51
|
Motor speech disorders in the nonfluent, semantic and logopenic variants of primary progressive aphasia. Cortex 2021; 140:66-79. [PMID: 33933931 DOI: 10.1016/j.cortex.2021.03.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/12/2021] [Accepted: 03/22/2021] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Motor speech disorders (MSDs) are characteristic for nonfluent primary progressive aphasia (nfvPPA). In primary progressive aphasia (PPA) of the semantic (svPPA) and of the logopenic type (lvPPA), speech motor function is considered typically intact. However, knowledge on the prevalence of MSDs in svPPA and lvPPA is mainly based on studies with a priori knowledge of PPA syndrome diagnosis. This fully blinded retrospective study aims to provide data on the prevalence of all types of MSDs in a large sample of German-speaking patients with different subtypes of PPA. METHOD Two raters, blinded for PPA subtype, independently evaluated connected speech samples for MSD syndrome and severity from 161 patients diagnosed with nfvPPA, svPPA or lvPPA in the database of the German Consortium of Frontotemporal Lobar Degeneration (FTLDc). In case of disagreement, a third experienced rater re-evaluated the speech samples, followed by a consensus procedure. Consensus was reached for 160 patients (74 nfvPPA, 49 svPPA, 37 lvPPA). MAIN RESULTS Across all PPA syndromes, 43.8% of the patients showed MSDs. Patients with nfvPPA demonstrated the highest proportion of MSDs (62.2%), but MSDs were also identified in svPPA (26.5%) and lvPPA (29.7%), respectively. Overall, dysarthria was the most common class of MSDs, followed by apraxia of speech. In addition, we identified speech abnormalities presenting as "syllabic speech", "dysfluent speech", and "adynamic speech". DISCUSSION Our study confirmed MSDs as frequently occurring in PPA. The study also confirmed MSDs to be most common in patients with nfvPPA. However, MSDs were also found in substantial proportions of patients with svPPA and lvPPA. Furthermore, our study identified speech motor deficits that have not received attention in previous studies on PPA. The results are discussed against the background of the existing literature on MSDs in PPA, including theoretical considerations of the neuroanatomical conditions described for each of the different subtypes of PPA.
Collapse
|
52
|
Giunta M, Solje E, Gardoni F, Borroni B, Benussi A. Experimental Disease-Modifying Agents for Frontotemporal Lobar Degeneration. J Exp Pharmacol 2021; 13:359-376. [PMID: 33790662 PMCID: PMC8005747 DOI: 10.2147/jep.s262352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia is a clinically, genetically and pathologically heterogeneous neurodegenerative disorder, enclosing a wide range of different pathological entities, associated with the accumulation of proteins such as tau and TPD-43. Characterized by a high hereditability, mutations in three main genes, MAPT, GRN and C9orf72, can drive the neurodegenerative process. The connection between different genes and proteinopathies through specific mechanisms has shed light on the pathophysiology of the disease, leading to the identification of potential pharmacological targets. New experimental strategies are emerging, in both preclinical and clinical settings, which focus on small molecules rather than gene therapy. In this review, we provide an insight into the aberrant mechanisms leading to FTLD-related proteinopathies and discuss recent therapies with the potential to ameliorate neurodegeneration and disease progression.
Collapse
Affiliation(s)
- Marcello Giunta
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
53
|
Geser F, Jellinger KA, Fellner L, Wenning GK, Yilmazer-Hanke D, Haybaeck J. Emergent creativity in frontotemporal dementia. J Neural Transm (Vienna) 2021; 128:279-293. [PMID: 33709181 DOI: 10.1007/s00702-021-02325-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022]
Abstract
Numerous papers report on connections between creative work and dementing illness, particularly in frontotemporal dementia (FTD), which may combine with motor neuron disease (FTD-MND). However, the emergence of FTD(-MND) patients' de novo artistic activities is rarely reported and underappreciated. Therefore, the present review summarizes relevant case studies' outcomes, capturing creativity's multifaceted nature. Here, we systematically searched for case reports by paying particular attention to the chronological development of individual patients' clinical symptoms, signs, and life events. We synoptically compared the various art domains to the pattern of brain atrophy, the clinical and pathological FTD subtypes. 22 FTD(-MND) patients were identified with creativity occurring either at the same time (41%) or starting after the disease onset (59%); the median lag between the first manifestation of disease and the beginning of creativity was two years. In another five patients, novel artistic activity was developed by a median of 8 years before the start of dementia symptoms. Artistic activity usually evolved over time with a peak in performance, followed by a decline that was further hampered by physical impairment during disease progression. Early on, the themes and objects depicted were often concrete and realistic, but they could become more abstract or symbolic at later stages. Emergent artistic processes may occur early on in the disease process. They appear to be a communication of inner life and may also reflect an attempt of compensation or "self-healing". The relative preservation of primary neocortical areas such as the visual, auditory, or motor cortex may enable the development of artistic activity in the face of degeneration of association cortical areas and subcortical, deeper central nervous system structures. It is crucial to understand the differential loss of function and an individual's creative abilities to implement caregiver-guided, personalized therapeutic strategies such as art therapy.
Collapse
Affiliation(s)
- Felix Geser
- Department of Geriatric Psychiatry, Klinikum Christophsbad, Faurndauer Str. 6-28, 73035, Göppingen, Germany.
| | | | - Lisa Fellner
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor K Wenning
- Division of Neurobiology, Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Deniz Yilmazer-Hanke
- Department of Neurology, Clinical Neuroanatomy, University Hospital, Ulm University, Ulm, Germany
| | - Johannes Haybaeck
- Department of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, Graz, Austria
| |
Collapse
|
54
|
Low YH, Asi Y, Foti SC, Lashley T. Heterogeneous Nuclear Ribonucleoproteins: Implications in Neurological Diseases. Mol Neurobiol 2021; 58:631-646. [PMID: 33000450 PMCID: PMC7843550 DOI: 10.1007/s12035-020-02137-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022]
Abstract
Heterogenous nuclear ribonucleoproteins (hnRNPs) are a complex and functionally diverse family of RNA binding proteins with multifarious roles. They are involved, directly or indirectly, in alternative splicing, transcriptional and translational regulation, stress granule formation, cell cycle regulation, and axonal transport. It is unsurprising, given their heavy involvement in maintaining functional integrity of the cell, that their dysfunction has neurological implications. However, compared to their more established roles in cancer, the evidence of hnRNP implication in neurological diseases is still in its infancy. This review aims to consolidate the evidences for hnRNP involvement in neurological diseases, with a focus on spinal muscular atrophy (SMA), Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), multiple sclerosis (MS), congenital myasthenic syndrome (CMS), and fragile X-associated tremor/ataxia syndrome (FXTAS). Understanding more about hnRNP involvement in neurological diseases can further elucidate the pathomechanisms involved in these diseases and perhaps guide future therapeutic advances.
Collapse
Affiliation(s)
- Yi-Hua Low
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Disorders, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Duke-NUS Medical School, Singapore, Singapore
| | - Yasmine Asi
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Disorders, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
| | - Sandrine C Foti
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Disorders, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Tammaryn Lashley
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Disorders, UCL Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
55
|
Leroy M, Bertoux M, Skrobala E, Mode E, Adnet-Bonte C, Le Ber I, Bombois S, Cassagnaud P, Chen Y, Deramecourt V, Lebert F, Mackowiak MA, Sillaire AR, Wathelet M, Pasquier F, Lebouvier T. Characteristics and progression of patients with frontotemporal dementia in a regional memory clinic network. ALZHEIMERS RESEARCH & THERAPY 2021; 13:19. [PMID: 33419472 PMCID: PMC7796569 DOI: 10.1186/s13195-020-00753-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022]
Abstract
Background Due to heterogeneous clinical presentation, difficult differential diagnosis with Alzheimer’s disease (AD) and psychiatric disorders, and evolving clinical criteria, the epidemiology and natural history of frontotemporal lobar degeneration (FTD) remain elusive. In order to better characterize FTD patients, we relied on the database of a regional memory clinic network with standardized diagnostic procedures and chose AD patients as a comparator. Methods Patients that were first referred to our network between January 2010 and December 2016 and whose last clinical diagnosis was degenerative or vascular dementia were included. Comparisons were conducted between FTD and AD as well as between the different FTD syndromes, divided into language variants (lvFTD), behavioral variant (bvFTD), and FTD with primarily motor symptoms (mFTD). Cognitive progression was estimated with the yearly decline in Mini Mental State Examination (MMSE). Results Among the patients that were referred to our network in the 6-year time span, 690 were ultimately diagnosed with FTD and 18,831 with AD. Patients with FTD syndromes represented 2.6% of all-cause dementias. The age-standardized incidence was 2.90 per 100,000 person-year and incidence peaked between 75 and 79 years. Compared to AD, patients with FTD syndromes had a longer referral delay and delay to diagnosis. Patients with FTD syndromes had a higher MMSE score than AD at first referral while their progression was similar. mFTD patients had the shortest survival while survival in bvFTD, lvFTD, and AD did not significantly differ. FTD patients, especially those with the behavioral variant, received more antidepressants, anxiolytics, and antipsychotics than AD patients. Conclusions FTD syndromes differ with AD in characteristics at baseline, progression rate, and treatment. Despite a broad use of the new diagnostic criteria in an organized memory clinic network, FTD syndromes are longer to diagnose and account for a low proportion of dementia cases, suggesting persistent underdiagnosis. Congruent with recent publications, the late peak of incidence warns against considering FTD as being exclusively a young-onset dementia.
