience, Mayo Clinic Jacksonville, FL, USA
| | | | | |
Collapse
|
29
|
Pressman PS, Miller BL. Diagnosis and management of behavioral variant frontotemporal dementia. Biol Psychiatry 2014; 75:574-81. [PMID: 24315411 PMCID: PMC4194080 DOI: 10.1016/j.biopsych.2013.11.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 10/31/2013] [Accepted: 11/01/2013] [Indexed: 12/11/2022]
Abstract
Frontotemporal dementia was documented over a century ago. The last decade, however, has seen substantial changes in our conceptions of this increasingly recognized disorder. Different clinical variants have been delineated, the most common of which is the behavioral variant (bvFTD). Updated diagnostic criteria have been established. New histopathological findings and genetic etiologies have been discovered. Research continues to uncover molecular mechanisms by which abnormal proteins accumulate in degenerating brain tissue. Novel neuroimaging techniques suggest that functional networks are diminished in bvFTD that might be relevant to empathy and social behavior. Despite rapid advances in our understanding of bvFTD, the disease is still under-recognized and commonly misdiagnosed. The result is inappropriate patient care. Recognizing the various presentations of bvFTD and its histological and genetic subtypes might further diagnosis, treatment, and research.
Collapse
Affiliation(s)
- Peter S Pressman
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, San Francisco, California.
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, San Francisco, California
| |
Collapse
|
30
|
Abstract
25% of all people aged 55 years and older have a family history of dementia. For most, the family history is due to genetically complex disease, where many genetic variations of small effect interact to increase risk of dementia. The lifetime risk of dementia for these families is about 20%, compared with 10% in the general population. A small proportion of families have an autosomal dominant family history of early-onset dementia, which is often due to mendelian disease, caused by a mutation in one of the dementia genes. Each family member has a 50% chance of inheriting the mutation, which confers a lifetime dementia risk of over 95%. In this Review, we focus on the evidence for, and the approach to, genetic testing in Alzheimer's disease (APP, PSEN1, and PSEN2 genes), frontotemporal dementia (MAPT, GRN, C9ORF72, and other genes), and other familial dementias. We conclude by discussing the practical aspects of genetic counselling.
Collapse
Affiliation(s)
- Clement T Loy
- School of Public Health, University of Sydney, Sydney, NSW, Australia; Neuroscience Research Australia, Randwick, NSW, Australia; Huntington Disease Service, Westmead Hospital, Westmead, NSW, Australia
| | - Peter R Schofield
- Neuroscience Research Australia, Randwick, NSW, Australia; University of New South Wales, Kensington, NSW, Australia
| | - Anne M Turner
- Department of Medical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia
| | - John B J Kwok
- Neuroscience Research Australia, Randwick, NSW, Australia; University of New South Wales, Kensington, NSW, Australia.
| |
Collapse
|
31
|
Abstract
Over the past decade, there has been a dramatic evolution of genetic methodologies that can be used to identify genes contributing to disease. Initially, the focus was primarily on classical linkage analysis; more recently, genomewide association studies, and high-throughput whole genome and whole exome sequencing have provided efficient approaches to detect common and rare variation contributing to disease risk. Application of these methodologies to dementias has led to the nomination of dozens of causative and susceptibility genes, solidifying the recognition that genetic factors are important contributors to the disease processes. In this review, the authors focus on current knowledge of the genetics of Alzheimer's disease and frontotemporal lobar degeneration. A working understanding of the genes relevant to common dementias will become increasingly critical, as options for genetic testing and eventually gene-specific therapeutics are developed.
