1
|
Stefanova E, Marjanović A, Dobričić V, Mandić-Stojmenović G, Stojković T, Branković M, Šarčević M, Novaković I, Kostić VS. Frequency of C9orf72, GRN, and MAPT pathogenic variants in patients recruited at the Belgrade Memory Center. Neurogenetics 2024; 25:193-200. [PMID: 38847891 DOI: 10.1007/s10048-024-00766-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/02/2024] [Indexed: 07/16/2024]
Abstract
Most of the heritability in frontotemporal dementia (FTD) is accounted for by autosomal dominant hexanucleotide expansion in the chromosome 9 open reading frame 72 (C9orf72), pathogenic/likely pathogenic variants in progranulin (GRN), and microtubule-associated protein tau (MAPT) genes. Until now, there has been no systematic analysis of these genes in the Serbian population. Herein, we assessed the frequency of the C9orf72 expansion, pathogenic/likely pathogenic variants in GRN and MAPT in a well-characterized group of 472 subjects (FTD, Alzheimer's disease - AD, mild cognitive impairment - MCI, and unspecified dementia - UnD), recruited in the Memory Center, Neurology Clinic, University Clinical Center of Serbia. The C9orf72 repeat expansion was detected in 6.98% of FTD cases (13.46% familial; 2.6% sporadic). In the UnD subgroup, C9orf72 repeat expansions were detected in 4.08% (8% familial) individuals. Pathogenic variants in the GRN were found in 2.85% of familial FTD cases. Interestingly, no MAPT pathogenic/likely pathogenic variants were detected, suggesting possible geographical specificity. Our findings highlight the importance of wider implementation of genetic testing in neurological and psychiatric practice managing patients with cognitive-behavioral and motor symptoms.
Collapse
Affiliation(s)
- Elka Stefanova
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, Belgrade, 11000, Serbia.
- Neurology Clinic, University Clinical Center of Serbia (UCCS), Dr Subotića 6, Belgrade, 11000, Serbia.
| | - Ana Marjanović
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, Belgrade, 11000, Serbia
| | - Valerija Dobričić
- Neurology Clinic, University Clinical Center of Serbia (UCCS), Dr Subotića 6, Belgrade, 11000, Serbia
- University of Lübeck-Lübeck Interdisciplinary Platform for Genome Analytics, 11000, Lübeck, Germany
| | - Gorana Mandić-Stojmenović
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, Belgrade, 11000, Serbia
- Neurology Clinic, University Clinical Center of Serbia (UCCS), Dr Subotića 6, Belgrade, 11000, Serbia
| | - Tanja Stojković
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, Belgrade, 11000, Serbia
- Neurology Clinic, University Clinical Center of Serbia (UCCS), Dr Subotića 6, Belgrade, 11000, Serbia
| | - Marija Branković
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, Belgrade, 11000, Serbia
| | - Maksim Šarčević
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, Belgrade, 11000, Serbia
| | - Ivana Novaković
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, Belgrade, 11000, Serbia
- Neurology Clinic, University Clinical Center of Serbia (UCCS), Dr Subotića 6, Belgrade, 11000, Serbia
| | - Vladimir S Kostić
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, Belgrade, 11000, Serbia
- Neurology Clinic, University Clinical Center of Serbia (UCCS), Dr Subotića 6, Belgrade, 11000, Serbia
| |
Collapse
|
2
|
Almeida MR, Tábuas-Pereira M, Baldeiras I, Lima M, Durães J, Massano J, Pinto M, Cruto C, Santana I. Characterization of Progranulin Gene Mutations in Portuguese Patients with Frontotemporal Dementia. Int J Mol Sci 2023; 25:511. [PMID: 38203682 PMCID: PMC10778719 DOI: 10.3390/ijms25010511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
In Portugal, heterozygous loss-of-function mutations in the progranulin (GRN) gene account for approximately half of the genetic mediated forms of frontotemporal dementia (FTD). GRN mutations reported thus far cause FTD through a haploinsufficiency disease mechanism. Herein, we aim to unveil the GRN mutation spectrum, investigated in 257 FTD patients and 19 family members from the central/north region of Portugal using sequencing methods. Seven different pathogenic variants were identified in 46 subjects including 40 patients (16%) and 6 relatives (32%). bvFTD was the most common clinical presentation among the GRN mutation patients, who showed a global pattern of moderate-to-severe frontotemporoparietal deficits in the neuropsychological evaluation. Interestingly, two mutations were novel (p.Thr238Profs*18, p.Leu354Profs*16), and five were previously described, although three of them only in the Portuguese population, suggesting a population-specific GRN mutational spectrum. The subjects harboring a GRN mutation showed a significant reduction in serum PGRN levels, supporting the pathogenic nature of these variants. This work broadens the mutation spectrum of GRN and the identification of the underlying GRN mutations provided an accurate genetic counselling and allowed the enrolment of subjects with GRN mutations (both asymptomatic and symptomatic) in ongoing clinical trials, which is essential to test new drugs for the disease.
Collapse
Affiliation(s)
- Maria Rosário Almeida
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (I.B.); (I.S.)
| | - Miguel Tábuas-Pereira
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal; (M.T.-P.); (M.L.); (J.D.)
| | - Inês Baldeiras
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (I.B.); (I.S.)
- Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal
| | - Marisa Lima
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal; (M.T.-P.); (M.L.); (J.D.)
| | - João Durães
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal; (M.T.-P.); (M.L.); (J.D.)
| | - João Massano
- Neurology Department, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal; (J.M.); (M.P.)
| | - Madalena Pinto
- Neurology Department, Centro Hospitalar Universitário de São João, 4200-319 Porto, Portugal; (J.M.); (M.P.)
| | - Catarina Cruto
- Neurology Department, Hospital Pedro Hispano, Unidade Local de Saúde de Matosinhos, 4464-513 Matosinhos, Portugal;
| | - Isabel Santana
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (I.B.); (I.S.)
- Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal
| |
Collapse
|
3
|
Muraleedharan A, Vanderperre B. The endo-lysosomal system in Parkinson's disease: expanding the horizon. J Mol Biol 2023:168140. [PMID: 37148997 DOI: 10.1016/j.jmb.2023.168140] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/08/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease, and its prevalence is increasing with age. A wealth of genetic evidence indicates that the endo-lysosomal system is a major pathway driving PD pathogenesis with a growing number of genes encoding endo-lysosomal proteins identified as risk factors for PD, making it a promising target for therapeutic intervention. However, detailed knowledge and understanding of the molecular mechanisms linking these genes to the disease are available for only a handful of them (e.g. LRRK2, GBA1, VPS35). Taking on the challenge of studying poorly characterized genes and proteins can be daunting, due to the limited availability of tools and knowledge from previous literature. This review aims at providing a valuable source of molecular and cellular insights into the biology of lesser-studied PD-linked endo-lysosomal genes, to help and encourage researchers in filling the knowledge gap around these less popular genetic players. Specific endo-lysosomal pathways discussed range from endocytosis, sorting, and vesicular trafficking to the regulation of membrane lipids of these membrane-bound organelles and the specific enzymatic activities they contain. We also provide perspectives on future challenges that the community needs to tackle and propose approaches to move forward in our understanding of these poorly studied endo-lysosomal genes. This will help harness their potential in designing innovative and efficient treatments to ultimately re-establish neuronal homeostasis in PD but also other diseases involving endo-lysosomal dysfunction.
Collapse
Affiliation(s)
- Amitha Muraleedharan
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois and Biological Sciences Department, Université du Québec à Montréal
| | - Benoît Vanderperre
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois and Biological Sciences Department, Université du Québec à Montréal
| |
Collapse
|
4
|
Tan YJ, Yong ACW, Foo JN, Lian MM, Lim WK, Dominguez J, Fong ZH, Narasimhalu K, Chiew HJ, Ng KP, Ting SKS, Kandiah N, Ng ASL. C9orf72 expansions are the most common cause of genetic frontotemporal dementia in a Southeast Asian cohort. Ann Clin Transl Neurol 2023; 10:568-578. [PMID: 36799407 PMCID: PMC10109321 DOI: 10.1002/acn3.51744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/18/2023] Open
Abstract
OBJECTIVE Frontotemporal dementia (FTD) encompasses a spectrum of neurodegenerative disorders, including behavioural variant FTD (bvFTD), semantic variant primary progressive aphasia (svPPA) and non-fluent variant PPA (nfvPPA). While a strong genetic component is implicated in FTD, genetic FTD in Asia is less frequently reported. We aimed to investigate the frequency of Southeast Asian FTD patients harbouring known genetic FTD variants. METHODS A total of 60 FTD-spectrum patients (25 familial and 35 sporadic) from Singapore and the Philippines were included. All underwent next-generation sequencing and repeat-primed PCR for C9orf72 expansion testing. Neurofilament light chain (NfL) levels were measured in a subset of patients. RESULTS Overall, 26.6% (16/60 cases) carried pathogenic or likely pathogenic variants in a FTD-related gene, including: MAPT Gln351Arg (n = 1); GRN Cys92Ter (n = 1), Ser301Ter (n = 2), c.462 + 1G > C (n = 1); C9orf72 expansion (35-70 repeats; n = 8); TREM2 Arg47Cys (n = 1); and OPTN frameshift insertion (n = 2). Genetic mutations accounted for 48% (12/25) of patients with familial FTD, and 11.4% (4/35) of patients with sporadic FTD. C9orf72 repeat expansions were the most common genetic mutation (13.3%, 8/60), followed by GRN (6.7%, 4/60) variants. Within mutation carriers, plasma NfL was highest in a C9orf72 expansion carrier, and CSF NfL was highest in a GRN splice variant carrier. INTERPRETATION In our cohort, genetic mutations are present in one-quarter of FTD-spectrum cases, and up to half of those with family history. Our findings highlight the importance of wider implementation of genetic testing in FTD patients from Southeast Asia.