Collapse
Affiliation(s)
- Mélanie Leroy
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | - Maxime Bertoux
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | | | - Elisa Mode
- Univ. Lille, Inserm, CHU Lille, F-59000, Lille, France
| | - Catherine Adnet-Bonte
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | - Isabelle Le Ber
- Sorbonne Université, Inserm U1127, CNRS UMR 7225, Institut du Cerveau (ICM), AP-HP - Hôpital Pitié-Salpêtrière, Paris, France.,Centre de référence des démences rares ou précoces, IM2A, Département de Neurologie, AP-HP - Hôpital Pitié-Salpêtrière, Paris, France
| | - Stéphanie Bombois
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | - Pascaline Cassagnaud
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | - Yaohua Chen
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | - Vincent Deramecourt
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | - Florence Lebert
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | - Marie Anne Mackowiak
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | - Adeline Rollin Sillaire
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | | | - Florence Pasquier
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France
| | - Thibaud Lebouvier
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, CNRMAJ, LiCEND, DistAlz, F-59000, Lille, France.
| | | |
Collapse
|
56
|
Fluid Biomarkers of Frontotemporal Lobar Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:123-139. [PMID: 33433873 DOI: 10.1007/978-3-030-51140-1_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A timely diagnosis of frontotemporal degeneration (FTD) is frequently challenging due to the heterogeneous symptomatology and poor phenotype-pathological correlation. Fluid biomarkers that reflect FTD pathophysiology could be instrumental in both clinical practice and pharmaceutical trials. In recent years, significant progress has been made in developing biomarkers of neurodegenerative diseases: amyloid-β and tau in cerebrospinal fluid (CSF) can be used to exclude Alzheimer's disease, while neurofilament light chain (NfL) is emerging as a promising, albeit nonspecific, marker of neurodegeneration in both CSF and blood. Gene-specific biomarkers such as PGRN in GRN mutation carriers and dipeptide repeat proteins in C9orf72 mutation carriers are potential target engagement markers in genetic FTD trials. Novel techniques capable of measuring very low concentrations of brain-derived proteins in peripheral fluids are facilitating studies of blood biomarkers as a minimally invasive alternative to CSF. A major remaining challenge is the identification of a biomarker that can be used to predict the neuropathological substrate in sporadic FTD patients.
Collapse
|
57
|
Wong CE, Jin LW, Chu YP, Wei WY, Ho PC, Tsai KJ. TDP-43 proteinopathy impairs mRNP granule mediated postsynaptic translation and mRNA metabolism. Theranostics 2021; 11:330-345. [PMID: 33391478 PMCID: PMC7681104 DOI: 10.7150/thno.51004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Local protein synthesis and mRNA metabolism mediated by mRNP granules in the dendrites and the postsynaptic compartment is essential for synaptic remodeling and plasticity in neuronal cells. Dysregulation of these processes caused by TDP-43 proteinopathy leads to neurodegenerative diseases, such as frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Methods: Using biochemical analysis and imaging techniques, including super-resolution microscopy, we provide evidence, for the first time, for the postsynaptic localization of TDP-43 in mammalian synapses and we show that TDP-43 is a component of neuronal mRNP granules. Results: With activity stimulation and various molecular approaches, we further demonstrate activity-dependent mRNP granule dynamics involving disassembly of mRNP granules, release of mRNAs, activation of local protein translation, and the impairment of granule disassembly in cellular, animal and human models of TDP-43 proteinopathy. Conclusion: Our study elucidates the interplay between TDP-43 and neuronal mRNP granules in normal physiology and TDP-43 proteinopathy in the regulation of local protein translation and mRNA metabolism in the postsynaptic compartment.
Collapse
Affiliation(s)
- Chia-En Wong
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, UC Davis Medical Center, California, USA
| | - Yuan-Ping Chu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Yen Wei
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chuan Ho
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
58
|
Neuroprotective Effects of Coffee Bioactive Compounds: A Review. Int J Mol Sci 2020; 22:ijms22010107. [PMID: 33374338 PMCID: PMC7795778 DOI: 10.3390/ijms22010107] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Coffee is one of the most widely consumed beverages worldwide. It is usually identified as a stimulant because of a high content of caffeine. However, caffeine is not the only coffee bioactive component. The coffee beverage is in fact a mixture of a number of bioactive compounds such as polyphenols, especially chlorogenic acids (in green beans) and caffeic acid (in roasted coffee beans), alkaloids (caffeine and trigonelline), and the diterpenes (cafestol and kahweol). Extensive research shows that coffee consumption appears to have beneficial effects on human health. Regular coffee intake may protect from many chronic disorders, including cardiovascular disease, type 2 diabetes, obesity, and some types of cancer. Importantly, coffee consumption seems to be also correlated with a decreased risk of developing some neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease, and dementia. Regular coffee intake may also reduce the risk of stroke. The mechanism underlying these effects is, however, still poorly understood. This review summarizes the current knowledge on the neuroprotective potential of the main bioactive coffee components, i.e., caffeine, chlorogenic acid, caffeic acid, trigonelline, kahweol, and cafestol. Data from both in vitro and in vivo preclinical experiments, including their potential therapeutic applications, are reviewed and discussed. Epidemiological studies and clinical reports on this matter are also described. Moreover, potential molecular mechanism(s) by which coffee bioactive components may provide neuroprotection are reviewed.
Collapse
|
59
|
Frontotemporal Dementia Knowledge Scale: Development and Preliminary Psychometric Properties. Alzheimer Dis Assoc Disord 2020; 34:59-65. [PMID: 31460870 DOI: 10.1097/wad.0000000000000344] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Frontotemporal dementia (FTD) accounts for ∼10% of dementia cases and is the most common cause of early-onset dementia. However, no well-validated instrument currently exists to measure knowledge about FTD. In this study, we used systematic scale development procedures to create a scale to measure knowledge of FTD based on a contemporary understanding of the disease. METHODS Standard scale development methods were used to create items and evaluate their psychometric properties. A total of 72 health care professionals and 102 caregivers of people with FTD responded to items measuring FTD knowledge, general dementia knowledge, crystallized intelligence, experience with FTD, and demographic information. RESULTS The Frontotemporal Dementia Knowledge Scale (FTDKS) contains 18 items that cover key, basic knowledge about FTD in the domains of risk factors, symptoms, course, caregiving, and treatment. The scale uses a 4-point True/False format with a Don't Know option and takes ∼5 minutes to complete. In the current sample the FTDKS had good psychometric properties in terms of reliability and validity. IMPLICATIONS The FTDKS can be used with health care professionals and caregivers of people with FTD to assess their knowledge about the disease. The scale may be useful to evaluate knowledge in clinical care and educational program contexts.
Collapse
|
60
|
The role of hnRNPs in frontotemporal dementia and amyotrophic lateral sclerosis. Acta Neuropathol 2020; 140:599-623. [PMID: 32748079 PMCID: PMC7547044 DOI: 10.1007/s00401-020-02203-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022]
Abstract
Dysregulated RNA metabolism is emerging as a crucially important mechanism underpinning the pathogenesis of frontotemporal dementia (FTD) and the clinically, genetically and pathologically overlapping disorder of amyotrophic lateral sclerosis (ALS). Heterogeneous nuclear ribonucleoproteins (hnRNPs) comprise a family of RNA-binding proteins with diverse, multi-functional roles across all aspects of mRNA processing. The role of these proteins in neurodegeneration is far from understood. Here, we review some of the unifying mechanisms by which hnRNPs have been directly or indirectly linked with FTD/ALS pathogenesis, including their incorporation into pathological inclusions and their best-known roles in pre-mRNA splicing regulation. We also discuss the broader functionalities of hnRNPs including their roles in cryptic exon repression, stress granule assembly and in co-ordinating the DNA damage response, which are all emerging pathogenic themes in both diseases. We then present an integrated model that depicts how a broad-ranging network of pathogenic events can arise from declining levels of functional hnRNPs that are inadequately compensated for by autoregulatory means. Finally, we provide a comprehensive overview of the most functionally relevant cellular roles, in the context of FTD/ALS pathogenesis, for hnRNPs A1-U.
Collapse
|
61
|
Omer A, Patel D, Moran JL, Lian XJ, Di Marco S, Gallouzi IE. Autophagy and heat-shock response impair stress granule assembly during cellular senescence. Mech Ageing Dev 2020; 192:111382. [PMID: 33049246 DOI: 10.1016/j.mad.2020.111382] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/21/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023]
Abstract
Stress granules (SGs) are membraneless organelles formed in response to insult. These granules are related to pathological granules found in age-related neurogenerative diseases such as Parkinson's and Alzheimer's. Previously, we demonstrated that senescent cells, which accumulate with age, exposed to chronic oxidative stress, are unable to form SGs. Here, we show that the senescent cells' inability to form SGs correlates with an upregulation in both the heat-shock response and autophagy pathways, both of which are well-established promoters of SG disassembly. Our data also reveals that the knockdown of HSP70 and ATG5, important components of the heat-shock response and autophagy pathways, respectively, restores the number of SGs formed in senescent cells exposed to chronic oxidative stress. Surprisingly, under these conditions, the depletion of HSP70 or ATG5 did not affect the clearance of these SGs during their recovery from chronic stress. These data reveal that senescent cells possess a unique heat-shock and autophagy-dependent ability to impair the formation of SGs in response to chronic stress, thereby expanding the existing understanding of SG dynamics in senescent cells and their potential contribution to age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Amr Omer
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada.
| | - Devang Patel
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Julian Lucas Moran
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Xian Jin Lian
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Sergio Di Marco
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada
| | - Imed-Eddine Gallouzi
- McGill University, Department of Biochemistry, Rosalind and Morris Goodman Cancer Centre, Montreal, Quebec, Canada.
| |
Collapse
|
62
|
Modelling frontotemporal dementia using patient-derived induced pluripotent stem cells. Mol Cell Neurosci 2020; 109:103553. [PMID: 32956830 DOI: 10.1016/j.mcn.2020.103553] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 08/27/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) describes a group of clinically heterogeneous conditions that frequently affect people under the age of 65 (Le Ber et al., 2013). There are multiple genetic causes of FTD, including coding or splice-site mutations in MAPT, GRN mutations that lead to haploinsufficiency of progranulin protein, and a hexanucleotide GGGGCC repeat expansion in C9ORF72. Pathologically, FTD is characterised by abnormal protein accumulations in neurons and glia. These aggregates can be composed of the microtubule-associated protein tau (observed in FTD with MAPT mutations), the DNA/RNA-binding protein TDP-43 (seen in FTD with mutations in GRN or C9ORF72 repeat expansions) or dipeptide proteins generated by repeat associated non-ATG translation of the C9ORF72 repeat expansion. There are currently no disease-modifying therapies for FTD and the availability of in vitro models that recapitulate pathologies in a disease-relevant cell type would accelerate the development of novel therapeutics. It is now possible to generate patient-specific stem cells through the reprogramming of somatic cells from a patient with a genotype/phenotype of interest into induced pluripotent stem cells (iPSCs). iPSCs can subsequently be differentiated into a plethora of cell types including neurons, astrocytes and microglia. Using this approach has allowed researchers to generate in vitro models of genetic FTD in human cell types that are largely inaccessible during life. In this review we explore the recent progress in the use of iPSCs to model FTD, and consider the merits, limitations and future prospects of this approach.