Collapse
Affiliation(s)
- Janice L Farlow
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana
| | | |
Collapse
|
32
|
Paulsen JS, Nance M, Kim JI, Carlozzi NE, Panegyres PK, Erwin C, Goh A, McCusker E, Williams JK. A review of quality of life after predictive testing for and earlier identification of neurodegenerative diseases. Prog Neurobiol 2013; 110:2-28. [PMID: 24036231 PMCID: PMC3833259 DOI: 10.1016/j.pneurobio.2013.08.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 08/26/2013] [Accepted: 08/26/2013] [Indexed: 12/12/2022]
Abstract
The past decade has witnessed an explosion of evidence suggesting that many neurodegenerative diseases can be detected years, if not decades, earlier than previously thought. To date, these scientific advances have not provoked any parallel translational or clinical improvements. There is an urgency to capitalize on this momentum so earlier detection of disease can be more readily translated into improved health-related quality of life for families at risk for, or suffering with, neurodegenerative diseases. In this review, we discuss health-related quality of life (HRQOL) measurement in neurodegenerative diseases and the importance of these "patient reported outcomes" for all clinical research. Next, we address HRQOL following early identification or predictive genetic testing in some neurodegenerative diseases: Huntington disease, Alzheimer's disease, Parkinson's disease, Dementia with Lewy bodies, frontotemporal dementia, amyotrophic lateral sclerosis, prion diseases, hereditary ataxias, Dentatorubral-pallidoluysian atrophy and Wilson's disease. After a brief report of available direct-to-consumer genetic tests, we address the juxtaposition of earlier disease identification with assumed reluctance toward predictive genetic testing. Forty-one studies examining health-related outcomes following predictive genetic testing for neurodegenerative disease suggested that (a) extreme or catastrophic outcomes are rare; (b) consequences commonly include transiently increased anxiety and/or depression; (c) most participants report no regret; (d) many persons report extensive benefits to receiving genetic information; and (e) stigmatization and discrimination for genetic diseases are poorly understood and policy and laws are needed. Caution is appropriate for earlier identification of neurodegenerative diseases but findings suggest further progress is safe, feasible and likely to advance clinical care.
Collapse
Affiliation(s)
- Jane S Paulsen
- Department of Neurology, University of Iowa, Carver College of Medicine, Iowa City, IA, USA; Department of Psychiatry, University of Iowa, Carver College of Medicine, Iowa City, IA, USA; Department of Psychology, University of Iowa, Iowa City, IA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Gil-Navarro S, Lladó A, Rami L, Castellví M, Bosch B, Bargalló N, Lomeña F, Reñé R, Montagut N, Antonell A, Molinuevo JL, Sánchez-Valle R. Neuroimaging and biochemical markers in the three variants of primary progressive aphasia. Dement Geriatr Cogn Disord 2013; 35:106-17. [PMID: 23392204 DOI: 10.1159/000346289] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2012] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND/AIM To investigate in variants of primary progressive aphasia (PPA) the association between current clinical and neuroimaging criteria and biochemical/genetic markers at the individual level. METHODS Thirty-two PPA patients were classified as non-fluent/agrammatic (nfvPPA), semantic (svPPA), or logopenic variant (lvPPA) or as unclassifiable (uPPA). In all patients, we evaluated the neuroimaging criteria (magnetic resonance imaging and/or single photon emission computed tomography/positron emission tomography) of each variant and studied serum progranulin levels, APOE genotype and Alzheimer's disease (AD)-cerebrospinal fluid (CSF) biomarkers. Cases with a first-degree family history of early-onset dementia were genetically tested. RESULTS Ten of 15 (66%) nfvPPA, 5/5 (100%) svPPA and 7/7 (100%) lvPPA patients showed at least one positive neuroimaging-supported diagnostic criterion. All lvPPA and 3/5 (60%) uPPA patients presented AD-CSF biomarkers, which were absent in nfvPPA and svPPA cases. Four (27%) nfvPPA patients had dementia-causing mutations: 2 carried a GRN mutation and 2 the C9ORF72 hexanucleotide expansion. CONCLUSIONS There was an excellent association between clinical criteria and neuroimaging-supported biomarkers in svPPA and lvPPA, as well as with AD-CSF biochemical markers in the lvPPA. Neuroimaging, biochemical and genetic findings in nfvPPA were heterogeneous. Incorporating biochemical/genetic markers into the PPA clinical diagnosis would allow clinicians to improve their predictions of PPA neuropathology, especially in nfvPPA and uPPA cases.