Collapse
Affiliation(s)
- Yi Jayne Tan
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Alisa C W Yong
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Jia Nee Foo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Human Genetics, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Michelle M Lian
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Weng Khong Lim
- Singhealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore.,Cancer & Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | | | - Zhi Hui Fong
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Kaavya Narasimhalu
- Singhealth Duke-NUS Institute of Precision Medicine, Singapore, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
| | - Hui Jin Chiew
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Kok Pin Ng
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Simon K S Ting
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore, Singapore
| | - Nagaendran Kandiah
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Adeline S L Ng
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Singapore, Singapore.,Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
5
|
Kashyap SN, Boyle NR, Roberson ED. Preclinical Interventions in Mouse Models of Frontotemporal Dementia Due to Progranulin Mutations. Neurotherapeutics 2023; 20:140-153. [PMID: 36781744 PMCID: PMC10119358 DOI: 10.1007/s13311-023-01348-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 02/15/2023] Open
Abstract
Heterozygous loss-of-function mutations in progranulin (GRN) cause frontotemporal dementia (FTD), a leading cause of early-onset dementia characterized clinically by behavioral, social, and language deficits. There are currently no FDA-approved therapeutics for FTD-GRN, but this has been an active area of investigation, and several approaches are now in clinical trials. Here, we review preclinical development of therapies for FTD-GRN with a focus on testing in mouse models. Since most FTD-GRN-associated mutations cause progranulin haploinsufficiency, these approaches focus on raising progranulin levels. We begin by considering the disorders associated with altered progranulin levels, and then review the basics of progranulin biology including its lysosomal, neurotrophic, and immunomodulatory functions. We discuss mouse models of progranulin insufficiency and how they have been used in preclinical studies on a variety of therapeutic approaches. These include approaches to raise progranulin expression from the normal allele or facilitate progranulin production by the mutant allele, as well as approaches to directly increase progranulin levels by delivery across the blood-brain barrier or by gene therapy. Several of these approaches have entered clinical trials, providing hope that new therapies for FTD-GRN may be the next frontier in the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
- Shreya N Kashyap
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Medical Scientist Training Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nicholas R Boyle
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Medical Scientist Training Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Medical Scientist Training Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
6
|
Piehl N, van Olst L, Ramakrishnan A, Teregulova V, Simonton B, Zhang Z, Tapp E, Channappa D, Oh H, Losada PM, Rutledge J, Trelle AN, Mormino EC, Elahi F, Galasko DR, Henderson VW, Wagner AD, Wyss-Coray T, Gate D. Cerebrospinal fluid immune dysregulation during healthy brain aging and cognitive impairment. Cell 2022; 185:5028-5039.e13. [PMID: 36516855 PMCID: PMC9815831 DOI: 10.1016/j.cell.2022.11.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/27/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022]
Abstract
Cerebrospinal fluid (CSF) contains a tightly regulated immune system. However, knowledge is lacking about how CSF immunity is altered with aging or neurodegenerative disease. Here, we performed single-cell RNA sequencing on CSF from 45 cognitively normal subjects ranging from 54 to 82 years old. We uncovered an upregulation of lipid transport genes in monocytes with age. We then compared this cohort with 14 cognitively impaired subjects. In cognitively impaired subjects, downregulation of lipid transport genes in monocytes occurred concomitantly with altered cytokine signaling to CD8 T cells. Clonal CD8 T effector memory cells upregulated C-X-C motif chemokine receptor 6 (CXCR6) in cognitively impaired subjects. The CXCR6 ligand, C-X-C motif chemokine ligand 16 (CXCL16), was elevated in the CSF of cognitively impaired subjects, suggesting CXCL16-CXCR6 signaling as a mechanism for antigen-specific T cell entry into the brain. Cumulatively, these results reveal cerebrospinal fluid immune dysregulation during healthy brain aging and cognitive impairment.
Collapse
Affiliation(s)
- Natalie Piehl
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lynn van Olst
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Abhirami Ramakrishnan
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Victoria Teregulova
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Brooke Simonton
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ziyang Zhang
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emma Tapp
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Divya Channappa
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hamilton Oh
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Graduate Program in Stem Cell and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Patricia M Losada
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Jarod Rutledge
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Elizabeth C Mormino
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Department of Psychology, Stanford University, Stanford, CA, USA
| | - Fanny Elahi
- Departments of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, La Jolla, CA, USA
| | - Douglas R Galasko
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, USA
| | - Victor W Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony D Wagner
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Department of Psychology, Stanford University, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; The Phil and Penny Initiative for Brain Resilience, Stanford University, Stanford, CA, USA; Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
7
|
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: 1.0] [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
|
8
|
Zheng X, Mi T, Wang R, Zhang Z, Li W, Zhao J, Yang P, Xia H, Mao Q. Progranulin deficiency promotes persistent neuroinflammation and causes regional pathology in the hippocampus following traumatic brain injury. Glia 2022; 70:1317-1336. [PMID: 35362178 DOI: 10.1002/glia.24175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 11/07/2022]
Abstract
Traumatic brain injury (TBI) can be progressive and can lead to the development of a long-term complication termed chronic traumatic encephalopathy. The mechanisms underlying the progressive changes are still unknown; however, studies have suggested that microglia-mediated neuroinflammation in response to TBI may play a fundamental role. This study aimed to determine whether progranulin (PGRN), a major modulator of microglial activity, plays a role in the progressive damage following TBI. PGRN-deficient and wild-type mice were subjected to controlled cortical impact and were observed neuropathologically after 3 days, 7 days, and 5 months. Compared to sham and wild-type mice, the PGRN-deficient mice showed overall stronger microgliosis and astrocytosis. The astrocytosis involved broader areas than the microgliosis and was more prominent in the basal ganglia, hippocampus, and internal capsule in PGRN-deficient mice. Ongoing neuronal death was uniquely observed in the hippocampal CA3 region of PGRN-deficient mice at 5 months after TBI, accompanying the regional chronic microgliosis and astrocytosis involving the CA3 commissural pathway. In addition, there was M1 microglial polarization in the pericontusional area with activated TLR4/MyD88/NF-κB signaling; however, the hippocampus showed only mild M1 polarization 7 days after TBI. Lastly, Morris water maze tests showed PGRN-deficient mice had poorer spatial learning and memory 5 months after TBI than wild-type or sham mice. The data indicated the PGRN deficiency caused TBI progression by promoting persistent microgliosis with microglial polarization and astrocytosis, as well as regional pathology in the hippocampus. The study suggests that PGRN should be evaluated as a potential therapy for TBI.
Collapse
Affiliation(s)
- Xiaojing Zheng
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Tiantian Mi
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Rong Wang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Zihan Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Wenyan Li
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Junli Zhao
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Peiyan Yang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
9
|
TDP-43 pathology: from noxious assembly to therapeutic removal. Prog Neurobiol 2022; 211:102229. [DOI: 10.1016/j.pneurobio.2022.102229] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/08/2021] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
|
10
|
Christodoulidou A, McKenna GE, Holden ST, Rowe JB, Cope TE. Are the UK genetic testing criteria for dementia too exclusive? J Neurol 2021; 269:2222-2226. [PMID: 34748083 DOI: 10.1007/s00415-021-10867-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 11/27/2022]
Affiliation(s)
- Annita Christodoulidou
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Georgina E McKenna
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Simon T Holden
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- East Anglian Medical Genetics Service, Cambridge, UK
| | - James B Rowe
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Thomas E Cope
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
11
|
Nalls MA, Blauwendraat C, Sargent L, Vitale D, Leonard H, Iwaki H, Song Y, Bandres-Ciga S, Menden K, Faghri F, Heutink P, Cookson MR, Singleton AB. Evidence for GRN connecting multiple neurodegenerative diseases. Brain Commun 2021; 3:fcab095. [PMID: 34693284 DOI: 10.1093/braincomms/fcab095] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
Previous research using genome-wide association studies has identified variants that may contribute to lifetime risk of multiple neurodegenerative diseases. However, whether there are common mechanisms that link neurodegenerative diseases is uncertain. Here, we focus on one gene, GRN, encoding progranulin, and the potential mechanistic interplay between genetic risk, gene expression in the brain and inflammation across multiple common neurodegenerative diseases. We utilized genome-wide association studies, expression quantitative trait locus mapping and Bayesian colocalization analyses to evaluate potential causal and mechanistic inferences. We integrate various molecular data types from public resources to infer disease connectivity and shared mechanisms using a data-driven process. Expression quantitative trait locus analyses combined with genome-wide association studies identified significant functional associations between increasing genetic risk in the GRN region and decreased expression of the gene in Parkinson's, Alzheimer's and amyotrophic lateral sclerosis. Additionally, colocalization analyses show a connection between blood-based inflammatory biomarkers relating to platelets and GRN expression in the frontal cortex. GRN expression mediates neuroinflammation function related to multiple neurodegenerative diseases. This analysis suggests shared mechanisms for Parkinson's, Alzheimer's and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA.,Data Tecnica International LLC, Glen Echo, MD 20812, USA
| | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lana Sargent
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dan Vitale
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA.,Data Tecnica International LLC, Glen Echo, MD 20812, USA
| | - Hampton Leonard
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA.,Data Tecnica International LLC, Glen Echo, MD 20812, USA.,German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
| | - Hirotaka Iwaki
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA.,Data Tecnica International LLC, Glen Echo, MD 20812, USA
| | - Yeajin Song
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA.,Data Tecnica International LLC, Glen Echo, MD 20812, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin Menden
- German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
| | - Faraz Faghri
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA.,Data Tecnica International LLC, Glen Echo, MD 20812, USA
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE), Tuebingen, Germany
| | - Mark R Cookson
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Dominguez J, Ng A, Yu J, Guevarra AC, Daroy ML, Alfon A, Catindig JA, Dizon M, Santiago J, Del Moral MC, Yu J, Jamerlan A, Ligsay A, Bagyinszky E, An SS, Kim S. Autosomal Dominant Frontotemporal Lobar Degeneration in a Filipino Family with Progranulin Mutation. Dement Geriatr Cogn Disord 2021; 49:557-564. [PMID: 33486486 DOI: 10.1159/000510106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/10/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Compared to Western populations, familial frontotemporal lobar degeneration (FTLD) is rare among Asians. Progranulin (GRN) gene mutation, which is a major cause of FTLD, is likewise rare. We present a family with FTLD from the Philippines with an autosomal dominant pattern of inheritance and GRN mutation and briefly review reports of GRN mutations in Asia. CASE PRESENTATION The proband is 66 years old with progressive nonfluent aphasia (PNFA)-corticobasal syndrome . We assessed 3 generations of her pedigree and found 11 affected relatives with heterogenous phenotypes, usually behavioral variant frontotemporal dementia (FTD) and PNFA. Neuroimaging showed atrophy and hypometabolism consistent with FTD syndromes. White matter hyperintensities were seen in affected members even in the absence of vascular risk factors. A GRN mutation R110X was found in 6 members, 3 with symptoms and 3 were asymptomatic. Plasma GRN was low (<112 ng/mL) in all mutation carriers. No mutations were found in microtubule-associated protein tau, APP, PSEN1, and PSEN2 genes, and all were APOE3. CONCLUSION This is the first Filipino family with autosomal dominant FTD documented with GRN mutation. Identifying families and cohorts would contribute to therapeutic developments in an area with FTD-GRN.
Collapse
Affiliation(s)
- Jacqueline Dominguez
- Institute for Neurosciences, St. Luke's Medical Center, Quezon City, Philippines,
| | - Arlene Ng
- Institute for Neurosciences, St. Luke's Medical Center, Quezon City, Philippines
| | - Jeryl Yu
- Institute for Neurosciences, St. Luke's Medical Center, Quezon City, Philippines
| | - Anne Cristine Guevarra
- Research and Biotechnology Division, St. Luke's Medical Center, Quezon City, Philippines
| | - Maria Luisa Daroy
- Research and Biotechnology Division, St. Luke's Medical Center, Quezon City, Philippines
| | - Alicia Alfon
- Research and Biotechnology Division, St. Luke's Medical Center, Quezon City, Philippines
| | - Joseree-Ann Catindig
- Memory Center-Institute for Neurosciences, St. Luke's Medical Center, Taguig City, Philippines
| | - Mercedes Dizon
- Institute of Radiology, St. Luke's Medical Center, Quezon City, Philippines
| | - Jonas Santiago
- PET Center, St. Luke's Medical Center, Quezon City, Philippines
| | | | - Justine Yu
- Memory Center-Institute for Neurosciences, St. Luke's Medical Center, Taguig City, Philippines
| | - Angelo Jamerlan
- Department of Bionano Technology, Gachon University, Seongnam, Republic of Korea
| | - Antonio Ligsay
- Section of Clinical Research, St. Luke's Medical Center - College of Medicine, Quezon City, Philippines
| | - Eva Bagyinszky
- Department of Industrial and Environmental Engineering, Gachon University, Seongnam, Republic of Korea
| | - Seong Soo An
- Department of Bionano Technology, Gachon University, Seongnam, Republic of Korea
| | - Sangyun Kim
- Department of Neurology, Seoul National University College of Medicine & Neurocognitive Behavior Center, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
13
|
Kucikova L, Goerdten J, Dounavi ME, Mak E, Su L, Waldman AD, Danso S, Muniz-Terrera G, Ritchie CW. Resting-state brain connectivity in healthy young and middle-aged adults at risk of progressive Alzheimer's disease. Neurosci Biobehav Rev 2021; 129:142-153. [PMID: 34310975 DOI: 10.1016/j.neubiorev.2021.07.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 05/18/2021] [Accepted: 07/21/2021] [Indexed: 11/15/2022]
Abstract
Functional brain connectivity of the resting-state networks has gained recent attention as a possible biomarker of Alzheimer's Disease (AD). In this paper, we review the literature of functional connectivity differences in young adults and middle-aged cognitively intact individuals with non-modifiable risk factors of AD (n = 17). We focus on three main intrinsic resting-state networks: The Default Mode network, Executive network, and the Salience network. Overall, the evidence from the literature indicated early vulnerability of functional connectivity across different at-risk groups, particularly in the Default Mode Network. While there was little consensus on the interpretation on directionality, the topography of the findings showed frequent overlap across studies, especially in regions that are characteristic of AD (i.e., precuneus, posterior cingulate cortex, and medial prefrontal cortex areas). We conclude that while resting-state functional connectivity markers have great potential to identify at-risk individuals, implementing more data-driven approaches, further longitudinal and cross-validation studies, and the analysis of greater sample sizes are likely to be necessary to fully establish the effectivity and utility of resting-state network-based analyses.