Collapse
|
63
|
Woollacott IO, Nicholas JM, Heller C, Foiani MS, Moore KM, Russell LL, Paterson RW, Keshavan A, Schott JM, Warren JD, Heslegrave A, Zetterberg H, Rohrer JD. Cerebrospinal Fluid YKL-40 and Chitotriosidase Levels in Frontotemporal Dementia Vary by Clinical, Genetic and Pathological Subtype. Dement Geriatr Cogn Disord 2020; 49:56-76. [PMID: 32344399 PMCID: PMC7513620 DOI: 10.1159/000506282] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Chronic glial dysfunction may contribute to the pathogenesis of frontotemporal dementia (FTD). Cerebrospinal fluid (CSF) levels of glia-derived proteins YKL-40 and chitotriosidase are increased in Alzheimer's disease (AD) but have not been explored in detail across the spectrum of FTD. METHODS We investigated whether CSF YKL-40 and chitotriosidase levels differed between FTD patients and controls, across different clinical and genetic subtypes of FTD, and between individuals with a clinical FTD syndrome due to AD versus non-AD (frontotemporal lobar degeneration, FTLD) pathology (based on CSF neurodegenerative biomarkers). Eighteen healthy controls and 64 people with FTD (behavioural variant FTD, n = 20; primary progressive aphasia [PPA], n = 44: nfvPPA, n = 16, svPPA, n = 11, lvPPA, n = 14, PPA-NOS, n = 3) were included. 10/64 had familial FTD, with mutations in GRN(n = 3), MAPT(n = 4), or C9orf72 (n = 3). 15/64 had neurodegenerative biomarkers consistent with AD pathology. Levels were measured by immunoassay and compared using multiple linear regressions. We also examined relationships of YKL-40 and chitotriosidase with CSF total tau (T-tau), phosphorylated tau 181 (P-tau) and β-amyloid 1-42 (Aβ42), with each other, and with age and disease du-ration. RESULTS CSF YKL-40 and chitotriosidase levels were higher in FTD, particularly lvPPA (both) and nfvPPA (YKL-40), compared with controls. GRN mutation carriers had higher levels of both proteins than controls and C9orf72 expansion carriers, and YKL-40 was higher in MAPT mutation carriers than controls. Individuals with underlying AD pathology had higher YKL-40 and chitotriosidase levels than both controls and those with likely FTLD pathology. CSF YKL-40 and chitotriosidase levels were variably associated with levels of T-tau, P-tau and Aβ42, and with each other, depending on clinical syndrome and underlying pathology. CSF YKL-40 but not chitotriosidase was associated with age, but not disease duration. CONCLUSION CSF YKL-40 and chitotriosidase levels are increased in individuals with clinical FTD syndromes, particularly due to AD pathology. In a preliminary analysis of genetic groups, levels of both proteins are found to be highly elevated in FTD due to GRN mutations, while YKL-40 is increased in individuals with MAPT mutations. As glia-derived protein levels generally correlate with T-tau and P-tau levels, they may reflect the glial response to neurodegeneration in FTLD.
Collapse
Affiliation(s)
- Ione O.C. Woollacott
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Jennifer M. Nicholas
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Carolin Heller
- UK Dementia Research Institute, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Martha S. Foiani
- UK Dementia Research Institute, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Katrina M. Moore
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Lucy L. Russell
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Ross W. Paterson
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Ashvini Keshavan
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Jonathan M. Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Jason D. Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Amanda Heslegrave
- UK Dementia Research Institute, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom
| | - Henrik Zetterberg
- UK Dementia Research Institute, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jonathan D. Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London, United Kingdom,*Dr. Jonathan D. Rohrer, Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square UCL Institute of Neurology, London WC1N 3BG (UK),
| |
Collapse
|
64
|
Ikeda M, Kuwabara T, Takai E, Kasahara H, Furuta M, Sekine A, Makioka K, Yamazaki T, Fujita Y, Nagashima K, Higuchi T, Tsushima Y, Ikeda Y. Increased Neurofilament Light Chain and YKL-40 CSF Levels in One Japanese IBMPFD Patient With VCP R155C Mutation: A Clinical Case Report With CSF Biomarker Analyses. Front Neurol 2020; 11:757. [PMID: 32849216 PMCID: PMC7431878 DOI: 10.3389/fneur.2020.00757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022] Open
Abstract
Inclusion body myopathy (IBM) with Paget's disease of bone (PDB) and frontotemporal dementia (IBMPFD) presents with multiple symptoms and an unknown etiology. Valosin-containing protein (VCP) has been identified as the main causative gene of IBMPFD. However, no studies on neurofilament light chain (NFL) as a cerebrospinal fluid (CSF) marker of axonal neurodegeneration or on YKL-40 as a CSF marker of glial neuroinflammation have been conducted in IBMPFD patients with VCP mutations. A 65-year-old man presented with progressive muscle atrophy and weakness of all limbs, non-fluent aphasia, and changes in personality and behavior. Cerebral MRI revealed bilateral frontal and temporal atrophy. 99mTc-HMDP bone scintigraphy and pelvic CT revealed remodeling changes and active osteoblastic accumulations in the right medial iliac bone. Muscle biopsy demonstrated multiple rimmed vacuoles in muscle cells with myogenic and neurogenic pathological alterations. After the patient was clinically diagnosed with IBMPFD, DNA analysis of the VCP gene revealed a cytosine (C) to thymine (T) (C→ T) mutation, resulting in an amino acid exchange of arginine to cysteine (p.R155C mutation). The CSF levels of NFL at two time points (12 years apart) were higher than those in non-dementia controls (CTR) and Alzheimer's disease (AD); lower than those in frontotemporal dementia with motor neuron disease (FTD-MND); and comparable to those in patients with behavioral variant frontotemporal dementia (bvFTD), progressive supranuclear palsy (PSP), and corticobasal syndrome (CBS). The CSF levels of YKL-40 were comparable at both time points and higher than those in CTR; lower than those in FTD-MND; and comparable to those in bvFTD, PSP, CBS, and AD. The CSF levels of phosphorylated tau 181 (P-Tau) and total tau (T-Tau) were not significantly different from those in CTR and other neurodegenerative diseases, except those in AD, which were significantly elevated. This is the first report that demonstrates increased NFL and YKL-40 CSF levels in an IBMPFD patient with a VCP mutation (p.R155C); NFL and YKL-40 levels were comparable to those in bvFTD, PSP, CBS, and AD and higher than those in CTR. Our results suggest that IBMPFD neuropathology may involve both axonal neurodegeneration and glial neuroinflammation.
Collapse
Affiliation(s)
- Masaki Ikeda
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Takeo Kuwabara
- Department of Neurology, Jobu Hospital for Respiratory Diseases, Maebashi, Japan
| | - Eriko Takai
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroo Kasahara
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Minori Furuta
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akiko Sekine
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kouki Makioka
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tsuneo Yamazaki
- Department of Occupational Therapy, Gunma University Graduate School of Health Sciences, Maebashi, Japan
| | - Yukio Fujita
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kazuaki Nagashima
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tetsuya Higuchi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshio Ikeda
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
65
|
Woollacott IOC, Toomey CE, Strand C, Courtney R, Benson BC, Rohrer JD, Lashley T. Microglial burden, activation and dystrophy patterns in frontotemporal lobar degeneration. J Neuroinflammation 2020; 17:234. [PMID: 32778130 PMCID: PMC7418403 DOI: 10.1186/s12974-020-01907-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Background Microglial dysfunction is implicated in frontotemporal lobar degeneration (FTLD). Although studies have reported excessive microglial activation or senescence (dystrophy) in Alzheimer’s disease (AD), few have explored this in FTLD. We examined regional patterns of microglial burden, activation and dystrophy in sporadic and genetic FTLD, sporadic AD and controls. Methods Immunohistochemistry was performed in frontal and temporal grey and white matter from 50 pathologically confirmed FTLD cases (31 sporadic, 19 genetic: 20 FTLD-tau, 26 FTLD-TDP, four FTLD-FUS), five AD cases and five controls, using markers to detect phagocytic (CD68-positive) and antigen-presenting (CR3/43-positive) microglia, and microglia in general (Iba1-positive). Microglial burden and activation (morphology) were assessed quantitatively for each microglial phenotype. Iba1-positive microglia were assessed semi-quantitatively for dystrophy severity and qualitatively for rod-shaped and hypertrophic morphology. Microglia were compared in each region between FTLD, AD and controls, and between different pathological subtypes of FTLD, including its main subtypes (FTLD-tau, FTLD-TDP, FTLD-FUS), and subtypes of FTLD-tau, FTLD-TDP and genetic FTLD. Microglia were also compared between grey and white matter within each lobe for each group. Results There was a higher burden of phagocytic and antigen-presenting microglia in FTLD and AD cases than controls, but activation was often not increased. Burden was generally higher in white matter than grey matter, but activation was greater in grey matter. However, microglia varied regionally according to FTLD subtype and disease mechanism. Dystrophy was more severe in FTLD and AD than controls, and more severe in white than grey matter, but this also varied regionally and was particularly extensive in FTLD due to progranulin (GRN) mutations. Presence of rod-shaped and hypertrophic microglia also varied by FTLD subtype. Conclusions This study demonstrates regionally variable microglial involvement in FTLD and links this to underlying disease mechanisms. This supports investigation of microglial dysfunction in disease models and consideration of anti-senescence therapies in clinical trials.