Collapse
Affiliation(s)
- S Gil-Navarro
- Alzheimer's Disease and Other Cognitive Disorders Unit, Department of Neurology, Hospital Clínic, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sorbi S, Hort J, Erkinjuntti T, Fladby T, Gainotti G, Gurvit H, Nacmias B, Pasquier F, Popescu BO, Rektorova I, Religa D, Rusina R, Rossor M, Schmidt R, Stefanova E, Warren JD, Scheltens P. EFNS-ENS Guidelines on the diagnosis and management of disorders associated with dementia. Eur J Neurol 2013; 19:1159-79. [PMID: 22891773 DOI: 10.1111/j.1468-1331.2012.03784.x] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND OBJECTIVES The last version of the EFNS dementia guidelines is from 2007. In 2010, the revised guidelines for Alzheimer's disease (AD) were published. The current guidelines involve the revision of the dementia syndromes outside of AD, notably vascular cognitive impairment, frontotemporal lobar degeneration, dementia with Lewy bodies, corticobasal syndrome, progressive supranuclear palsy, Parkinson's disease dementia, Huntington's disease, prion diseases, normal-pressure hydrocephalus, limbic encephalitis and other toxic and metabolic disorders. The aim is to present a peer-reviewed evidence-based statement for the guidance of practice for clinical neurologists, geriatricians, psychiatrists and other specialist physicians responsible for the care of patients with dementing disorders. It represents a statement of minimum desirable standards for practice guidance. METHODS The task force working group reviewed evidence from original research articles, meta-analyses and systematic reviews, published by June 2011. The evidence was classified (I, II, III, IV) and consensus recommendations graded (A, B, or C) according to the EFNS guidance. Where there was a lack of evidence, but clear consensus, good practice points were provided. RESULTS AND CONCLUSIONS New recommendations and good practice points are made for clinical diagnosis, blood tests, neuropsychology, neuroimaging, electroencephalography, cerebrospinal fluid (CSF) analysis, genetic testing, disclosure of diagnosis, treatment of behavioural and psychological symptoms in dementia, legal issues, counselling and support for caregivers. All recommendations were revised as compared with the previous EFNS guidelines. The specialist neurologist together with primary care physicians play an important role in the assessment, interpretation and treatment of symptoms, disability and needs of dementia patients.
Collapse
Affiliation(s)
- S Sorbi
- Department of Neurological and Psychiatric Sciences, University of Florence, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Grossman M. Multimodal comparative studies of neurodegenerative diseases. J Alzheimers Dis 2013; 33 Suppl 1:S379-83. [PMID: 22531420 DOI: 10.3233/jad-2012-129002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Here we provide a brief description of our program to improve diagnostic accuracy in cases with phenotypically similar presentations that are due to distinct histopathologic abnormalities. We propose a staged approach to diagnosis, beginning with a screening assessment of specific, quantitative neuropsychological measures, and followed by assessments of imaging and biofluid biomarkers. Our goal is to determine the specific histopathologic abnormalities contributing to an individual's neurodegenerative condition.
Collapse
Affiliation(s)
- Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104-4283, USA.