Collapse
Affiliation(s)
- Ludmila Kucikova
- Edinburgh Dementia Prevention and Centre for Clinical Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom.
| | - Jantje Goerdten
- Department of Epidemiological Methods and Etiological Research, Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
| | - Maria-Eleni Dounavi
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Elijah Mak
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Li Su
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Adam D Waldman
- Edinburgh Dementia Prevention and Centre for Clinical Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| | - Samuel Danso
- Edinburgh Dementia Prevention and Centre for Clinical Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| | - Graciela Muniz-Terrera
- Edinburgh Dementia Prevention and Centre for Clinical Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| | - Craig W Ritchie
- Edinburgh Dementia Prevention and Centre for Clinical Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
14
|
Bhopatkar AA, Rangachari V. Are granulins copper sequestering proteins? Proteins 2020; 89:450-461. [PMID: 33252789 DOI: 10.1002/prot.26031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023]
Abstract
Granulins (GRN 1-7) are short (~6 kDa), cysteine-rich proteins that are generated upon the proteolytic processing of progranulin (PGRN). These peptides, along with their precursor, have been implicated in multiple pathophysiological roles, especially in neurodegenerative diseases. Previously we showed that GRN-3 and GRN-5 are fully disordered in the reduced form implicating redox sensitive attributes to the proteins. Redox-based modulations are often carried out by metalloproteins in mitigating oxidative stress and maintaining metal-homeostasis within cells. To probe whether GRNs play a role in metal sequestration, we tested the metal binding propensity of the reduced forms of GRNs -3 and - 5 under neutral and acidic pH mimicking cytosolic and lysosomal conditions, respectively. We found, at neutral pH, both GRNs selectively bind Cu and no other divalent metal cations, with a greater specificity for Cu(I). Binding of Cu did not result in a disorder-to-order structural transition but partly triggered the multimerization of GRNs via uncoordinated cystines at both pH conditions. Overall, the results indicate that GRNs -3 and - 5 have surprisingly strong affinity for Cu in the pM range, comparable to other known copper sequestering proteins. The results also hint at a potential of GRNs to reduce Cu(II) to Cu(I), a process that has significance in mitigating Cu-induced ROS cytotoxicity in cells. Together, this report uncovers metal-coordinating property of GRNs for the first time, which may have profound significance in their structure and pathophysiological functions.
Collapse
Affiliation(s)
- Anukool A Bhopatkar
- Department of Chemistry and Biochemistry, School of Mathematics and Natural Sciences and, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Vijayaraghavan Rangachari
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| |
Collapse
|
15
|
Cipriani G, Danti S, Nuti A, Di Fiorino M, Cammisuli DM. Is that schizophrenia or frontotemporal dementia? Supporting clinicians in making the right diagnosis. Acta Neurol Belg 2020; 120:799-804. [PMID: 32314269 DOI: 10.1007/s13760-020-01352-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/31/2020] [Indexed: 01/01/2023]
Abstract
Schizophrenia (SCH) and frontotemporal dementia (FTD) are neurobehavioral syndromes characterized by a profound alteration in personal and social conduct. Differential diagnosis between SCH and FTD remains a challenge. In this short narrative review, we summarize evidences regarding similarities and differences between these disorders to support clinicians in making the right diagnosis. Reports of FTD misdiagnosed as schizophrenia or schizophrenia-like psychosis are frequently reported in the literature. The behavioural variant of FTD (bvFTD) along with familial FTD characterized by delusions and hallucinations represent the medical conditions that best illustrate overlaps between psychiatry and neurology. Neuropsychological patterns of core deficits and anatomical and physiological brain alterations primarily concur in differencing such disorders while additional research on genetic alterations and their reflection on clinical phenotypes should be implemented in the near future. In some cases, a correct diagnosis should be made within an interdisciplinary clinical setting by complementary competences and follow-up visits to evaluate pathology evolution.
Collapse
Affiliation(s)
| | - Sabrina Danti
- Psychology Unit, Hospital of Pontedera, Pontedera, Italy
| | - Angelo Nuti
- Neurology Unit, Versilia Hospital, Lido di Camaiore, Italy
| | | | - Davide M Cammisuli
- Department of Biomedical and Clinical Sciences "L. Sacco", University of Milan (La Statale), Milan, Italy.
| |
Collapse
|
16
|
Sellami L, Rucheton B, Ben Younes I, Camuzat A, Saracino D, Rinaldi D, Epelbaum S, Azuar C, Levy R, Auriacombe S, Hannequin D, Pariente J, Barbier M, Boutoleau-Bretonnière C, Couratier P, Pasquier F, Deramecourt V, Sauvée M, Sarazin M, Lagarde J, Roué-Jagot C, Forlani S, Jornea L, David I, LeGuern E, Dubois B, Brice A, Clot F, Lamari F, Le Ber I. Plasma progranulin levels for frontotemporal dementia in clinical practice: a 10-year French experience. Neurobiol Aging 2020; 91:167.e1-167.e9. [DOI: 10.1016/j.neurobiolaging.2020.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/12/2022]
|
17
|
Yliranta A, Jehkonen M. Limb and face apraxias in frontotemporal dementia: A systematic scoping review. Cortex 2020; 129:529-547. [PMID: 32418629 DOI: 10.1016/j.cortex.2020.03.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/12/2020] [Accepted: 03/30/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE To investigate the literature for frequencies, profiles and neural correlates of limb and face apraxias in frontotemporal dementia (FTD). METHOD The search conducted in Ovid Medline, PsycINFO and Scopus yielded 487 non-duplicate records, and 43 were included in the final analysis. RESULTS Apraxias are evident in diverse forms in all clinical variants of FTD within the first four years of the disease. Face apraxia and productive limb apraxia co-occur in the behavioural and nonfluent variants. The logopenic variant resembles Alzheimer's disease in terms of pronounced parietal limb apraxia and absence of face apraxia. The semantic variant exhibits conceptual praxis deficits together with relatively preserved imitation skills. Concerning the genetic variants of FTD, productive limb apraxia is common among carriers of the progranulin gene mutation, and subtle gestural alterations have been documented among carriers of the chromosome 9 open reading frame 72 gene mutation before the expected disease onset. The data on neural correlations suggest that the breakdown of praxis results from bilateral cortical and subcortical damage in FTD and that Alzheimer-type pathology of the cerebrospinal fluid increases the severity of limb apraxia in all of the variants. Face apraxia correlates with degeneration of the medial and superior frontal cortices. CONCLUSIONS Each of the clinical variants of FTD exhibits a characteristic profile of apraxias that may support early differentiation between the variants and from Alzheimer's disease. However, the screening procedures developed for stroke populations seem insufficient, and a multifaceted assessment tool is needed. Although valid and practical tests already exist for dementia populations, a concise selection of test items that covers all of the critical domains is called for.
Collapse
Affiliation(s)
- Aino Yliranta
- Neurology Clinic, Lapland Central Hospital, Rovaniemi, Finland; Faculty of Social Sciences, Tampere University, Tampere, Finland.
| | - Mervi Jehkonen
- Faculty of Social Sciences, Tampere University, Tampere, Finland.
| |
Collapse
|
18
|
Guven G, Bilgic B, Tufekcioglu Z, Erginel Unaltuna N, Hanagasi H, Gurvit H, Singleton A, Hardy J, Emre M, Gulec C, Bras J, Guerreiro R, Lohmann E. Peripheral GRN mRNA and Serum Progranulin Levels as a Potential Indicator for Both the Presence of Splice Site Mutations and Individuals at Risk for Frontotemporal Dementia. J Alzheimers Dis 2020; 67:159-167. [PMID: 30475763 DOI: 10.3233/jad-180599] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Progranulin (GRN) gene mutations are a major cause of frontotemporal dementia (FTD). Most mutations identified to date are null mutations, which are predicted to cause the pathology via haploinsufficiency. Decreased peripheral progranulin protein (PGRN) levels are associated with the presence of GRN null mutations and are accepted as reliable biomarkers. In this study, our aim was to test whether the presence of specific GRN splice site mutations (c.- 8+2T>G and c.708+6_9del), could be predicted by peripheral mRNA or protein GRN levels, by studying affected and asymptomatic individuals from FTD families. We also tested four missense GRN variants to assess if altered GRN levels depended on the type of mutation.Our results confirmed a reduction in both mRNA and protein PGRN levels in the splice site mutation carriers, which is consistent with previous reports for null mutations. Our results also suggested that both decreased peripheral GRN mRNA and serum PGRN levels indicate the presence of pathogenic mutations in affected individuals, and identify the asymptomatic individuals at risk, without previous knowledge of genetic status. Both inferences suggest a potential use of peripheral GRN mRNA or serum PGRN levels as biomarkers for families with FTD.
Collapse
Affiliation(s)
- Gamze Guven
- Department of Genetics, Aziz Sancar Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Başar Bilgic
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Zeynep Tufekcioglu
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Nihan Erginel Unaltuna
- Department of Genetics, Aziz Sancar Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Hasmet Hanagasi
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hakan Gurvit
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Murat Emre
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Cagri Gulec
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Jose Bras
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK.,Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal.,UK Dementia Research Institute at UCL (UK DRI), London, UK
| | - Rita Guerreiro
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK.,Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal.,UK Dementia Research Institute at UCL (UK DRI), London, UK
| | - Ebba Lohmann
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany.,DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| |
Collapse
|
19
|
Coppola C, Oliva M, Saracino D, Pappatà S, Zampella E, Cimini S, Ricci M, Giaccone G, Di Iorio G, Rossi G. One novel GRN null mutation, two different aphasia phenotypes. Neurobiol Aging 2019; 87:141.e9-141.e14. [PMID: 31837909 DOI: 10.1016/j.neurobiolaging.2019.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/04/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022]
Abstract
Progranulin gene (GRN) mutations are among the leading causes of frontotemporal lobar degeneration, a group of neurodegenerative diseases characterized by remarkable clinical heterogeneity. In this article, we report the new GRN 708+4A>T splicing mutation, identified in 2 siblings of a family with several members affected by cognitive, behavioral, and motor disorders. Plasma progranulin dosage and GRN expression analysis, together with in silico prediction studies, supported the pathogenicity of the mutation. Both the patients displayed a clinical syndrome in which language impairment was largely predominant. However, motor speech deficits were the major feature in one case, diagnosed as progressive nonfluent aphasia, whereas marked semantic alterations were present in the other, whose clinical phenotype was in favor of a mixed aphasia. The profile of neuroanatomical alterations from imaging studies was in line with the clinical phenotypes. Therefore, also this novel GRN mutation is associated with haploinsufficiency and phenotypic heterogeneity, which are both typical features of progranulinopathies.