Collapse
Affiliation(s)
- Ione O C Woollacott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Christina E Toomey
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Catherine Strand
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Robert Courtney
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Bridget C Benson
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK. .,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK.
| |
Collapse
|
66
|
van der Ende EL, Xiao M, Xu D, Poos JM, Panman JL, Jiskoot LC, Meeter LH, Dopper EG, Papma JM, Heller C, Convery R, Moore K, Bocchetta M, Neason M, Peakman G, Cash DM, Teunissen CE, Graff C, Synofzik M, Moreno F, Finger E, Sánchez-Valle R, Vandenberghe R, Laforce R, Masellis M, Tartaglia MC, Rowe JB, Butler CR, Ducharme S, Gerhard A, Danek A, Levin J, Pijnenburg YA, Otto M, Borroni B, Tagliavini F, de Mendonca A, Santana I, Galimberti D, Seelaar H, Rohrer JD, Worley PF, van Swieten JC. Neuronal pentraxin 2: a synapse-derived CSF biomarker in genetic frontotemporal dementia. J Neurol Neurosurg Psychiatry 2020; 91:612-621. [PMID: 32273328 PMCID: PMC7279197 DOI: 10.1136/jnnp-2019-322493] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/26/2020] [Accepted: 03/02/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Synapse dysfunction is emerging as an early pathological event in frontotemporal dementia (FTD), however biomarkers are lacking. We aimed to investigate the value of cerebrospinal fluid (CSF) neuronal pentraxins (NPTXs), a family of proteins involved in homeostatic synapse plasticity, as novel biomarkers in genetic FTD. METHODS We included 106 presymptomatic and 54 symptomatic carriers of a pathogenic mutation in GRN, C9orf72 or MAPT, and 70 healthy non-carriers participating in the Genetic Frontotemporal dementia Initiative (GENFI), all of whom had at least one CSF sample. We measured CSF concentrations of NPTX2 using an in-house ELISA, and NPTX1 and NPTX receptor (NPTXR) by Western blot. We correlated NPTX2 with corresponding clinical and neuroimaging datasets as well as with CSF neurofilament light chain (NfL) using linear regression analyses. RESULTS Symptomatic mutation carriers had lower NPTX2 concentrations (median 643 pg/mL, IQR (301-872)) than presymptomatic carriers (1003 pg/mL (624-1358), p<0.001) and non-carriers (990 pg/mL (597-1373), p<0.001) (corrected for age). Similar results were found for NPTX1 and NPTXR. Among mutation carriers, NPTX2 concentration correlated with several clinical disease severity measures, NfL and grey matter volume of the frontal, temporal and parietal lobes, insula and whole brain. NPTX2 predicted subsequent decline in phonemic verbal fluency and Clinical Dementia Rating scale plus FTD modules. In longitudinal CSF samples, available in 13 subjects, NPTX2 decreased around symptom onset and in the symptomatic stage. DISCUSSION We conclude that NPTX2 is a promising synapse-derived disease progression biomarker in genetic FTD.
Collapse
Affiliation(s)
- Emma L van der Ende
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Meifang Xiao
- Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Desheng Xu
- Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jackie M Poos
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jessica L Panman
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Lize C Jiskoot
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, Rotterdam, Netherlands
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Lieke H Meeter
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Elise Gp Dopper
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Janne M Papma
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carolin Heller
- Dementia Research Institute, Department of Neurodegenerative Disease, University College London, London, United Kingdom
| | - Rhian Convery
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Katrina Moore
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Martina Bocchetta
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Mollie Neason
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Georgia Peakman
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Caroline Graff
- Karolinska Institutet, Dept NVS, Division of Neurogeriatrics, Bioclinicum, Stockholm, Sweden
- Unit of Hereditary Dementia, Theme Aging, Karolinska University Hospital-Solna, Stockholm, Sweden
| | - Matthis Synofzik
- German Center for Neurodegenerative Diseases (DZNE), University of Tübingen, Tübingen, Germany
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, Tübingen, Germany
| | - Fermin Moreno
- Department of Neurology, Donostia University Hospital, San Sebastian, Gipuzkoa, Spain
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Raquel Sánchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire du CHU de Québec, Département des Sciences Neurologiques, Université Laval, Québec, Quebec City, Canada
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
| | - James B Rowe
- Cambridge University Centre for Frontotemporal Dementia, University of Cambridge, Cambridge, United Kingdom
| | | | - Simon Ducharme
- Montreal Neurological Institute and McGill University Health Centre, McGill University, Montreal, Québec, Canada
| | - Alex Gerhard
- Department of Nuclear Medicine and Geriatric Medicine, University Hospital Essen, Essen, Germany
- Divison of Neuroscience and Experimental Psychology, University of Manchester, Manchester, United Kingdom
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Neurodegenerative Diseases, (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology, (SyNergy), Munich, Germany
| | - Yolande Al Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Markus Otto
- Department of Neurology, Universität Ulm, Ulm, Germany
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | | | | | - Isabel Santana
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Daniela Galimberti
- Department of Neurological Sciences, Dino Ferrari Center, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Harro Seelaar
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jonathan D Rohrer
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - Paul F Worley
- Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
67
|
Chen SD, Li HQ, Cui M, Dong Q, Yu JT. Pluripotent stem cells for neurodegenerative disease modeling: an expert view on their value to drug discovery. Expert Opin Drug Discov 2020; 15:1081-1094. [PMID: 32425128 DOI: 10.1080/17460441.2020.1767579] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Neurodegenerative diseases have become a major global health concern, posing a huge disease burden on patients and their families. Although there has been rapid progress in the development of therapies, a lack of accurate disease models and efficient drug screening platforms have made achieving a breakthrough difficult. The technology of human-induced pluripotent stem cells (iPSCs) shows better recapitulation of disease pathophysiology and provides a more accessible supply of patient-specific samples compared to other modeling methods. It has been a powerful tool for mechanism exploration and drug development. AREAS COVERED This review describes the recent use of human iPSC-derived cells for modeling neurodegenerative disorders and discovering potential drugs. EXPERT OPINION Model systems based on iPSC-derived cells have created a paradigm shift in drug discovery. Accuracy, consistency, translatability, and cost-effectiveness are the four major focuses of academic and industrial communities to fulfill the potential of iPSC technology for their purposes. It is the art of balance between these four factors to generate efficacious outputs with maximum efficiency. Future studies should persist in refining this technology and promote its application in this field to benefit all the disease-affected population eventually.
Collapse
Affiliation(s)
- Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University , Shanghai, China
| | - Hong-Qi Li
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University , Shanghai, China
| | - Mei Cui
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University , Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University , Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University , Shanghai, China
| |
Collapse
|
68
|
Ehrenberg AJ, Khatun A, Coomans E, Betts MJ, Capraro F, Thijssen EH, Senkevich K, Bharucha T, Jafarpour M, Young PNE, Jagust W, Carter SF, Lashley T, Grinberg LT, Pereira JB, Mattsson-Carlgren N, Ashton NJ, Hanrieder J, Zetterberg H, Schöll M, Paterson RW. Relevance of biomarkers across different neurodegenerative diseases. ALZHEIMERS RESEARCH & THERAPY 2020; 12:56. [PMID: 32404143 PMCID: PMC7222479 DOI: 10.1186/s13195-020-00601-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/16/2020] [Indexed: 01/11/2023]
Abstract
Background The panel of fluid- and imaging-based biomarkers available for neurodegenerative disease research is growing and has the potential to close important gaps in research and the clinic. With this growth and increasing use, appropriate implementation and interpretation are paramount. Various biomarkers feature nuanced differences in strengths, limitations, and biases that must be considered when investigating disease etiology and clinical utility. For example, neuropathological investigations of Alzheimer’s disease pathogenesis can fall in disagreement with conclusions reached by biomarker-based investigations. Considering the varied strengths, limitations, and biases of different research methodologies and approaches may help harmonize disciplines within the neurodegenerative disease field. Purpose of review Along with separate review articles covering fluid and imaging biomarkers in this issue of Alzheimer’s Research and Therapy, we present the result of a discussion from the 2019 Biomarkers in Neurodegenerative Diseases course at the University College London. Here, we discuss themes of biomarker use in neurodegenerative disease research, commenting on appropriate use, interpretation, and considerations for implementation across different neurodegenerative diseases. We also draw attention to areas where biomarker use can be combined with other disciplines to understand issues of pathophysiology and etiology underlying dementia. Lastly, we highlight novel modalities that have been proposed in the landscape of neurodegenerative disease research and care.