| |
Collapse
|
36
|
Stamelou M, Quinn NP, Bhatia KP. “Atypical” atypical parkinsonism: New genetic conditions presenting with features of progressive supranuclear palsy, corticobasal degeneration, or multiple system atrophy-A diagnostic guide. Mov Disord 2013; 28:1184-99. [DOI: 10.1002/mds.25509] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 04/01/2013] [Accepted: 04/09/2013] [Indexed: 12/13/2022] Open
Affiliation(s)
- Maria Stamelou
- Sobell Department of Motor Neuroscience and Movement Disorders; UCL Institute of Neurology; London United Kingdom
- Neurology Clinic; Philipps-University; Marburg Germany
| | - Niall P. Quinn
- Sobell Department of Motor Neuroscience and Movement Disorders; UCL Institute of Neurology; London United Kingdom
| | - Kailash P. Bhatia
- Sobell Department of Motor Neuroscience and Movement Disorders; UCL Institute of Neurology; London United Kingdom
| |
Collapse
|
37
|
Goldman JS. New approaches to genetic counseling and testing for Alzheimer's disease and frontotemporal degeneration. Curr Neurol Neurosci Rep 2013; 12:502-10. [PMID: 22773362 DOI: 10.1007/s11910-012-0296-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The discovery of new autosomal dominant and susceptibility genes for Alzheimer's disease (AD) and frontotemporal degeneration (FTD) is revealing important new information about the neurodegenerative process and the risk for acquiring these diseases. It is becoming increasingly clear that both the mechanisms that drive these diseases and their phenotypes overlap. New technologies will assist access to genetic testing but may increase difficulty with genetic test interpretation. Thus, the process of genetic counseling and testing for these diseases is becoming more complex. This article will review current knowledge on the genetics of AD and FTD and suggest clinical guidelines for helping families to navigate through these complexities. The implications of future discoveries will be offered.
Collapse
Affiliation(s)
- Jill S Goldman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 630 W. 168th St., P & S Box 16, New York, NY 10032, USA.
| |
Collapse
|
38
|
The Challenges of Service Provision in Younger-Onset Dementia. J Am Med Dir Assoc 2013; 14:230-2. [DOI: 10.1016/j.jamda.2013.01.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 01/16/2013] [Indexed: 11/22/2022]
|
39
|
Beldarrain MG, Ojeda JR, Ferrer I, Garcia-Monco JC. Aprosodic speech with insular hyperintensities and 4R Tau pathology on autopsy. Neurocase 2013; 19:583-6. [PMID: 22992154 DOI: 10.1080/13554794.2012.713489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We describe a 46-year-old woman who presented with a 2-year history of aprosodic speech together with apathy and disinhibition. Brain magnetic resonance imaging showed subcortical hyperintensities over both insular regions that later extended to both frontal and temporal cortices. The post-mortem exam showed a massive tau protein deposition throughout the brain. No mutation in the gene MAPT was detected. This case illustrates an atypical clinical-radiological presentation of a frontotemporal dementia with an unusual speech and abnormal signal of both insulae. Furthermore, it reinforces the crucial role of the insula in the development of symptoms in frontotemporal dementia.
Collapse
|
40
|
Abstract
Frontotemporal dementia (FTD) is a common dementia syndrome in patients under the age of 65 years with many features overlapping with amyotrophic lateral sclerosis (ALS). The link between FTD and ALS has been strengthened by the discovery that a hexanucleotide repeat expansion in a non-coding region of the C9ORF72 gene causes both familial and sporadic types of these two diseases. As we begin to understand the pathophysiological mechanisms by which this mutation leads to FTD and ALS (c9FTD/ALS), new targets for disease-modifying therapies will likely be unveiled. Putative C9ORF72 expansion pathogenic mechanisms include loss of C9ORF72 protein function, sequestration of nucleic acid binding proteins due to expanded hexanucleotide repeats, or a combination of the two. New animal models and other research tools informed by work in other repeat expansion neurodegenerative diseases such as the spinocerebellar ataxias will help to elucidate the mechanisms of C9ORF72-mediated disease. Similarly, re-examining previous studies of drugs developed to treat ALS in light of this new mutation may identify novel FTD treatments. Ultimately, research consortiums incorporating animal models and well-characterized clinical populations will be necessary to fully understand the natural history of the c9FTD/ALS clinical phenotypes and identify biomarkers and therapeutic agents that can cure the most common form of genetically determined FTD and ALS.