Collapse
Affiliation(s)
- Cinzia Coppola
- Second Division of Neurology, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Mariano Oliva
- Second Division of Neurology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Dario Saracino
- Second Division of Neurology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Sabina Pappatà
- Department of Biomedical Sciences, Institute of Biostructure and Bioimaging, National Council of Research, Naples, Italy
| | - Emilia Zampella
- Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Sara Cimini
- Division of Neurology V - Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Martina Ricci
- Division of Neurology V - Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giorgio Giaccone
- Division of Neurology V - Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Di Iorio
- Second Division of Neurology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giacomina Rossi
- Division of Neurology V - Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
20
|
Mao Q, Zheng X, Gefen T, Rogalski E, Spencer CL, Rademakers R, Fought AJ, Kohler M, Weintraub S, Xia H, Mesulam MM, Bigio EH. FTLD-TDP With and Without GRN Mutations Cause Different Patterns of CA1 Pathology. J Neuropathol Exp Neurol 2019; 78:844-853. [PMID: 31361008 PMCID: PMC7967835 DOI: 10.1093/jnen/nlz059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 06/03/2019] [Accepted: 06/20/2019] [Indexed: 12/12/2022] Open
Abstract
Heterozygous loss-of-function mutations in the GRN gene lead to progranulin (PGRN) haploinsufficiency and cause frontotemporal lobar degeneration with TDP-43 pathology type A (FTLD-TDP type A). PGRN is a highly conserved, secreted glycoprotein and functions in the central nervous system as a key modulator of microglial function. Hence, altered microglial function caused by PGRN deficiency may be tied to the pathogenesis of FTLD-TDP. Our previous studies showed that haploinsufficiency of GRN mutations extends to microglial PGRN expression in the hippocampal CA1 region. In this study, we found that the CA1 sector was associated with less neuronal loss and more frequent TDP-43 inclusions in FTLD-TDP type A cases with GRN mutations than in sporadic cases. In addition, the CA1 region in GRN mutation cases contained more rod-like microglia, which also had reduced PGRN expression. These findings suggest that the profile of TDP-43 inclusions, neuronal number, and microgliosis in the CA1 sector of FTLD-TDP type A cases may be influenced by GRN gene expression status.
Collapse
Affiliation(s)
- Qinwen Mao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Xiaojing Zheng
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, P.R. China
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Callen L Spencer
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Angela J Fought
- Division of Biostatistics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine
| | | | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi, P.R. China
| | - Marek-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Eileen H Bigio
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
21
|
Van Mossevelde S, Engelborghs S, van der Zee J, Van Broeckhoven C. Genotype-phenotype links in frontotemporal lobar degeneration. Nat Rev Neurol 2019; 14:363-378. [PMID: 29777184 DOI: 10.1038/s41582-018-0009-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) represents a group of neurodegenerative brain diseases with highly heterogeneous clinical, neuropathological and genetic characteristics. This high degree of heterogeneity results from the presence of several different underlying molecular disease processes; consequently, it is unlikely that all patients with FTLD will benefit from a single therapy. Therapeutic strategies for FTLD are currently being explored, and tools are urgently needed that enable the selection of patients who are the most likely to benefit from a particular therapy. Definition of the phenotypic characteristics in patients with different FTLD subtypes that share the same underlying disease processes would assist in the stratification of patients into homogeneous groups. The most common subtype of FTLD is characterized by TAR DNA-binding protein 43 (TDP43) pathology (FTLD-TDP). In this group, pathogenic mutations have been identified in four genes: C9orf72, GRN, TBK1 and VCP. Here, we provide a comprehensive overview of the phenotypic characteristics of patients with FTLD-TDP, highlighting shared features and differences among groups of patients who have a pathogenic mutation in one of these four genes.
Collapse
Affiliation(s)
- Sara Van Mossevelde
- Neurodegenerative Brain Diseases Group, VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp, Middelheim and Hoge Beuken, Antwerp, Belgium.,Department of Neurology and Memory Clinic, University Hospital Antwerp, Edegem, Belgium
| | - Sebastiaan Engelborghs
- Institute Born-Bunge, UAntwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Network Antwerp, Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Julie van der Zee
- Neurodegenerative Brain Diseases Group, VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium.,Institute Born-Bunge, UAntwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, VIB-UAntwerp Center for Molecular Neurology, Antwerp, Belgium. .,Institute Born-Bunge, UAntwerp, Antwerp, Belgium.
| |
Collapse
|
22
|
Ciani M, Bonvicini C, Scassellati C, Carrara M, Maj C, Fostinelli S, Binetti G, Ghidoni R, Benussi L. The Missing Heritability of Sporadic Frontotemporal Dementia: New Insights from Rare Variants in Neurodegenerative Candidate Genes. Int J Mol Sci 2019; 20:ijms20163903. [PMID: 31405128 PMCID: PMC6721049 DOI: 10.3390/ijms20163903] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/02/2019] [Accepted: 08/08/2019] [Indexed: 12/13/2022] Open
Abstract
Frontotemporal dementia (FTD) is a common form of dementia among early-onset cases. Several genetic factors for FTD have been revealed, but a large proportion of FTD cases still have an unidentified genetic origin. Recent studies highlighted common pathobiological mechanisms among neurodegenerative diseases. In the present study, we investigated a panel of candidate genes, previously described to be associated with FTD and/or other neurodegenerative diseases by targeted next generation sequencing (NGS). We focused our study on sporadic FTD (sFTD), devoid of disease-causing mutations in GRN, MAPT and C9orf72. Since genetic factors have a substantially higher pathogenetic contribution in early onset patients than in late onset dementia, we selected patients with early onset (<65 years). Our study revealed that, in 50% of patients, rare missense potentially pathogenetic variants in genes previously associated with Alzheimer's disease, Parkinson disease, amyotrophic lateral sclerosis and Lewy body dementia (GBA, ABCA7, PARK7, FUS, SORL1, LRRK2, ALS2), confirming genetic pleiotropy in neurodegeneration. In parallel, a synergic genetic effect on FTD is suggested by the presence of variants in five different genes in one single patient. Further studies employing genome-wide approaches might highlight pathogenic variants in novel genes that explain the still missing heritability of FTD.
Collapse
Affiliation(s)
- Miriam Ciani
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Cristian Bonvicini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Catia Scassellati
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Matteo Carrara
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Carlo Maj
- Institute of Genomic Statistics and Bioinformatics, University of Bonn, 53127 Bonn, Germany
| | - Silvia Fostinelli
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Giuliano Binetti
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy.
| |
Collapse
|
23
|
Novel GRN mutations in Koreans with Alzheimer’s disease. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0038-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
24
|
Lee SE, Sias AC, Kosik EL, Flagan TM, Deng J, Chu SA, Brown JA, Vidovszky AA, Ramos EM, Gorno-Tempini ML, Karydas AM, Coppola G, Geschwind DH, Rademakers R, Boeve BF, Boxer AL, Rosen HJ, Miller BL, Seeley WW. Thalamo-cortical network hyperconnectivity in preclinical progranulin mutation carriers. Neuroimage Clin 2019; 22:101751. [PMID: 30921613 PMCID: PMC6438992 DOI: 10.1016/j.nicl.2019.101751] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/02/2019] [Accepted: 03/09/2019] [Indexed: 12/12/2022]
Abstract
Mutations in progranulin (GRN) cause heterogeneous clinical syndromes, including behavioral variant frontotemporal dementia (bvFTD), primary progressive aphasia (PPA), corticobasal syndrome (CBS) and Alzheimer-type dementia (AD-type dementia). Human studies have shown that presymptomatic GRN carriers feature reduced connectivity in the salience network, a system targeted in bvFTD. Mice with homozygous deletion of GRN, in contrast, show thalamo-cortical hypersynchrony due to aberrant pruning of inhibitory synapses onto thalamo-cortical projection neurons. No studies have systematically explored the intrinsic connectivity networks (ICNs) targeted by the four GRN-associated clinical syndromes, or have forged clear links between human and mouse model findings. We compared 17 preclinical GRN carriers (14 "presymptomatic" clinically normal and three "prodromal" with mild cognitive symptoms) to healthy controls to assess for differences in cognitive testing and gray matter volume. Using task-free fMRI, we assessed connectivity in the salience network, a non-fluent variant primary progressive aphasia network (nfvPPA), the perirolandic network (CBS), and the default mode network (AD-type dementia). GRN carriers and controls showed similar performance on cognitive testing. Although carriers showed little evidence of brain atrophy, markedly enhanced connectivity emerged in all four networks, and thalamo-cortical hyperconnectivity stood out as a unifying feature. Voxelwise assessment of whole brain degree centrality, an unbiased graph theoretical connectivity metric, confirmed thalamic hyperconnectivity. These results show that human GRN disease and the prevailing GRN mouse model share a thalamo-cortical network hypersynchrony phenotype. Longitudinal studies will determine whether this network physiology represents a compensatory response as carriers approach symptom onset, or an early and sustained preclinical manifestation of lifelong progranulin haploinsufficiency.
Collapse
Affiliation(s)
- Suzee E Lee
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States.
| | - Ana C Sias
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Eena L Kosik
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Taru M Flagan
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Jersey Deng
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Stephanie A Chu
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Jesse A Brown
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Anna A Vidovszky
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Eliana Marisa Ramos
- University of California, Neurobehavior Division, Department of Neurology, Los Angeles, United States
| | - Maria Luisa Gorno-Tempini
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Anna M Karydas
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Giovanni Coppola
- University of California, Neurobehavior Division, Department of Neurology, Los Angeles, United States
| | - Daniel H Geschwind
- University of California, Neurobehavior Division, Department of Neurology, Los Angeles, United States
| | - Rosa Rademakers
- Mayo Clinic Jacksonville, Department of Neuroscience, Jacksonville, United States
| | - Bradley F Boeve
- Mayo Clinic, Department of Neurology, Rochester, United States
| | - Adam L Boxer
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Howard J Rosen
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Bruce L Miller
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - William W Seeley
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States; University of California, Department of Pathology, San Francisco, United States
| |
Collapse
|
25
|
Cysteine-rich granulin-3 rapidly promotes amyloid-β fibrils in both redox states. Biochem J 2019; 476:859-873. [PMID: 30782973 DOI: 10.1042/bcj20180916] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/07/2019] [Accepted: 02/19/2019] [Indexed: 01/03/2023]
Abstract
Granulins (GRNs 1-7) are cysteine-rich proteolytic products of progranulin (PGRN) that have recently been implicated in neurodegenerative diseases including frontotemporal dementia (FTD) and Alzheimer's disease (AD). Their precise mechanism in these pathologies remains uncertain, but both inflammatory and lysosomal roles have been observed for GRNs. Among the seven GRNs, GRN-3 is well characterized and is implicated within the context of FTD. However, the relationship between GRN-3 and amyloid-β (Aβ), a protein relevant in AD pathology, has not yet been explored. To gain insight into this mechanism, we investigated the effect of both oxidized and reduced GRN-3 on Aβ aggregation and found that both GRN-3 (oxidized) and rGRN-3 (reduced) bind to monomeric and oligomeric Aβ42 to promote rapid fibril formation with subtle rate differences. As low molecular weight oligomers of Aβ are well-established neurotoxins, rapid promotion of fibrils by GRN-3 mitigates Aβ42-induced cellular apoptosis. These data provide valuable insights in understanding GRN-3's ability to modulate Aβ-induced toxicity under redox control and presents a new perspective toward AD pathology. These results also prompt further investigation into the role(s) of other GRNs in AD pathogenesis.