Collapse
Affiliation(s)
- Alexander J Ehrenberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA. .,Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA. .,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA.
| | - Ayesha Khatun
- Dementia Research Centre, University College London Institute of Neurology, London, UK
| | - Emma Coomans
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, location VUmc, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Matthew J Betts
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Federica Capraro
- The Francis Crick Institute, London, UK.,Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, London, UK
| | - Elisabeth H Thijssen
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Department of Clinical Chemistry, Amsterdam UMC, Amsterdam, The Netherlands
| | - Konstantin Senkevich
- Petersburg Nuclear Physics Institute names by B.P. Konstantinov of National Research Center, Kurchatov Institute, St. Petersburg, Russia.,First Pavlov State Medical University of St. Petersburg, St. Petersburg, Russia
| | - Tehmina Bharucha
- Oxford Glycobiology Institute, Department of Biochemistry , University of Oxford, Oxford, UK
| | - Mehrsa Jafarpour
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK
| | - Peter N E Young
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Center for Molecular and Translational Medicine, Lund University, Lund, Sweden
| | - William Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA.,Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Stephen F Carter
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK.,Wolfson Molecular Imaging Centre, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Tammaryn Lashley
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK.,Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Lea T Grinberg
- Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,University of São Paulo Medical School, São Paulo, Brazil.,Global Brain Health Institute, San Francisco, CA, USA
| | - Joana B Pereira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Clinical Memory Research Unit, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Center for Molecular and Translational Medicine, Lund University, Lund, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Wallenberg Center for Molecular and Translational Medicine, Lund University, Lund, Sweden.,King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, UK.,NIHR Biomedical Research Centre for Mental Health & Biomedical Research Unit for Dementia at South London & Maudsley NHS Foundation, London, UK
| | - Jörg Hanrieder
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at University College London, London, UK
| | - Michael Schöll
- Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK.,Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Memory Research Unit, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Ross W Paterson
- Dementia Research Centre, University College London Institute of Neurology, London, UK
| |
Collapse
|
69
|
Cell-to-Cell Transmission of Tau and α-Synuclein. Trends Mol Med 2020; 26:936-952. [PMID: 32371172 DOI: 10.1016/j.molmed.2020.03.012] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022]
Abstract
The stereotypical spread of pathological protein inclusions and clinicopathological heterogeneity are well described in neurodegenerative diseases. Accumulating evidence suggests that the former can be attributed to consecutive cell-to-cell transmission of pathological proteins between anatomically connected brain regions, while the latter has been hypothesized to result from the spread of conformationally distinct pathological protein aggregates, or strains. These emerging concepts have dramatically changed our understanding of neurodegenerative diseases. In this review, we first summarize the background and recent findings underpinning these concepts with a focus on two major pathological proteins: tau and α-synuclein. We then discuss their clinical implications for tauopathies and synucleinopathies and propose a working hypothesis for future research.
Collapse
|
70
|
Ling H, Gelpi E, Davey K, Jaunmuktane Z, Mok KY, Jabbari E, Simone R, R'Bibo L, Brandner S, Ellis MJ, Attems J, Mann D, Halliday GM, Al-Sarraj S, Hedreen J, Ironside JW, Kovacs GG, Kovari E, Love S, Vonsattel JPG, Allinson KSJ, Hansen D, Bradshaw T, Setó-Salvia N, Wray S, de Silva R, Morris HR, Warner TT, Hardy J, Holton JL, Revesz T. Fulminant corticobasal degeneration: a distinct variant with predominant neuronal tau aggregates. Acta Neuropathol 2020; 139:717-734. [PMID: 31950334 PMCID: PMC7096362 DOI: 10.1007/s00401-019-02119-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/15/2019] [Accepted: 12/20/2019] [Indexed: 02/01/2023]
Abstract
Corticobasal degeneration typically progresses gradually over 5–7 years from onset till death. Fulminant corticobasal degeneration cases with a rapidly progressive course were rarely reported (RP-CBD). This study aimed to investigate their neuropathological characteristics. Of the 124 autopsy-confirmed corticobasal degeneration cases collected from 14 centres, we identified 6 RP-CBD cases (4.8%) who died of advanced disease within 3 years of onset. These RP-CBD cases had different clinical phenotypes including rapid global cognitive decline (N = 2), corticobasal syndrome (N = 2) and Richardson’s syndrome (N = 2). We also studied four corticobasal degeneration cases with an average disease duration of 3 years or less, who died of another unrelated illness (Intermediate-CBD). Finally, we selected 12 age-matched corticobasal degeneration cases out of a cohort of 110, who had a typical gradually progressive course and reached advanced clinical stage (End-stage-CBD). Quantitative analysis showed high overall tau burden (p = 0.2) and severe nigral cell loss (p = 0.47) in both the RP-CBD and End-stage-CBD groups consistent with advanced pathological changes, while the Intermediate-CBD group (mean disease duration = 3 years) had milder changes than End-stage-CBD (p < 0.05). These findings indicated that RP-CBD cases had already developed advanced pathological changes as those observed in End-stage-CBD cases (mean disease duration = 6.7 years), but within a significantly shorter duration (2.5 years; p < 0.001). Subgroup analysis was performed to investigate the cellular patterns of tau aggregates in the anterior frontal cortex and caudate by comparing neuronal-to-astrocytic plaque ratios between six RP-CBD cases, four Intermediate-CBD and 12 age-matched End-stage-CBD. Neuronal-to-astrocytic plaque ratios of Intermediate-CBD and End-stage-CBD, but not RP-CBD, positively correlated with disease duration in both the anterior frontal cortex and caudate (p = 0.02). In contrast to the predominance of astrocytic plaques we previously reported in preclinical asymptomatic corticobasal degeneration cases, neuronal tau aggregates predominated in RP-CBD exceeding those in Intermediate-CBD (anterior frontal cortex: p < 0.001, caudate: p = 0.001) and End-stage-CBD (anterior frontal cortex: p = 0.03, caudate: p = 0.01) as demonstrated by its higher neuronal-to-astrocytic plaque ratios in both anterior frontal cortex and caudate. We did not identify any difference in age at onset, any pathogenic tau mutation or concomitant pathologies that could have contributed to the rapid progression of these RP-CBD cases. Mild TDP-43 pathology was observed in three RP-CBD cases. All RP-CBD cases were men. The MAPT H2 haplotype, known to be protective, was identified in one RP-CBD case (17%) and 8 of the matched End-stage-CBD cases (67%). We conclude that RP-CBD is a distinct aggressive variant of corticobasal degeneration with characteristic neuropathological substrates resulting in a fulminant disease process as evident both clinically and pathologically. Biological factors such as genetic modifiers likely play a pivotal role in the RP-CBD variant and should be the subject of future research.
Collapse
Affiliation(s)
- Helen Ling
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK.
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Ellen Gelpi
- Neurological Tissue Bank of the Biobanc-Hospital Clinic-IDIBAPS, Barcelona, Spain
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Karen Davey
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Zane Jaunmuktane
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London Hospital Trust, Queen Square, London, UK
| | - Kin Y Mok
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Division of Life Science, Institute for Advanced Study, Hong Kong University of Science and Technology, Hong Kong Special Administrative Region, Hong Kong, China
| | - Edwin Jabbari
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Roberto Simone
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Lea R'Bibo
- UK Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sebastian Brandner
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London Hospital Trust, Queen Square, London, UK
| | - Matthew J Ellis
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Johannes Attems
- Newcastle Brain Tissue Resource, Institute of Neuroscience, Newcastle University, Newcastle, UK
| | - David Mann
- Manchester Brain Bank, University of Manchester, Manchester, UK
| | - Glenda M Halliday
- Sydney Brain Bank, Neuroscience Research Australia (NeuRA), Sydney, Australia
- Brain and Mind Centre and Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - S Al-Sarraj
- The London Neurodegeneration Brain Bank, The Institute of Psychiatry Psychology and Neurosciences (IOPPN), Kings College London, London, UK
| | - J Hedreen
- The Harvard Brain Tissue Resource Centre, McLean Hospital, Belmont, USA
| | - James W Ironside
- National CJD Research and Surveillance Unit, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Gabor G Kovacs
- University of Toronto, University Health Network, and Tanz Centre for Research in Neurodegenerative Disease, Toronto, Canada
| | - E Kovari
- Department of Psychiatry, HUG Belle-Idée, University of Geneva School of Medicine, Geneva, Switzerland
| | - S Love
- South West Dementia Brain Bank, University of Bristol, Bristol, UK
| | - Jean Paul G Vonsattel
- Taub Institute for Research on AD and the Aging Brain, Columbia University Medical Center, New York, USA
| | | | - Daniela Hansen
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Teisha Bradshaw
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Núria Setó-Salvia
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Selina Wray
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Rohan de Silva
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Huw R Morris
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Thomas T Warner
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - John Hardy
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Janice L Holton
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Tamas Revesz
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, University College London, 1 Wakefield Street, London, WC1N 1PJ, UK.