Collapse
Affiliation(s)
- Sharon J Sha
- University of California, San Francisco, Memory and Aging Center, Box 1207, San Francisco, CA 94143-1207, USA
| | - Adam Boxer
- University of California, San Francisco, Memory and Aging Center, Box 1207, San Francisco, CA 94143-1207, USA
| |
Collapse
|
41
|
Boxer AL, Gold M, Huey E, Gao FB, Burton EA, Chow T, Kao A, Leavitt BR, Lamb B, Grether M, Knopman D, Cairns NJ, Mackenzie IR, Mitic L, Roberson ED, Van Kammen D, Cantillon M, Zahs K, Salloway S, Morris J, Tong G, Feldman H, Fillit H, Dickinson S, Khachaturian Z, Sutherland M, Farese R, Miller BL, Cummings J. Frontotemporal degeneration, the next therapeutic frontier: molecules and animal models for frontotemporal degeneration drug development. Alzheimers Dement 2012; 9:176-88. [PMID: 23043900 DOI: 10.1016/j.jalz.2012.03.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Accepted: 03/07/2012] [Indexed: 02/04/2023]
Abstract
Frontotemporal degeneration (FTD) is a common cause of dementia for which there are currently no approved therapies. Over the past decade, there has been an explosion of knowledge about the biology and clinical features of FTD that has identified a number of promising therapeutic targets as well as animal models in which to develop drugs. The close association of some forms of FTD with neuropathological accumulation of tau protein or increased neuroinflammation due to progranulin protein deficiency suggests that a drug's success in treating FTD may predict efficacy in more common diseases such as Alzheimer's disease. A variety of regulatory incentives, clinical features of FTD such as rapid disease progression, and relatively pure molecular pathology suggest that there are advantages to developing drugs for FTD as compared with other more common neurodegenerative diseases such as Alzheimer's disease. In March 2011, the Frontotemporal Degeneration Treatment Study Group sponsored a conference entitled "FTD, the Next Therapeutic Frontier," which focused on preclinical aspects of FTD drug development. The goal of the meeting was to promote collaborations between academic researchers and biotechnology and pharmaceutical researchers to accelerate the development of new treatments for FTD. Here we report the key findings from the conference, including the rationale for FTD drug development; epidemiological, genetic, and neuropathological features of FTD; FTD animal models and how best to use them; and examples of successful drug development collaborations in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
The non-fluent/agrammatic variant of primary progressive aphasia (naPPA) is a young-onset neurodegenerative disorder characterised by poor grammatical comprehension and expression and a disorder of speech sound production. In an era of disease-modifying treatments, the identification of naPPA might be an important step in establishing a specific cause of neurodegenerative disease. However, difficulties in defining the characteristic language deficits and heterogeneity in the anatomical distribution of disease in naPPA have led to controversy. Findings from imaging studies have linked an impairment of this uniquely human language capacity with disruption of large-scale neural networks centred in left inferior frontal and anterior superior temporal regions. Accordingly, the pathological burden of disease in naPPA is anatomically focused in these regions. Most cases of naPPA are associated with the spectrum of pathological changes found in frontotemporal lobar degeneration involving the microtubule-associated protein tau. Knowledge of these unique clinical-pathological associations should advance care for patients with this important class of neurodegenerative diseases while supplementing our knowledge of human cognitive neuroscience.
Collapse
Affiliation(s)
- Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104-4283, USA.