Collapse
|
26
|
Nicolas G, Veltman JA. The role of de novo mutations in adult-onset neurodegenerative disorders. Acta Neuropathol 2019; 137:183-207. [PMID: 30478624 PMCID: PMC6513904 DOI: 10.1007/s00401-018-1939-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 12/13/2022]
Abstract
The genetic underpinnings of the most common adult-onset neurodegenerative disorders (AOND) are complex in majority of the cases. In some families, however, the disease can be inherited in a Mendelian fashion as an autosomal-dominant trait. Next to that, patients carrying mutations in the same disease genes have been reported despite a negative family history. Although challenging to demonstrate due to the late onset of the disease in most cases, the occurrence of de novo mutations can explain this sporadic presentation, as demonstrated for severe neurodevelopmental disorders. Exome or genome sequencing of patient-parent trios allows a hypothesis-free study of the role of de novo mutations in AOND and the discovery of novel disease genes. Another hypothesis that may explain a proportion of sporadic AOND cases is the occurrence of a de novo mutation after the fertilization of the oocyte (post-zygotic mutation) or even as a late-somatic mutation, restricted to the brain. Such somatic mutation hypothesis, that can be tested with the use of novel sequencing technologies, is fully compatible with the seeding and spreading mechanisms of the pathological proteins identified in most of these disorders. We review here the current knowledge and future perspectives on de novo mutations in known and novel candidate genes identified in the most common AONDs such as Alzheimer's disease, Parkinson's disease, the frontotemporal lobar degeneration spectrum and Prion disorders. Also, we review the first lessons learned from recent genomic studies of control and diseased brains and the challenges which remain to be addressed.
Collapse
Affiliation(s)
- Gaël Nicolas
- Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, 22, Boulevard Gambetta, 76000, 76031, Rouen Cedex, France.
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Joris A Veltman
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
27
|
Abstract
Frontotemporal dementia (FTD) is the second commonest cause of young onset dementia. Our understanding of FTD and its related syndromes has advanced significantly in recent years. Among the most prominent areas of progress is the overlap between FTD, MND, and other neurodegenerative conditions at a clinicopathologic and genetic level. In parallel major advances in neuroimaging techniques, the discovery of new genetic mutations as well as the development of potential biomarkers may serve to further expand knowledge of the biologic processes at play in FTD and may in turn propel research toward identifying curative and preventative pharmacologic therapies. The aim of this chapter is to discuss the clinical, pathologic, and genetic complexities of FTD and related disorders.
Collapse
Affiliation(s)
- Emma M Devenney
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia
| | - Rebekah M Ahmed
- Department of Clinical Neuroscience, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - John R Hodges
- Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
28
|
Kim EJ, Kim YE, Jang JH, Cho EH, Na DL, Seo SW, Jung NY, Jeong JH, Kwon JC, Park KH, Park KW, Lee JH, Roh JH, Kim HJ, Yoon SJ, Choi SH, Jang JW, Ki CS, Kim SH. Analysis of frontotemporal dementia, amyotrophic lateral sclerosis, and other dementia-related genes in 107 Korean patients with frontotemporal dementia. Neurobiol Aging 2018; 72:186.e1-186.e7. [PMID: 30054184 DOI: 10.1016/j.neurobiolaging.2018.06.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/16/2018] [Accepted: 06/24/2018] [Indexed: 11/15/2022]
Abstract
To identify pathogenic variants in 107 Korean patients with sporadic frontotemporal dementia (FTD), 46 genes related to FTD, amyotrophic lateral sclerosis, and other dementias were screened by next-generation sequencing. Hexanucleotide repeats in C9orf72 gene were also tested by repeat-primed polymerase chain reaction. Next-generation sequencing revealed one known pathogenic variant (c.708+1G>A) in the GRN gene in a patient with behavioral variant FTD (bvFTD). In addition, a novel in-frame deletion (c.2675_2683del) in the CSF1R gene was identified in a patient with bvFTD who had severe bifrontal atrophy with frontal subcortical white matter changes. Novel compound heterozygous variants in the AARS2 gene, c.1040+1G>A and c.636G>A (p.Met212Ile), were found in a patient with bvFTD. Forty-six variants of uncertain significance were detected in other patients. None of the patients had expanded hexanucleotide repeats in C9orf72. These results show that pathogenic variants of known FTD genes are rare in Korean FTD patients but the CSF1R and AARS2 genes should be screened for a genetic diagnosis of FTD or other dementias.
Collapse
Affiliation(s)
- Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Young-Eun Kim
- Department of Laboratory Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Ja-Hyun Jang
- Green Cross Genome, Yongin, Gyeonggi-do, Republic of Korea
| | - Eun-Hae Cho
- Green Cross Genome, Yongin, Gyeonggi-do, Republic of Korea
| | - Duk L Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Na-Yeon Jung
- Department of Neurology, Pusan National University Yangsan Hospital, Research Institute for Convergence of Biomedical Science and Technology, Busan, Republic of Korea
| | - Jee H Jeong
- Department of Neurology, Ewha Womans University Hospital, Seoul, Republic of Korea
| | - Jay C Kwon
- Department of Neurology, Changwon Fatima Hospital, Changwon, Gyeongsangnam-do, Republic of Korea
| | - Kee Hyung Park
- Department of Neurology, Gachon University Gil Hospital, Incheon, Republic of Korea
| | - Kyung Won Park
- Department of Neurology, Dong-A Medical Center, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jee Hoon Roh
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee-Jin Kim
- Department of Neurology, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Soo Jin Yoon
- Department of Neurology, Eulgi University Hospital, Daejeon, Republic of Korea
| | - Seong Hye Choi
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Jae-Won Jang
- Department of Neurology, Kangwon National University Hospital, Chuncheon, Republic of Korea
| | - Chang-Seok Ki
- Green Cross Genome, Yongin, Gyeonggi-do, Republic of Korea.
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Kuuluvainen L, Pöyhönen M, Pasanen P, Siitonen M, Rummukainen J, Tienari PJ, Paetau A, Myllykangas L. A Novel Loss-of-Function GRN Mutation p.(Tyr229*): Clinical and Neuropathological Features. J Alzheimers Dis 2018; 55:1167-1174. [PMID: 27767988 DOI: 10.3233/jad-160647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mutations in the progranulin (GRN) gene represent about 5-10% of frontotemporal lobar degeneration (FTLD). We describe a proband with a novel GRN mutation c.687T>A, p.(Tyr229*), presenting with dyspraxia, dysgraphia, and dysphasia at the age of 60 and a very severe FTLD neuropathological phenotype with TDP43 inclusions. The nephew of the proband had signs of dementia and personality changes at the age of 60 and showed similar but milder FTLD pathology. Three other family members had had early-onset dementia. Gene expression studies showed decreased GRN gene expression in mutation carriers' blood samples. In conclusion, we describe a novel GRN, p.(Tyr229*) mutation, resulting in haploinsufficiency of GRN and a severe neuropathologic FTLD phenotype.
Collapse
Affiliation(s)
- Liina Kuuluvainen
- Department of Clinical Genetics, Helsinki University Central Hospital and Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Minna Pöyhönen
- Department of Clinical Genetics, Helsinki University Central Hospital and Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Petra Pasanen
- Department of Medical Biochemistry and Genetics, Institute of Biomedicine, University of Turku, and Tyks Microbiology and Genetics, Department of Medical Genetics, Turku University Hospital, Turku, Finland
| | - Maija Siitonen
- Department of Medical Biochemistry and Genetics, Institute of Biomedicine, University of Turku, and Tyks Microbiology and Genetics, Department of Medical Genetics, Turku University Hospital, Turku, Finland
| | - Jaana Rummukainen
- Department of Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Pentti J Tienari
- Department of Neurology, Helsinki University Hospital, and Molecular Neurology, Research Programs Unit, Biomedicum, University of Helsinki, Helsinki, Finland
| | - Anders Paetau
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki, Finland
| | - Liisa Myllykangas
- Department of Pathology, University of Helsinki and HUSLAB, Helsinki, Finland
| |
Collapse
|
30
|
Fujita K, Chen X, Homma H, Tagawa K, Amano M, Saito A, Imoto S, Akatsu H, Hashizume Y, Kaibuchi K, Miyano S, Okazawa H. Targeting Tyro3 ameliorates a model of PGRN-mutant FTLD-TDP via tau-mediated synaptic pathology. Nat Commun 2018; 9:433. [PMID: 29382817 PMCID: PMC5789822 DOI: 10.1038/s41467-018-02821-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Abstract
Mutations in the progranulin (PGRN) gene cause a tau pathology-negative and TDP43 pathology-positive form of frontotemporal lobar degeneration (FTLD-TDP). We generated a knock-in mouse harboring the R504X mutation (PGRN-KI). Phosphoproteomic analysis of this model revealed activation of signaling pathways connecting PKC and MAPK to tau prior to TDP43 aggregation and cognitive impairments, and identified PKCα as the kinase responsible for the early-stage tau phosphorylation at Ser203. Disinhibition of Gas6 binding to Tyro3 due to PGRN reduction results in activation of PKCα via PLCγ, inducing tau phosphorylation at Ser203, mislocalization of tau to dendritic spines, and spine loss. Administration of a PKC inhibitor, B-Raf inhibitor, or knockdown of molecules in the Gas6-Tyro3-tau axis rescues spine loss and cognitive impairment of PGRN-KI mice. Collectively, these results suggest that targeting of early-stage and aggregation-independent tau signaling represents a promising therapeutic strategy for this disease.
Collapse
Affiliation(s)
- Kyota Fujita
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Xigui Chen
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Hidenori Homma
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kazuhiko Tagawa
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Mutsuki Amano
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65, Tsurumai, Showa, Nagoya, Aichi, 466-8550, Japan
| | - Ayumu Saito
- Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Seiya Imoto
- Health Intelligence Center, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Hiroyasu Akatsu
- Department of Medicine for Aging in Place and Community-Based Medical Education, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, 467-8601, Japan
| | - Yoshio Hashizume
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Kozo Kaibuchi
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, 65, Tsurumai, Showa, Nagoya, Aichi, 466-8550, Japan
| | - Satoru Miyano
- Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Hitoshi Okazawa
- Department of Neuropathology, Medical Research Institute and Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan.
| |
Collapse
|
31
|
Sieben A, Van Mossevelde S, Wauters E, Engelborghs S, van der Zee J, Van Langenhove T, Santens P, Praet M, Boon P, Miatton M, Van Hoecke S, Vandenbulcke M, Vandenberghe R, Cras P, Cruts M, De Deyn PP, Van Broeckhoven C, Martin JJ. Extended FTLD pedigree segregating a Belgian GRN-null mutation: neuropathological heterogeneity in one family. ALZHEIMERS RESEARCH & THERAPY 2018; 10:7. [PMID: 29370838 PMCID: PMC6389176 DOI: 10.1186/s13195-017-0334-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/20/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND In this paper, we describe the clinical and neuropathological findings of nine members of the Belgian progranulin gene (GRN) founder family. In this family, the loss-of-function mutation IVS1 + 5G > C was identified in 2006. In 2007, a clinical description of the mutation carriers was published that revealed the clinical heterogeneity among IVS1 + 5G > C carriers. We report our comparison of our data with the published clinical and neuropathological characteristics of other GRN mutations as well as other frontotemporal lobar degeneration (FTLD) syndromes, and we present a review of the literature. METHODS For each case, standardized sampling and staining were performed to identify proteinopathies, cerebrovascular disease, and hippocampal sclerosis. RESULTS The neuropathological substrate in the studied family was compatible in all cases with transactive response DNA-binding protein (TDP) proteinopathy type A, as expected. Additionally, most of the cases presented also with primary age-related tauopathy (PART) or mild Alzheimer's disease (AD) neuropathological changes, and one case had extensive Lewy body pathology. An additional finding was the presence of cerebral small vessel changes in every patient in this family. CONCLUSIONS Our data show not only that the IVS1 + 5G > C mutation has an exclusive association with FTLD-TDP type A proteinopathy but also that other proteinopathies can occur and should be looked for. Because the penetrance rate of the clinical phenotype of carriers of GRN mutations is age-dependent, further research is required to investigate the role of co-occurring age-related pathologies such as AD, PART, and cerebral small vessel disease.