- Reta Lila Weston Institute for Neurological Studies, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
71
|
Genetic architecture of neurodegenerative dementias. Neuropharmacology 2020; 168:108014. [PMID: 32097768 DOI: 10.1016/j.neuropharm.2020.108014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/03/2020] [Accepted: 02/14/2020] [Indexed: 12/14/2022]
Abstract
Molecular genetics has been an invaluable tool to help understand the molecular basis of neurodegenerative dementias. In this review, we provide an overview of the genetic architecture underlying some of the most prevalent causes of dementia, including Alzheimer's dementia, frontotemporal lobar degeneration, Lewy body dementia, and prion diseases. We also discuss the complexity of the human genome and how the novel technologies have revolutionized and accelerated the way we screen the variety of our DNA. Finally, we also provide some examples about how this genetic knowledge is being transferred into the clinic through personalized medicine. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
|
72
|
Convery RS, Neason MR, Cash DM, Cardoso MJ, Modat M, Ourselin S, Warren JD, Rohrer JD, Bocchetta M. Basal forebrain atrophy in frontotemporal dementia. NEUROIMAGE-CLINICAL 2020; 26:102210. [PMID: 32143137 PMCID: PMC7058403 DOI: 10.1016/j.nicl.2020.102210] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/22/2020] [Accepted: 02/12/2020] [Indexed: 12/16/2022]
Abstract
Background The basal forebrain is a subcortical structure that plays an important role in learning, attention, and memory. Despite the known subcortical involvement in frontotemporal dementia (FTD), there is little research into the role of the basal forebrain in this disease. We aimed to investigate differences in basal forebrain volumes between clinical, genetic, and pathological diagnoses of FTD. Methods 356 patients with FTD were recruited from the UCL Dementia Research Centre and matched on age and gender with 83 cognitively normal controls. All subjects had a T1-weighted MR scan suitable for analysis. Basal forebrain volumes were calculated using the Geodesic Information Flow (GIF) parcellation method and were compared between clinical (148 bvFTD, 82 svPPA, 103 nfvPPA, 14 PPA–NOS, 9 FTD–MND), genetic (24 MAPT, 15 GRN, 26 C9orf72) and pathological groups (28 tau, 3 FUS, 35 TDP-43) and controls. A subanalysis was also performed comparing pathological subgroups of tau (11 Pick's disease, 6 FTDP-17, 7 CBD, 4 PSP) and TDP-43 (12 type A, 2 type B, 21 type C). Results All clinical subtypes of FTD showed significantly smaller volumes than controls (p ≤ 0.010, ANCOVA), with svPPA (10% volumetric difference) and bvFTD (9%) displaying the smallest volumes. Reduced basal forebrain volumes were also seen in MAPT mutations (18%, p < 0.0005) and in individuals with pathologically confirmed FTDP-17 (17%), Pick's disease (12%), and TDP-43 type C (8%) (p < 0.001). Conclusion Involvement of the basal forebrain is a common feature in FTD, although the extent of volume reduction differs between clinical, genetic, and pathological diagnoses. Tauopathies, particularly those with MAPT mutations, had the smallest volumes. However, atrophy was also seen in those with TDP-43 type C pathology (most of whom have svPPA clinically). This suggests that the basal forebrain is vulnerable to multiple types of FTD-associated protein inclusions.
Collapse
Affiliation(s)
- Rhian S Convery
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Mollie R Neason
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom; Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - M Jorge Cardoso
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Marc Modat
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Sebastien Ourselin
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Jason D Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| | - Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
73
|
Poos JM, Jiskoot LC, Leijdesdorff SMJ, Seelaar H, Panman JL, van der Ende EL, Mol MO, Meeter LHH, Pijnenburg YAL, Donker Kaat L, de Jong FJ, van Swieten JC, Papma JM, van den Berg E. Cognitive profiles discriminate between genetic variants of behavioral frontotemporal dementia. J Neurol 2020; 267:1603-1612. [PMID: 32052166 PMCID: PMC7293665 DOI: 10.1007/s00415-020-09738-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 12/12/2022]
Abstract
Introduction Trials to test disease-modifying treatments for frontotemporal dementia are eagerly awaited and sensitive instruments to assess potential treatment effects are increasingly urgent, yet lacking thus far. We aimed to identify gene-specific instruments assessing clinical onset and disease progression by comparing cognitive functioning between bvFTD patients across genetic mutations. Methods We examined differences in 7 cognitive domains between bvFTD patients with GRN (n = 20), MAPT (n = 29) or C9orf72 (n = 31) mutations, and non-carriers (n = 24), and described longitudinal (M = 22.6 months, SD = 16.6) data in a subsample (n = 27). Results Patients showed overall cognitive impairment, except memory recall, working memory and visuoconstruction. GRN patients performed lower on executive function (mean difference − 2.1; 95%CI − 4.1 to − 0.5) compared to MAPT and lower on attention compared to MAPT (mean difference − 2.5; 95%CI − 4.7 to − 0.3) and C9orf72 (mean difference − 2.4; 95%CI − 4.5 to − 0.3). Only MAPT patients were impaired on delayed recall (mean difference − 1.4; 95%CI − 2.1 to − 0.7). GRN patients declined rapidly on attention and memory, MAPT declined in confrontation naming, whereas C9orf72 patients were globally impaired but remained relatively stable over time on all cognitive domains. Discussion This study shows gene-specific cognitive profiles in bvFTD, which underlines the value of neuropsychological tests as outcome measures in upcoming trials for genetic bvFTD. Electronic supplementary material The online version of this article (10.1007/s00415-020-09738-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J M Poos
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands. .,Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.
| | - L C Jiskoot
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands.,Dementia Research Center, University College London, London, UK
| | - S M J Leijdesdorff
- Department of Psychiatry and Psychology, Maastricht University, Maastricht, The Netherlands
| | - H Seelaar
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| | - J L Panman
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands.,Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - E L van der Ende
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| | - M O Mol
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| | - L H H Meeter
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| | - Y A L Pijnenburg
- Department of Neurology, Alzheimer Center, Location VU University Medical CenterAmsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - L Donker Kaat
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - F J de Jong
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| | - J C van Swieten
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| | - J M Papma
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| | - E van den Berg
- Department of Neurology, Alzheimer Center, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3000 CA, Rotterdam, The Netherlands
| |
Collapse
|
74
|
Khatun A, Paterson RW, Schöll M. University College London/University of Gothenburg PhD course "Biomarkers in neurodegenerative diseases" 2019-course organisation. ALZHEIMERS RESEARCH & THERAPY 2020; 12:18. [PMID: 32019594 PMCID: PMC7001332 DOI: 10.1186/s13195-020-0583-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/13/2020] [Indexed: 11/10/2022]
Abstract
Biomarkers are increasingly employed for effective research into neurodegenerative diseases. They have become essential for reaching an accurate clinical diagnosis, monitoring disease, and refining entry criteria for participation in clinical treatment trials, and will be key in measuring target engagement and treatment outcome in disease-modifying therapies. Emerging techniques and research combining different biomarker modalities continue to strengthen our understanding of the underlying pathology and the sequence of pathogenic events. Given recent advances, we are now at a pivotal stage in biomarker research. PhD students working in the field of neurodegenerative disease require a working knowledge of a range of biomarkers available and their limitations, to correctly interpret scientific literature and to design and conduct successful research studies themselves. Here, we outline the University College London/University of Gothenburg "Biomarkers in neurodegenerative diseases course", the first initiative of its kind aimed to bring together both experts and PhD students from all areas within the field of neurodegeneration, to provide comprehensive knowledge of biomarker research for the next generation of scientists.
Collapse
Affiliation(s)
- Ayesha Khatun
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Ross W Paterson
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK
| | - Michael Schöll
- Dementia Research Centre, Queen Square Institute of Neurology, University College London, London, UK. .,Wallenberg Centre for Molecular and Translational Medicine and the Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden. .,Department of Clinical Physiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
75
|
Takeda T, Kitagawa K, Arai K. Phenotypic variability and its pathological basis in amyotrophic lateral sclerosis. Neuropathology 2019; 40:40-56. [PMID: 31802540 DOI: 10.1111/neup.12606] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 12/19/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by its inherent clinicopathological variability. The concurrence of upper and lower motor neuron signs is a common feature in the majority of patients with ALS. However, some patients manifest an atypical clinical course, with only upper or lower motor neuron signs, or various extra-motor symptoms including cognitive dysfunction, parkinsonism, autonomic dysfunction, or ophthalmoparesis. This variability indicates different manifestations of ALS and is reflected by ALS pathology spreading into the central nervous system. The presence of cytoplasmic inclusions positive for transactivation response DNA-binding protein 43 kDa (TDP-43) is a key feature in ALS. Loss of TDP-43 from the nucleus and its subsequent aggregation in the cytoplasm may occur in susceptible regions and may be associated with neuronal loss. However, in some regions, there is no apparent neuronal loss while TDP-43 accumulation is evident; in contrast, in other regions, neuronal loss is apparent without any evidence of TDP-43 accumulation. Therefore, in addition to TDP-43 dysfunction, underlying region-specific cellular vulnerability may exist in the upper and lower motor neurons and frontotemporal system in patients with ALS. The microscopic discrepancy and selective vulnerability may be linked to the macroscopic propensities of the sites of onset, and may also determine the direction and rate of progression of the lesions. Thus, there may be multicentric sites of onset, region-oriented disease development, and different speeds of disease progression across patients with ALS. ALS lesions occur in motor-related areas but may spread to neighboring areas. However, since lesions may spread in a discontinuous manner, and the dynamics of disease propagation have not been able to be identified, it remains controversial whether the stepwise appearance of TDP-43-positive inclusions is based on direct cell-to-cell protein propagation. Further understanding of the phenotypic variability of ALS and its pathological basis may serve as a guide for investigating the underlying pathogenesis of ALS.