| |
Collapse
|
43
|
Ghidoni R, Paterlini A, Albertini V, Binetti G, Benussi L. Losing protein in the brain: the case of progranulin. Brain Res 2012; 1476:172-82. [PMID: 22348647 DOI: 10.1016/j.brainres.2012.01.075] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 01/18/2012] [Accepted: 01/28/2012] [Indexed: 01/22/2023]
Abstract
It is well known that progranulin protein is involved in wound repair, inflammation, and tumor formation. The wedding between progranulin and brain was celebrated in 2006 with the involvement of progranulin gene (GRN) in Frontotemporal lobar degeneration (FTLD), the most common form of early-onset dementia: up to date, 75 mutations have been detected in FTLD patients as well as in patients with widely variable clinical phenotypes. All pathogenic GRN mutations identified thus far cause the disease through a uniform mechanism, i.e. loss of functional progranulin or haploinsufficiency. Studies on GRN knockout mice suggest that progranulin-related neurodegenerative diseases may result from lifetime depletion of neurotrophic support together with cumulative damage in association with dysregulated inflammation, thus highlighting possible new molecular targets for GRN-related FTLD treatment. Recently, the dosage of plasma progranulin has been proposed as a useful tool for a quick and inexpensive large-scale screening of affected and unaffected carriers of GRN mutations. Before it is systematically translated into clinical practice and, more importantly, included into diagnostic criteria for dementias, further standardization of plasma progranulin test and harmonization of its use are required. Once a specific treatment becomes available for these pathologies, this test - being applicable on large scale - will represent an important step towards personalized healthcare. This article is part of a Special Issue entitled: Brain Integration.
Collapse
Affiliation(s)
- Roberta Ghidoni
- Proteomics Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| | | | | | | | | |
Collapse
|
44
|
Intrafamilial clinical phenotypic heterogeneity with progranulin gene p.Glu498fs mutation. J Neurol Sci 2012; 316:189-90. [PMID: 22280948 DOI: 10.1016/j.jns.2012.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Revised: 12/23/2011] [Accepted: 01/04/2012] [Indexed: 12/12/2022]
Abstract
A patient with a progressive aphasia syndrome underwent progranulin gene (GRN) testing in light of a family history of early-onset dementia in two of her brothers, one of whom had been previously examined and had the phenotype of frontal variant frontotemporal dementia. The proband was found to have the p.Glu498fs mutation. This is only the second English family, and the fifth family overall, to be described with this GRN mutation. There was marked intrafamilial phenotypic heterogeneity with respect to age at onset and clinical presentation. The mechanisms underpinning this heterogeneity, as seen with other GRN mutations, are currently unknown. Since all GRN mutations lead to progranulin haploinsufficiency, other modifying factors, possibly genetic, are implicated.
Collapse
|
45
|
Ghidoni R, Stoppani E, Rossi G, Piccoli E, Albertini V, Paterlini A, Glionna M, Pegoiani E, Agnati LF, Fenoglio C, Scarpini E, Galimberti D, Morbin M, Tagliavini F, Binetti G, Benussi L. Optimal Plasma Progranulin Cutoff Value for Predicting Null Progranulin Mutations in Neurodegenerative Diseases: A Multicenter Italian Study. NEURODEGENER DIS 2012; 9:121-7. [DOI: 10.1159/000333132] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 09/13/2011] [Indexed: 11/19/2022] Open
|
46
|
Parkinsonism and frontotemporal dementia: the clinical overlap. J Mol Neurosci 2011; 45:343-9. [PMID: 21892619 DOI: 10.1007/s12031-011-9632-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Accepted: 08/14/2011] [Indexed: 12/13/2022]
Abstract
Frontotemporal dementia is commonly associated with parkinsonism in several sporadic (i.e., progressive supranuclear palsy, corticobasal degeneration) and familial neurodegenerative disorders (i.e., frontotemporal dementia associated with parkinsonism and MAPT or progranulin mutations in chromosome 17). The clinical diagnosis of these disorders may be challenging in view of overlapping clinical features, particularly in speech, language, and behavior. The motor and cognitive phenotypes can be viewed within a spectrum of clinical, pathologic, and genetic disorders with no discrete clinicopathologic correlations but rather lying within a dementia-parkinsonism continuum. Neuroimaging and cerebrospinal fluid analysis can be helpful, but the poor specificity of clinical and imaging features has enormously challenged the development of biological markers that could differentiate these disorders premortem. This gap is critical to bridge in order to allow testing of novel biological therapies that may slow the progression of these proteinopathies.
Collapse
|