Collapse
Affiliation(s)
- Anne Sieben
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Universiteitsplein 1, B-2160, Antwerp, Belgium.,Department of Neurology, Ghent University Hospital, Ghent, Belgium.,Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB , Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Sara Van Mossevelde
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB , Universiteitsplein 1, B-2610, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Netwerk Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium.,Department of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Eline Wauters
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB , Universiteitsplein 1, B-2610, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Sebastiaan Engelborghs
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Universiteitsplein 1, B-2160, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Netwerk Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Julie van der Zee
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB , Universiteitsplein 1, B-2610, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Tim Van Langenhove
- Department of Neurology, Ghent University Hospital, Ghent, Belgium.,Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB , Universiteitsplein 1, B-2610, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Patrick Santens
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Marleen Praet
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - Paul Boon
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Marijke Miatton
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Sofie Van Hoecke
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Mathieu Vandenbulcke
- Department of Neurosciences, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Department of Old Age Psychiatry and Memory Clinic, University Hospitals Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurosciences, Faculty of Medicine, KU Leuven, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Cras
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Universiteitsplein 1, B-2160, Antwerp, Belgium.,Department of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Marc Cruts
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB , Universiteitsplein 1, B-2610, Antwerp, Belgium.,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Peter Paul De Deyn
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Universiteitsplein 1, B-2160, Antwerp, Belgium.,Department of Neurology and Memory Clinic, Hospital Netwerk Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium.,Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB , Universiteitsplein 1, B-2610, Antwerp, Belgium. .,Laboratory of Neurogenetics, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium.
| | - Jean-Jacques Martin
- Institute Born-Bunge, Neuropathology and Laboratory of Neurochemistry and Behavior, University of Antwerp, Universiteitsplein 1, B-2160, Antwerp, Belgium.
| |
Collapse
|
32
|
Chitramuthu BP, Bennett HPJ, Bateman A. Progranulin: a new avenue towards the understanding and treatment of neurodegenerative disease. Brain 2017; 140:3081-3104. [PMID: 29053785 DOI: 10.1093/brain/awx198] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 06/26/2017] [Indexed: 12/14/2022] Open
Abstract
Progranulin, a secreted glycoprotein, is encoded in humans by the single GRN gene. Progranulin consists of seven and a half, tandemly repeated, non-identical copies of the 12 cysteine granulin motif. Many cellular processes and diseases are associated with this unique pleiotropic factor that include, but are not limited to, embryogenesis, tumorigenesis, inflammation, wound repair, neurodegeneration and lysosome function. Haploinsufficiency caused by autosomal dominant mutations within the GRN gene leads to frontotemporal lobar degeneration, a progressive neuronal atrophy that presents in patients as frontotemporal dementia. Frontotemporal dementia is an early onset form of dementia, distinct from Alzheimer's disease. The GRN-related form of frontotemporal lobar dementia is a proteinopathy characterized by the appearance of neuronal inclusions containing ubiquitinated and fragmented TDP-43 (encoded by TARDBP). The neurotrophic and neuro-immunomodulatory properties of progranulin have recently been reported but are still not well understood. Gene delivery of GRN in experimental models of Alzheimer's- and Parkinson's-like diseases inhibits phenotype progression. Here we review what is currently known concerning the molecular function and mechanism of action of progranulin in normal physiological and pathophysiological conditions in both in vitro and in vivo models. The potential therapeutic applications of progranulin in treating neurodegenerative diseases are highlighted.
Collapse
Affiliation(s)
- Babykumari P Chitramuthu
- Endocrine Research Laboratory, Royal Victoria Hospital, and McGill University Health Centre Research Institute, Centre for Translational Biology, Platform in Metabolic Disorders and Complications, 1001 Decarie Boulevard, QC, Canada, H4A 3J1
| | - Hugh P J Bennett
- Endocrine Research Laboratory, Royal Victoria Hospital, and McGill University Health Centre Research Institute, Centre for Translational Biology, Platform in Metabolic Disorders and Complications, 1001 Decarie Boulevard, QC, Canada, H4A 3J1
| | - Andrew Bateman
- Endocrine Research Laboratory, Royal Victoria Hospital, and McGill University Health Centre Research Institute, Centre for Translational Biology, Platform in Metabolic Disorders and Complications, 1001 Decarie Boulevard, QC, Canada, H4A 3J1
| |
Collapse
|
33
|
Mao Q, Wang D, Li Y, Kohler M, Wilson J, Parton Z, Shmaltsuyeva B, Gursel D, Rademakers R, Weintraub S, Mesulam MM, Xia H, Bigio EH. Disease and Region Specificity of Granulin Immunopositivities in Alzheimer Disease and Frontotemporal Lobar Degeneration. J Neuropathol Exp Neurol 2017; 76:957-968. [PMID: 29044416 DOI: 10.1093/jnen/nlx085] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Heterozygous loss-of-function mutations in GRN, the progranulin gene, which result in progranulin (PGRN) protein haploinsufficiency, are a major cause of frontotemporal lobar degeneration with TDP-43 proteinopathy (FTLD-TDP). PGRN is composed of seven and a half repeats of a highly conserved granulin motif that is cleaved to produce the granulin peptides A-G and paragranulin. To better understand the role of PGRN and granulin (Grn) peptides in the pathogenesis of neurodegeneration, we evaluated PGRN/Grn in brains of patients with Alzheimer disease, FTLD-TDP type A with or without GRN mutations, and normal individuals, using a panel of monoclonal antibodies against Grn peptides A-G. In the neocortex, Grn peptide-specific immunostains were observed, for example, membranous Grn E immunopositivity in pyramidal neurons, and Grn C immunopositivity in ramified microglia. In the hippocampus, Grn immunopositivity in the CA1 and CA2 regions showed disease-specific changes in both neurons and microglia. Most interestingly, in FTLD-TDP type A with GRN mutations, there is a 60% decrease in the density of Grn-positive microglia in the hippocampal CA1, suggesting that haploinsufficiency of the GRN mutations also extends to PGRN expression in microglia. This study provides important insights into future studies of the pathogenesis and treatment of FTLD-TDP.
Collapse
Affiliation(s)
- Qinwen Mao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Dongyang Wang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Yanqing Li
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Missia Kohler
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Jayson Wilson
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Zachary Parton
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Bella Shmaltsuyeva
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Demirkan Gursel
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Rosa Rademakers
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Sandra Weintraub
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Marek-Marsel Mesulam
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | - Haibin Xia
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, P.R. China; The Cognitive Neurology and Alzheimer Disease Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University School of Medicine, Chicago, Illinois; Department of Neuroscience, Mayo Clinic, Jacksonville, Florida
| | | |
Collapse
|
34
|
Clinical and genetic analyses of familial and sporadic frontotemporal dementia patients in Southern Italy. Alzheimers Dement 2017; 13:858-869. [PMID: 28264768 DOI: 10.1016/j.jalz.2017.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/17/2016] [Accepted: 01/02/2017] [Indexed: 12/12/2022]
Abstract
INTRODUCTION We investigated the clinical differences between familial and sporadic frontotemporal dementia (FTD), screening for mutations in known FTD genes. METHODS We diagnosed 22 affected individuals belonging to eight families and 43 sporadic cases with FTD in Apulia, Southern Italy, in 2 years. Mutations in common causative FTD genes (GRN, MAPT, VCP, and TARDBP) and C9ORF72 expansions were screened. RESULTS Behavioral variant of FTD was the most common clinical subtype (50% and 69% in familial and sporadic cases, respectively). Social conduct impairment/disinhibition, loss of insight, and inflexibility were the most frequent clinical features observed at onset. One new mutation was identified in GRN in family A. DISCUSSION Disease onset in sporadic FTD was more frequently characterized by a clustering of behavioral symptoms with apathy and loss of personal hygiene. Mutations in common causative FTD genes are not a major cause of familial and sporadic FTD in the Southern Italian population.
Collapse
|
35
|
Mann DMA, Snowden JS. Frontotemporal lobar degeneration: Pathogenesis, pathology and pathways to phenotype. Brain Pathol 2017; 27:723-736. [PMID: 28100023 DOI: 10.1111/bpa.12486] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 01/12/2017] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal Lobar Degeneration (FTLD) is a clinically, pathologically and genetically heterogeneous group of disorders that affect principally the frontal and temporal lobes of the brain. There are three major associated clinical syndromes, behavioral variant frontotemporal dementia (bvFTD), semantic dementia (SD) and progressive non-fluent aphasia (PNFA); three principal histologies, involving tau, TDP-43 and FUS proteins; and mutations in three major genes, MAPT, GRN and C9orf72, along with several other less common gene mutations. All three clinical syndromes can exist separately or in combination with Amyotrophic Lateral Sclerosis (ALS). SD is exclusively a TDP-43 proteinopathy, and PNFA may be so, with both showing tight clinical, histological and genetic inter-relationships. bvFTD is more of a challenge with overlapping histological and genetic features, involvement of any of the three aggregating proteins, and changes in any of the three major genes. However, when ALS is present, all cases show a clear histological phenotype with TDP-43 aggregated proteins, and familial forms are associated with expansions in C9orf72. TDP-43 and FUS are nuclear carrier proteins involved in the regulation of RNA metabolism, whereas tau protein - the product of MAPT - is responsible for the assembly/disassembly of microtubules, which are vital for intracellular transport. Mutations in TDP-43 and FUS genes are linked to clinical ALS rather than FTLD (with or without ALS), suggesting that clinical ALS may be a disorder of RNA metabolism. Conversely, the protein products of GRN and C9orf72, along with those of the other minor genes, appear to form part of the cellular protein degradation machinery. It is possible therefore that FTLD is a reflection of dysfunction within lysosomal/proteasomal systems resulting in failure to remove potentially neurotoxic (TDP-43 and tau) aggregates, which ultimately overwhelm capacity to function. Spread of aggregates along distinct pathways may account for the different clinical phenotypes, and patterns of progression of disease.