Collapse
Affiliation(s)
- Takahiro Takeda
- Department of Neurology, National Hospital Organization Chibahigashi National Hospital, Chiba, Japan.,Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kimihito Arai
- Department of Neurology, National Hospital Organization Chibahigashi National Hospital, Chiba, Japan
| |
Collapse
|
76
|
van der Ende EL, Meeter LH, Poos JM, Panman JL, Jiskoot LC, Dopper EGP, Papma JM, de Jong FJ, Verberk IMW, Teunissen C, Rizopoulos D, Heller C, Convery RS, Moore KM, Bocchetta M, Neason M, Cash DM, Borroni B, Galimberti D, Sanchez-Valle R, Laforce R, Moreno F, Synofzik M, Graff C, Masellis M, Carmela Tartaglia M, Rowe JB, Vandenberghe R, Finger E, Tagliavini F, de Mendonça A, Santana I, Butler C, Ducharme S, Gerhard A, Danek A, Levin J, Otto M, Frisoni GB, Cappa S, Pijnenburg YAL, Rohrer JD, van Swieten JC, Warren JD, Fox NC, Woollacott IO, Shafei R, Greaves C, Guerreiro R, Bras J, Thomas DL, Nicholas J, Mead S, van Minkelen R, Barandiaran M, Indakoetxea B, Gabilondo A, Tainta M, de Arriba M, Gorostidi A, Zulaica M, Villanua J, Diaz Z, Borrego-Ecija S, Olives J, Lladó A, Balasa M, Antonell A, Bargallo N, Premi E, Cosseddu M, Gazzina S, Padovani A, Gasparotti R, Archetti S, Black S, Mitchell S, Rogaeva E, Freedman M, Keren R, Tang-Wai D, Öijerstedt L, Andersson C, Jelic V, Thonberg H, Arighi A, Fenoglio C, Scarpini E, Fumagalli G, Cope T, Timberlake C, Rittman T, Shoesmith C, Bartha R, Rademakers R, Wilke C, Karnath HO, Bender B, Bruffaerts R, Vandamme P, et alvan der Ende EL, Meeter LH, Poos JM, Panman JL, Jiskoot LC, Dopper EGP, Papma JM, de Jong FJ, Verberk IMW, Teunissen C, Rizopoulos D, Heller C, Convery RS, Moore KM, Bocchetta M, Neason M, Cash DM, Borroni B, Galimberti D, Sanchez-Valle R, Laforce R, Moreno F, Synofzik M, Graff C, Masellis M, Carmela Tartaglia M, Rowe JB, Vandenberghe R, Finger E, Tagliavini F, de Mendonça A, Santana I, Butler C, Ducharme S, Gerhard A, Danek A, Levin J, Otto M, Frisoni GB, Cappa S, Pijnenburg YAL, Rohrer JD, van Swieten JC, Warren JD, Fox NC, Woollacott IO, Shafei R, Greaves C, Guerreiro R, Bras J, Thomas DL, Nicholas J, Mead S, van Minkelen R, Barandiaran M, Indakoetxea B, Gabilondo A, Tainta M, de Arriba M, Gorostidi A, Zulaica M, Villanua J, Diaz Z, Borrego-Ecija S, Olives J, Lladó A, Balasa M, Antonell A, Bargallo N, Premi E, Cosseddu M, Gazzina S, Padovani A, Gasparotti R, Archetti S, Black S, Mitchell S, Rogaeva E, Freedman M, Keren R, Tang-Wai D, Öijerstedt L, Andersson C, Jelic V, Thonberg H, Arighi A, Fenoglio C, Scarpini E, Fumagalli G, Cope T, Timberlake C, Rittman T, Shoesmith C, Bartha R, Rademakers R, Wilke C, Karnath HO, Bender B, Bruffaerts R, Vandamme P, Vandenbulcke M, Ferreira CB, Miltenberger G, Maruta C, Verdelho A, Afonso S, Taipa R, Caroppo P, Di Fede G, Giaccone G, Prioni S, Redaelli V, Rossi G, Tiraboschi P, Duro D, Rosario Almeida M, Castelo-Branco M, João Leitão M, Tabuas-Pereira M, Santiago B, Gauthier S, Schonecker S, Semler E, Anderl-Straub S, Benussi L, Binetti G, Ghidoni R, Pievani M, Lombardi G, Nacmias B, Ferrari C, Bessi V. Serum neurofilament light chain in genetic frontotemporal dementia: a longitudinal, multicentre cohort study. Lancet Neurol 2019; 18:1103-1111. [PMID: 31701893 DOI: 10.1016/s1474-4422(19)30354-0] [Show More Authors] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 07/26/2019] [Accepted: 08/13/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neurofilament light chain (NfL) is a promising blood biomarker in genetic frontotemporal dementia, with elevated concentrations in symptomatic carriers of mutations in GRN, C9orf72, and MAPT. A better understanding of NfL dynamics is essential for upcoming therapeutic trials. We aimed to study longitudinal NfL trajectories in people with presymptomatic and symptomatic genetic frontotemporal dementia. METHODS We recruited participants from 14 centres collaborating in the Genetic Frontotemporal Dementia Initiative (GENFI), which is a multicentre cohort study of families with genetic frontotemporal dementia done across Europe and Canada. Eligible participants (aged ≥18 years) either had frontotemporal dementia due to a pathogenic mutation in GRN, C9orf72, or MAPT (symptomatic mutation carriers) or were healthy at-risk first-degree relatives (either presymptomatic mutation carriers or non-carriers), and had at least two serum samples with a time interval of 6 months or more. Participants were excluded if they had neurological comorbidities that were likely to affect NfL, including cerebrovascular events. We measured NfL longitudinally in serum samples collected between June 8, 2012, and Dec 8, 2017, through follow-up visits annually or every 2 years, which also included MRI and neuropsychological assessments. Using mixed-effects models, we analysed NfL changes over time and correlated them with longitudinal imaging and clinical parameters, controlling for age, sex, and study site. The primary outcome was the course of NfL over time in the various stages of genetic frontotemporal dementia. FINDINGS We included 59 symptomatic carriers and 149 presymptomatic carriers of a mutation in GRN, C9orf72, or MAPT, and 127 non-carriers. Nine presymptomatic carriers became symptomatic during follow-up (so-called converters). Baseline NfL was elevated in symptomatic carriers (median 52 pg/mL [IQR 24-69]) compared with presymptomatic carriers (9 pg/mL [6-13]; p<0·0001) and non-carriers (8 pg/mL [6-11]; p<0·0001), and was higher in converters than in non-converting carriers (19 pg/mL [17-28] vs 8 pg/mL [6-11]; p=0·0007; adjusted for age). During follow-up, NfL increased in converters (b=0·097 [SE 0·018]; p<0·0001). In symptomatic mutation carriers overall, NfL did not change during follow-up (b=0·017 [SE 0·010]; p=0·101) and remained elevated. Rates of NfL change over time were associated with rate of decline in Mini Mental State Examination (b=-94·7 [SE 33·9]; p=0·003) and atrophy rate in several grey matter regions, but not with change in Frontotemporal Lobar Degeneration-Clinical Dementia Rating scale score (b=-3·46 [SE 46·3]; p=0·941). INTERPRETATION Our findings show the value of blood NfL as a disease progression biomarker in genetic frontotemporal dementia and suggest that longitudinal NfL measurements could identify mutation carriers approaching symptom onset and capture rates of brain atrophy. The characterisation of NfL over the course of disease provides valuable information for its use as a treatment effect marker. FUNDING ZonMw and the Bluefield project.
Collapse
Affiliation(s)
- Emma L van der Ende
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands
| | - Lieke H Meeter
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jackie M Poos
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jessica L Panman
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Lize C Jiskoot
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands; Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Elise G P Dopper
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands
| | - Janne M Papma
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands
| | - Frank Jan de Jong
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands
| | - Inge M W Verberk
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Charlotte Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, Netherlands
| | - Dimitris Rizopoulos
- Department of Biostatistics, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands
| | - Carolin Heller
- Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Rhian S Convery
- Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Katrina M Moore
- Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Martina Bocchetta
- Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Mollie Neason
- Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - David M Cash
- Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Barbara Borroni
- Centre for Neurodegenerative Disorders, Neurology unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurodegenerative Diseases Unit, Milan, Italy; University of Milan, Centro Dino Ferrari, Milan, Italy
| | - Raquel Sanchez-Valle
- Hospital Clinic de Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - Fermin Moreno
- Department of Neurology, Hospital Universitario Donostia, Gipuzkoa, Spain
| | - Matthis Synofzik
- Hertie-Institute for Clinical Brain Research Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE) Tübingen, Tübingen, Germany
| | - Caroline Graff
- Karolinska Institutet, Dept NVS, Division of Neurogeriatrics, Stockholm, Sweden; Unit of Hereditary Dementia, Theme Aging, Karolinska University Hospital-Solna, Stockholm, Sweden
| | | | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, ON, Canada
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, ON, Canada
| | | | | | - Isabel Santana
- Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Chris Butler
- Department of Clinical Neurology, University of Oxford, Oxford, UK
| | - Simon Ducharme
- Montreal Neurological Institute and McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Alex Gerhard
- Faculty of Medical and Human Sciences, Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Markus Otto
- Department of Neurology, Universität Ulm, Ulm, Germany
| | - Giovanni B Frisoni
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Stefano Cappa
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam and Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center, location VU University Medical Center, Amsterdam, Netherlands
| | - Jonathan D Rohrer
- Dementia Research Institute, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - John C van Swieten
- Department of Neurology and Alzheimer Center, Erasmus Medical Center Rotterdam, Rotterdam, Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Fernandopulle M, Wang G, Nixon-Abell J, Qamar S, Balaji V, Morihara R, St George-Hyslop PH. Inherited and Sporadic Amyotrophic Lateral Sclerosis and Fronto-Temporal Lobar Degenerations arising from Pathological Condensates of Phase Separating Proteins. Hum Mol Genet 2019; 28:R187-R196. [PMID: 31595953 PMCID: PMC6872449 DOI: 10.1093/hmg/ddz162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022] Open
Abstract
Recent work on the biophysics of proteins with low complexity, intrinsically disordered domains that have the capacity to form biological condensates has profoundly altered the concepts about the pathogenesis of inherited and sporadic neurodegenerative disorders associated with pathological accumulation of these proteins. In the present review, we use the FUS, TDP-43 and A11 proteins as examples to illustrate how missense mutations and aberrant post-translational modifications of these proteins cause amyotrophic lateral sclerosis (ALS) and fronto-temporal lobar degeneration (FTLD).