Collapse
Affiliation(s)
- David M A Mann
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, M6 8HD, UK
| | - Julie S Snowden
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, M6 8HD, UK.,Cerebral Function Unit, Greater Manchester Neurosciences Centre, Salford Royal Hospital, Stott Lane, Salford, M6 8HD, UK
| |
Collapse
|
36
|
Coppola C, Saracino D, Puoti G, Lus G, Dato C, Le Ber I, Pariente J, Caroppo P, Piccoli E, Tagliavini F, Di Iorio G, Rossi G. A cluster of progranulin C157KfsX97 mutations in Southern Italy: clinical characterization and genetic correlations. Neurobiol Aging 2016; 49:219.e5-219.e13. [PMID: 27814992 DOI: 10.1016/j.neurobiolaging.2016.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/02/2016] [Indexed: 01/08/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) is a group of neurodegenerative diseases displaying high clinical, pathologic, and genetic heterogeneity. Several autosomal dominant progranulin (GRN) mutations have been reported, accounting for 5%-10% of FTLD cases worldwide. In this study, we described the clinical characteristics of 7 Italian patients, 5 with a diagnosis of frontotemporal dementia behavioral variant and 2 of corticobasal syndrome (CBS), carrying the GRN deletion g.101349_101355delCTGCTGT, resulting in the C157KfsX97 null mutation, and hypothesized the existence of a founder effect by means of haplotype sharing analysis. We performed plasma progranulin dosage, GRN gene sequencing, and haplotype sharing study, analyzing 10 short tandem repeat markers, spanning a region of 11.08 Mb flanking GRN on chromosome 17q21. We observed shared alleles among 6 patients for 8 consecutive short tandem repeat markers spanning a 7.29 Mb region. Therefore, also with this particular mutation, the elevated clinical variability described among GRN-mutated FTLD cases is confirmed. Moreover, this is the first study reporting the likely existence of a founder effect for C157KfsX97 mutation in Southern Italy.
Collapse
Affiliation(s)
- Cinzia Coppola
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy.
| | - Dario Saracino
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Gianfranco Puoti
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Giacomo Lus
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Clemente Dato
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Isabelle Le Ber
- Institut du Cerveau et de la Moelle épinière (ICM), INSERM U1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06, UPMC-P6 UMR S 1127 Hôpital de la Pitié-Salpêtrière, Paris, France; AP-HP, Hôpital de la Pitié-Salpêtrière, Centre de Référence des Démences Rares & Fédération des maladies du système nerveux, Paris, France
| | | | - Paola Caroppo
- Division of Neurology V-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Elena Piccoli
- Division of Neurology V-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Fabrizio Tagliavini
- Division of Neurology V-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giuseppe Di Iorio
- Second Division of Neurology, Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences, Second University of Naples, Naples, Italy
| | - Giacomina Rossi
- Division of Neurology V-Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
37
|
Mutation Frequency of the Major Frontotemporal Dementia Genes, MAPT, GRN and C9ORF72 in a Turkish Cohort of Dementia Patients. PLoS One 2016; 11:e0162592. [PMID: 27632209 PMCID: PMC5025192 DOI: 10.1371/journal.pone.0162592] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 08/25/2016] [Indexed: 12/13/2022] Open
Abstract
‘Microtubule-associated protein tau’ (MAPT), ‘granulin’ (GRN) and ‘chromosome 9 open reading frame72’ (C9ORF72) gene mutations are the major known genetic causes of frontotemporal dementia (FTD). Recent studies suggest that mutations in these genes may also be associated with other forms of dementia. Therefore we investigated whether MAPT, GRN and C9ORF72 gene mutations are major contributors to dementia in a random, unselected Turkish cohort of dementia patients. A combination of whole-exome sequencing, Sanger sequencing and fragment analysis/Southern blot was performed in order to identify pathogenic mutations and novel variants in these genes as well as other FTD-related genes such as the ‘charged multivesicular body protein 2B’ (CHMP2B), the ‘FUS RNA binding protein’ (FUS), the ‘TAR DNA binding protein’ (TARDBP), the ‘sequestosome1’ (SQSTM1), and the ‘valosin containing protein’ (VCP). We determined one pathogenic MAPT mutation (c.1906C>T, p.P636L) and one novel missense variant (c.38A>G, p.D13G). In GRN we identified a probably pathogenic TGAG deletion in the splice donor site of exon 6. Three patients were found to carry the GGGGCC expansions in the non-coding region of the C9ORF72 gene. In summary, a complete screening for mutations in MAPT, GRN and C9ORF72 genes revealed a frequency of 5.4% of pathogenic mutations in a random cohort of 93 Turkish index patients with dementia.
Collapse
|
38
|
Gasca-Salas C, Masellis M, Khoo E, Shah BB, Fisman D, Lang AE, Kleiner-Fisman G. Characterization of Movement Disorder Phenomenology in Genetically Proven, Familial Frontotemporal Lobar Degeneration: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0153852. [PMID: 27100392 PMCID: PMC4839564 DOI: 10.1371/journal.pone.0153852] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/05/2016] [Indexed: 12/11/2022] Open
Abstract
Background Mutations in granulin (PGRN) and tau (MAPT), and hexanucleotide repeat expansions near the C9orf72 genes are the most prevalent genetic causes of frontotemporal lobar degeneration. Although behavior, language and movement presentations are common, the relationship between genetic subgroup and movement disorder phenomenology is unclear. Objective We conducted a systematic review and meta-analysis of the literature characterizing the spectrum and prevalence of movement disorders in genetic frontotemporal lobar degeneration. Methods Electronic databases were searched using terms related to frontotemporal lobar degeneration and movement disorders. Articles were included when cases had a proven genetic cause. Study-specific prevalence estimates for clinical features were transformed using Freeman-Tukey arcsine transformation, allowing for pooled estimates of prevalence to be generated using random-effects models. Results The mean age at onset was earlier in those with MAPT mutations compared to PGRN (p<0.001) and C9orf72 (p = 0.024). 66.5% of subjects had an initial non-movement presentation that was most likely a behavioral syndrome (35.7%). At any point during the disease, parkinsonism was the most common movement syndrome reported in 79.8% followed by progressive supranuclear palsy (PSPS) and corticobasal (CBS) syndromes in 12.2% and 10.7%, respectively. The prevalence of movement disorder as initial presentation was higher in MAPT subjects (35.8%) compared to PGRN subjects (10.1). In those with a non-movement presentation, language disorder was more common in PGRN subjects (18.7%) compared to MAPT subjects (5.4%). Summary This represents the first systematic review and meta-analysis of the occurrence of movement disorder phenomenology in genetic frontotemporal lobar degeneration. Standardized prospective collection of clinical information in conjunction with genetic characterization will be crucial for accurate clinico-genetic correlation.
Collapse
Affiliation(s)
- Carmen Gasca-Salas
- The Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson's Disease, TWH, Toronto, Canada
- Department of Medicine, Division of Neurology, University of Toronto, Toronto, Canada
- Centro integral en Neurociencias A.C. (CINAC)/HM Hospitales- Puerta del Sur, CEU-San Pablo University, Madrid, Spain
- * E-mail:
| | - Mario Masellis
- Centro integral en Neurociencias A.C. (CINAC)/HM Hospitales- Puerta del Sur, CEU-San Pablo University, Madrid, Spain
- Cognitive & Movement Disorders Clinic, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Edwin Khoo
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Binit B. Shah
- Department of Neurology, University of Virginia, Charlottesville, Virginia, United States of America
| | - David Fisman
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Anthony E. Lang
- The Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson's Disease, TWH, Toronto, Canada
| | - Galit Kleiner-Fisman
- Department of Medicine, Division of Neurology, University of Toronto, Toronto, Canada
- Jeff and Diane Ross Movement Disorders Clinic, Baycrest Center for Geriatric Health, Toronto, Canada
| |
Collapse
|
39
|
Abstract
Although psychotic features have long been recognized in association with frontotemporal dementia (FTD), recent genetic discoveries enabling further subtyping of FTD have revealed that psychotic symptoms are frequent in some forms of FTD. Hallucinations and delusions can even precede onset of other cognitive or behavioural symptoms in patients with FTD. In this review, we explore the frequency and types of psychotic symptoms reported in patients with FTD, as well as in other neuropsychiatric disorders, to aid practitioners' consideration of these features in the diagnosis of FTD and related disorders.
Collapse
|
40
|
Taghdiri F, Sato C, Ghani M, Moreno D, Rogaeva E, Tartaglia MC. Novel GRN Mutations in Patients with Corticobasal Syndrome. Sci Rep 2016; 6:22913. [PMID: 26961809 PMCID: PMC4785496 DOI: 10.1038/srep22913] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 02/24/2016] [Indexed: 12/13/2022] Open
Abstract
Loss-of-function GRN mutations lead to GRN haploinsufficiency and consequently neurodegeneration with significant heterogeneity in clinical presentation of various syndromes. The aim of this study was to investigate the genetics and clinical features of patients with GRN-related frontotemporal lobar degeneration (FTLD) syndromes. We performed mutation analysis of GRN in 45 unrelated Canadian patients with a broad spectrum of FTLD-like syndromes (mean age at onset of 64.0 ± 11.2 years). In our cohort, two patients were carriers of two novel heterozygous alterations in GRN: 2 bp insertion (c.769-770insCC:p.Q257fs) and 12 bp deletion (c.1009-1020del:p.337-340del). Both patients presented with corticobasal syndrome supported by clinical and radiological findings. The absence of the mutant allele in the RT-PCR product was only observed for the sample with 2 bp insertion in GRN. In contrast, the allele with 12 bp deletion in GRN was not down-regulated at the RNA level and did not segregate with FTLD in the family. Our report extends the evidence for genetic and phenotypic variability in FTLD disorders, and detects a novel pathogenic GRN mutation, carriers of which could eventually help to evaluate the efficacy of different treatments at early stages of dementia.
Collapse
Affiliation(s)
- Foad Taghdiri
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, 60 Leonard avenue, Toronto, ON M5T 2S8, Canada
| | - Christine Sato
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, 60 Leonard avenue, Toronto, ON M5T 2S8, Canada
| | - Mahdi Ghani
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, 60 Leonard avenue, Toronto, ON M5T 2S8, Canada
| | - Danielle Moreno
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, 60 Leonard avenue, Toronto, ON M5T 2S8, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, 60 Leonard avenue, Toronto, ON M5T 2S8, Canada.,Department of Medicine, Division of Neurology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, 60 Leonard avenue, Toronto, ON M5T 2S8, Canada.,Devision of Neurology, University Health Network memory clinic, Toronto Western Hospital, 399 Bathurst street, ON M5T 2S8, Canada
| |
Collapse
|
41
|
Lucchese G. Understanding Neuropsychiatric Diseases, Analyzing the Peptide Sharing between Infectious Agents and the Language-Associated NMDA 2A Protein. Front Psychiatry 2016; 7:60. [PMID: 27148089 PMCID: PMC4827103 DOI: 10.3389/fpsyt.2016.00060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 03/29/2016] [Indexed: 11/13/2022] Open
Abstract
Language disorders and infections may occur together and often concur, to a different extent and via different modalities, in characterizing brain pathologies, such as schizophrenia, autism, epilepsies, bipolar disorders, frontotemporal neurodegeneration, and encephalitis, inter alia. The biological mechanism(s) that might channel language dysfunctions and infections into etiological pathways connected to neuropathologic sequelae are unclear. Searching for molecular link(s) between language disorders and infections, the present study explores the language-associated NMDA 2A subunit for peptide sharing with pathogens that have been described in concomitance with neuropsychiatric diseases. It was found that a vast peptide commonality links the human glutamate ionotropic receptor NMDA 2A subunit to infectious agents. Such a link expands to and interfaces with neuropsychiatric disorders in light of the specific allocation of NMDA 2A gene expression in brain areas related to language functions. The data hint at a possible pathologic scenario based on anti-pathogen immune responses cross-reacting with NMDA 2A in the brain.
Collapse
Affiliation(s)
- Guglielmo Lucchese
- Brain and Language Laboratory, Freie Universität Berlin , Berlin , Germany
| |
Collapse
|
42
|
Wallon D, Nicolas G. Genetica delle demenze degenerative. Neurologia 2015. [DOI: 10.1016/s1634-7072(15)73962-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
43
|
Snowden JS, Adams J, Harris J, Thompson JC, Rollinson S, Richardson A, Jones M, Neary D, Mann DM, Pickering-Brown S. Distinct clinical and pathological phenotypes in frontotemporal dementia associated with MAPT, PGRN and C9orf72 mutations. Amyotroph Lateral Scler Frontotemporal Degener 2015; 16:497-505. [PMID: 26473392 DOI: 10.3109/21678421.2015.1074700] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our objective was to compare the clinical and pathological characteristics of frontotemporal dementia patients with MAPT, GRN and C9orf72 gene mutations. We carried out a cross-sectional comparative study of 74 gene-positive patients (15 MAPT, 17 GRN and 42 C9orf72). Thirty had post mortem pathological data permitting clinico-pathological correlation. MAPT patients were younger than other groups, and showed more frequent behavioural disinhibition, repetitive and stereotyped behaviours, semantic impairment and temporal predominance of atrophy. GRN patients were older at death and more likely to present with non-fluent aphasia. C9orf72 patients alone showed a co-occurrence of ALS. They showed more psychotic symptoms and irrational behaviour, yet were more often reported clinically as socially appropriate and warm. They showed less dietary change than other groups. C9orf72 patients with and without ALS differed only in frequency of psychosis. Greater clinical overlap was observed between GRN and C9orf72 compared to MAPT cases. MAPT cases had tau and GRN and C9orf72, with one exception, TDP-43 pathology. Non-fluent aphasia was linked to TDP subtype A in both GRN and C9orf72 cases and ALS with subtype B. In conclusion, the findings reinforce clinical heterogeneity in FTD and strengthen evidence that genotype influences clinical presentation. Clinical features may inform targeted genetic testing.
Collapse
Affiliation(s)
- Julie S Snowden
- a Manchester Academic Health Sciences Centre, Cerebral Function Unit, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust , Salford.,b Institute of Brain, Behaviour and Mental Health, Faculty of Human and Medical Sciences, University of Manchester , Manchester , UK
| | - Jennifer Adams
- a Manchester Academic Health Sciences Centre, Cerebral Function Unit, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust , Salford.,b Institute of Brain, Behaviour and Mental Health, Faculty of Human and Medical Sciences, University of Manchester , Manchester , UK
| | - Jennifer Harris
- a Manchester Academic Health Sciences Centre, Cerebral Function Unit, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust , Salford.,b Institute of Brain, Behaviour and Mental Health, Faculty of Human and Medical Sciences, University of Manchester , Manchester , UK
| | - Jennifer C Thompson
- a Manchester Academic Health Sciences Centre, Cerebral Function Unit, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust , Salford.,b Institute of Brain, Behaviour and Mental Health, Faculty of Human and Medical Sciences, University of Manchester , Manchester , UK
| | - Sara Rollinson
- b Institute of Brain, Behaviour and Mental Health, Faculty of Human and Medical Sciences, University of Manchester , Manchester , UK
| | - Anna Richardson
- a Manchester Academic Health Sciences Centre, Cerebral Function Unit, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust , Salford.,b Institute of Brain, Behaviour and Mental Health, Faculty of Human and Medical Sciences, University of Manchester , Manchester , UK
| | - Matthew Jones
- a Manchester Academic Health Sciences Centre, Cerebral Function Unit, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust , Salford.,b Institute of Brain, Behaviour and Mental Health, Faculty of Human and Medical Sciences, University of Manchester , Manchester , UK
| | - David Neary
- a Manchester Academic Health Sciences Centre, Cerebral Function Unit, Greater Manchester Neuroscience Centre, Salford Royal NHS Foundation Trust , Salford
| | - David M Mann
- b Institute of Brain, Behaviour and Mental Health, Faculty of Human and Medical Sciences, University of Manchester , Manchester , UK
| | - Stuart Pickering-Brown
- b Institute of Brain, Behaviour and Mental Health, Faculty of Human and Medical Sciences, University of Manchester , Manchester , UK
| |
Collapse
|
44
|
Goldman JS. Genetic testing and counseling in the diagnosis and management of young-onset dementias. Psychiatr Clin North Am 2015; 38:295-308. [PMID: 25998117 DOI: 10.1016/j.psc.2015.01.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Young-onset dementia is hereditary, multifactorial, or sporadic. The most common hereditary dementias include Alzheimer disease, frontotemporal degeneration, Huntington disease, prion diseases, and cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy. Careful attainment of family history assists with diagnosis and determining the likelihood of a genetic cause, and can direct genetic testing. The type of genetic testing depends on confidence of the diagnosis, patient's and affected relatives' symptoms, and the number of disease genes. Single gene, disease-specific gene panels, and large dementia panels are available. Genetic counseling should be given and informed consent obtained. Predictive testing follows the Huntington disease protocol.
Collapse
Affiliation(s)
- Jill S Goldman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 630 West 168th Street, Box 16, New York, NY 10032, USA.
| |
Collapse
|
45
|
Devenney E, Vucic S, Hodges JR, Kiernan MC. Motor neuron disease-frontotemporal dementia: a clinical continuum. Expert Rev Neurother 2015; 15:509-22. [DOI: 10.1586/14737175.2015.1034108] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Fontana F, Siva K, Denti MA. A network of RNA and protein interactions in Fronto Temporal Dementia. Front Mol Neurosci 2015; 8:9. [PMID: 25852467 PMCID: PMC4365750 DOI: 10.3389/fnmol.2015.00009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/25/2015] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disorder characterized by degeneration of the fronto temporal lobes and abnormal protein inclusions. It exhibits a broad clinicopathological spectrum and has been linked to mutations in seven different genes. We will provide a picture, which connects the products of these genes, albeit diverse in nature and function, in a network. Despite the paucity of information available for some of these genes, we believe that RNA processing and post-transcriptional regulation of gene expression might constitute a common theme in the network. Recent studies have unraveled the role of mutations affecting the functions of RNA binding proteins and regulation of microRNAs. This review will combine all the recent findings on genes involved in the pathogenesis of FTD, highlighting the importance of a common network of interactions in order to study and decipher the heterogeneous clinical manifestations associated with FTD. This approach could be helpful for the research of potential therapeutic strategies.
Collapse
Affiliation(s)
- Francesca Fontana
- Laboratory of RNA Biology and Biotechnology, Centre for Integrative Biology, University of TrentoTrento, Italy
| | - Kavitha Siva
- Laboratory of RNA Biology and Biotechnology, Centre for Integrative Biology, University of TrentoTrento, Italy
| | - Michela A. Denti
- Laboratory of RNA Biology and Biotechnology, Centre for Integrative Biology, University of TrentoTrento, Italy
- CNR, Institute of NeurosciencePadua, Italy
| |
Collapse
|
47
|
Ng ASL, Rademakers R, Miller BL. Frontotemporal dementia: a bridge between dementia and neuromuscular disease. Ann N Y Acad Sci 2014; 1338:71-93. [PMID: 25557955 DOI: 10.1111/nyas.12638] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The concept that frontotemporal dementia (FTD) is a purely cortical dementia has largely been refuted by the recognition of its close association with motor neuron disease, and the identification of transactive response DNA-binding protein 43 (TDP-43) as a major pathological substrate underlying both diseases. Genetic findings have transformed this field and revealed connections between disorders that were previous thought clinically unrelated. The discovery that the C9ORF72 locus is responsible for the majority of hereditary FTD, amyotrophic lateral sclerosis (ALS), and FTD-ALS cases and the understanding that repeat-containing RNA plays a crucial role in pathogenesis of both disorders has paved the way for the development of potential biomarkers and therapeutic targets for these devastating diseases. In this review, we summarize the historical aspects leading up to our current understanding of the genetic, clinical, and neuropathological overlap between FTD and ALS, and include brief discussions on chronic traumatic encephalopathy (CTE), given its association with TDP-43 pathology, its associated increased dementia risk, and reports of ALS in CTE patients. In addition, we describe other genetic associations between dementia and neuromuscular disease, such as inclusion body myositis with Paget's disease and FTD.
Collapse
Affiliation(s)
- Adeline S L Ng
- Department of Neurology, National Neuroscience Institute, Tan Tock Seng Hospital, Novena, Singapore
| | | | | |
Collapse
|
48
|
Haapasalo A, Remes AM. Genetic and Molecular Aspects of Frontotemporal Lobar Degeneration. CURRENT GENETIC MEDICINE REPORTS 2014. [DOI: 10.1007/s40142-014-0063-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
49
|
Abstract
Objective:Primitive reflexes and parkinsonian signs are used by clinicians to differentiate among dementias. We reviewed our clinical sample to determine whether primitive reflexes were more prevalent in frontally-based dementias and whether parkinsonian signs were more common in dementia with Lewy bodies (DLB) than in other types of dementia.Design:We retrospectively reviewed charts from 204 patients with dementia who presented for consultation at Baycrest's Ross Memory Clinic between April, 2003, to December, 2007.Results:A greater proportion of subjects with DLB and dementia of the Alzheimer type with cardiovascular disease had primitive reflexes than subjects with frontotemporal dementia (FTD). Primitive reflexes were not positively predictive of FTD or vascular dementia (VaD). Dementia with Lewy bodies subjects were more likely to have parkinsonian signs than the other dementias, and bradykinesia and rigidity were positively predictive of FTD. The palmomental reflex was the most common primitive reflex in the sample, and cogwheeling was the most common parkinsonian sign. There was no significant difference between early- and late-stage groups in presence of primitive reflexes or parkinsonian signs.Conclusions:Primitive reflexes appear not to be clinically discriminative of frontally-based dementias such as FTD and VaD.
Collapse
|
50
|
Affiliation(s)
- Amanda R Mason
- From the Gladstone Institute of Neurological Disease (A.R.M., A.Z., S.F.), San Francisco; the Developmental and Stem Cell Biology Graduate Program (A.R.M.) and the Departments of Neurology (A.Z., S.F.) and Physiology (S.F.), University of California San Francisco; and the Taube/Koret Center for Neurodegenerative Disease Research (S.F.), San Francisco, CA. A.Z. is currently affiliated with Lundbeck, Deerfield, IL
| | - Adam Ziemann
- From the Gladstone Institute of Neurological Disease (A.R.M., A.Z., S.F.), San Francisco; the Developmental and Stem Cell Biology Graduate Program (A.R.M.) and the Departments of Neurology (A.Z., S.F.) and Physiology (S.F.), University of California San Francisco; and the Taube/Koret Center for Neurodegenerative Disease Research (S.F.), San Francisco, CA. A.Z. is currently affiliated with Lundbeck, Deerfield, IL
| | - Steven Finkbeiner
- From the Gladstone Institute of Neurological Disease (A.R.M., A.Z., S.F.), San Francisco; the Developmental and Stem Cell Biology Graduate Program (A.R.M.) and the Departments of Neurology (A.Z., S.F.) and Physiology (S.F.), University of California San Francisco; and the Taube/Koret Center for Neurodegenerative Disease Research (S.F.), San Francisco, CA. A.Z. is currently affiliated with Lundbeck, Deerfield, IL.
| |
Collapse
|