Collapse
Affiliation(s)
- Michael Fernandopulle
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - GuoZhen Wang
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Jonathon Nixon-Abell
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Seema Qamar
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Varun Balaji
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| | - Ryuta Morihara
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| | - Peter H St George-Hyslop
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| |
Collapse
|
78
|
Bocchetta M, Iglesias JE, Neason M, Cash DM, Warren JD, Rohrer JD. Thalamic nuclei in frontotemporal dementia: Mediodorsal nucleus involvement is universal but pulvinar atrophy is unique to C9orf72. Hum Brain Mapp 2019; 41:1006-1016. [PMID: 31696638 PMCID: PMC7267940 DOI: 10.1002/hbm.24856] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 10/09/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022] Open
Abstract
Thalamic atrophy is a common feature across all forms of FTD but little is known about specific nuclei involvement. We aimed to investigate in vivo atrophy of the thalamic nuclei across the FTD spectrum. A cohort of 402 FTD patients (age: mean(SD) 64.3(8.2) years; disease duration: 4.8(2.8) years) was compared with 104 age-matched controls (age: 62.5(10.4) years), using an automated segmentation of T1-weighted MRIs to extract volumes of 14 thalamic nuclei. Stratification was performed by clinical diagnosis (180 behavioural variant FTD (bvFTD), 85 semantic variant primary progressive aphasia (svPPA), 114 nonfluent variant PPA (nfvPPA), 15 PPA not otherwise specified (PPA-NOS), and 8 with associated motor neurone disease (FTD-MND), genetic diagnosis (27 MAPT, 28 C9orf72, 18 GRN), and pathological confirmation (37 tauopathy, 38 TDP-43opathy, 4 FUSopathy). The mediodorsal nucleus (MD) was the only nucleus affected in all FTD subgroups (16-33% smaller than controls). The laterodorsal nucleus was also particularly affected in genetic cases (28-38%), TDP-43 type A (47%), tau-CBD (44%), and FTD-MND (53%). The pulvinar was affected only in the C9orf72 group (16%). Both the lateral and medial geniculate nuclei were also affected in the genetic cases (10-20%), particularly the LGN in C9orf72 expansion carriers. Use of individual thalamic nuclei volumes provided higher accuracy in discriminating between FTD groups than the whole thalamic volume. The MD is the only structure affected across all FTD groups. Differential involvement of the thalamic nuclei among FTD forms is seen, with a unique pattern of atrophy in the pulvinar in C9orf72 expansion carriers.
Collapse
Affiliation(s)
- Martina Bocchetta
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Juan E Iglesias
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK.,Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, Boston, Massachusetts, USA
| | - Mollie Neason
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - David M Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK.,Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Jason D Warren
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
79
|
Wallings RL, Humble SW, Ward ME, Wade-Martins R. Lysosomal Dysfunction at the Centre of Parkinson's Disease and Frontotemporal Dementia/Amyotrophic Lateral Sclerosis. Trends Neurosci 2019; 42:899-912. [PMID: 31704179 DOI: 10.1016/j.tins.2019.10.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/25/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) and frontotemporal dementia/amyotrophic lateral sclerosis (FTD/ALS) are insidious and incurable neurodegenerative diseases that represent a significant burden to affected individuals, caregivers, and an ageing population. Both PD and FTD/ALS are defined at post mortem by the presence of protein aggregates and the loss of specific subsets of neurons. We examine here the crucial role of lysosome dysfunction in these diseases and discuss recent evidence for converging mechanisms. This review draws upon multiple lines of evidence from genetic studies, human tissue, induced pluripotent stem cells (iPSCs), and animal models to argue that lysosomal failure is a primary mechanism of disease, rather than merely reflecting association with protein aggregate end-points. This review provides compelling rationale for targeting lysosomes in future therapeutics for both PD and FTD/ALS.
Collapse
Affiliation(s)
- Rebecca L Wallings
- Department of Physiology, Emory University, Decatur, GA, USA; Current address: Department of Neuroscience, Center for Translational Research and Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Stewart W Humble
- Oxford Parkinson's Disease Centre, Department of Physiology Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK; National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3QX, UK.
| |
Collapse
|
80
|
Forrest SL, Kril JJ, Halliday GM. Cellular and regional vulnerability in frontotemporal tauopathies. Acta Neuropathol 2019; 138:705-727. [PMID: 31203391 DOI: 10.1007/s00401-019-02035-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/04/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022]
Abstract
The frontotemporal tauopathies all deposit abnormal tau protein aggregates, but often of only certain isoforms and in distinguishing pathologies of five main types (neuronal Pick bodies, neurofibrillary tangles, astrocytic plaques, tufted astrocytes, globular glial inclusions and argyrophilic grains). In those with isoform specific tau aggregates glial pathologies are substantial, even though there is limited evidence that these cells normally produce tau protein. This review will assess the differentiating features and clinicopathological correlations of the frontotemporal tauopathies, the genetic predisposition for these different pathologies, their neuroanatomical selectivity, current observations on how they spread through the brain, and any potential contributing cellular and molecular changes. The findings show that diverse clinical phenotypes relate most to the brain region degenerating rather than the type of pathology involved, that different regions on the MAPT gene and novel risk genes are associated with specific tau pathologies, that the 4-repeat glial tauopathies do not follow individual patterns of spreading as identified for neuronal pathologies, and that genetic and pathological data indicate that neuroinflammatory mechanisms are involved. Each pathological frontotemporal tauopathy subtype with their distinct pathological features differ substantially in the cell type affected, morphology, biochemical and anatomical distribution of inclusions, a fundamental concept central to future success in understanding the disease mechanisms required for developing therapeutic interventions. Tau directed therapies targeting genetic mechanisms, tau aggregation and pathological spread are being trialled, although biomarkers that differentiate these diseases are required. Suggested areas of future research to address the regional and cellular vulnerabilities in frontotemporal tauopathies are discussed.
Collapse
|
81
|
Miki Y, Ling H, Crampsie S, Mummery CJ, Rohrer JD, Jaunmuktane Z, Lashley T, Holton JL. Corticospinal tract degeneration and temporal lobe atrophy in frontotemporal lobar degeneration TDP-43 type C pathology. Neuropathol Appl Neurobiol 2019; 46:296-299. [PMID: 31602701 DOI: 10.1111/nan.12582] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/02/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Y Miki
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.,Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - H Ling
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.,Reta Lila Weston Institute of Neurological Studies, UCL Queen Square Institute of Neurology, London, UK
| | - S Crampsie
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - C J Mummery
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Queen Square Institute of Neurology, Queen Square, London, UK
| | - J D Rohrer
- Department of Neurodegenerative Disease, Dementia Research Centre, UCL Queen Square Institute of Neurology, Queen Square, London, UK
| | - Z Jaunmuktane
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.,Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London, UK
| | - T Lashley
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - J L Holton
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.,Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
82
|
Clark CN, Quaegebeur A, Nirmalananthan N, MacKinnon AD, Revesz T, Holton JL, Bridges LR, Pereira AC. Foix-Chavany-Marie syndrome due to type E TDP43 pathology. Neuropathol Appl Neurobiol 2019; 46:292-295. [PMID: 31520433 DOI: 10.1111/nan.12579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Affiliation(s)
- C N Clark
- Neurology Department, Atkinson Morley Regional Neuroscience Centre, St George's University Hospitals NHS Foundation Trust, London, UK
| | - A Quaegebeur
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - N Nirmalananthan
- Neurology Department, Atkinson Morley Regional Neuroscience Centre, St George's University Hospitals NHS Foundation Trust, London, UK
| | - A D MacKinnon
- Department of Neuroradiology, Atkinson Morley Regional Neuroscience Centre, St George's University Hospitals NHS Foundation Trust, London, UK
| | - T Revesz
- Queen Square Brain Bank for Neurological Disorders and Department of Neurodegenerative Disease, UCL, Queen Square Institute of Neurology, London, UK
| | - J L Holton
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - L R Bridges
- Department of Cellular Pathology, St George's Hospital, St George's University Hospitals NHS Foundation Trust, London, UK
| | - A C Pereira
- Neurology Department, Atkinson Morley Regional Neuroscience Centre, St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
83
|
Ishii K. Diagnostic imaging of dementia with Lewy bodies, frontotemporal lobar degeneration, and normal pressure hydrocephalus. Jpn J Radiol 2019; 38:64-76. [DOI: 10.1007/s11604-019-00881-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 09/10/2019] [Indexed: 10/25/2022]
|
84
|
Kovacs GG. Molecular pathology of neurodegenerative diseases: principles and practice. J Clin Pathol 2019; 72:725-735. [PMID: 31395625 DOI: 10.1136/jclinpath-2019-205952] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are characterised by selective dysfunction and progressive loss of synapses and neurons associated with pathologically altered proteins that deposit primarily in the human brain and spinal cord. Recent discoveries have identified a spectrum of distinct immunohistochemically and biochemically detectable proteins, which serve as a basis for protein-based disease classification. Diagnostic criteria have been updated and disease staging procedures have been proposed. These are based on novel concepts which recognise that (1) most of these proteins follow a sequential distribution pattern in the brain suggesting a seeding mechanism and cell-to-cell propagation; (2) some of the neurodegeneration-associated proteins can be detected in peripheral organs; and (3) concomitant presence of neurodegeneration-associated proteins is more the rule than the exception. These concepts, together with the fact that the clinical symptoms do not unequivocally reflect the molecular pathological background, place the neuropathological examination at the centre of requirements for an accurate diagnosis. The need for quality control in biomarker development, clinical and neuroimaging studies, and evaluation of therapy trials, as well as an increasing demand for the general public to better understand human brain disorders, underlines the importance for a renaissance of postmortem neuropathological studies at this time. This review summarises recent advances in neuropathological diagnosis and reports novel aspects of relevance for general pathological practice.
Collapse
Affiliation(s)
- Gabor G Kovacs
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
85
|
Andrés-Benito P, Gelpi E, Povedano M, Santpere G, Ferrer I. Gene Expression Profile in Frontal Cortex in Sporadic Frontotemporal Lobar Degeneration-TDP. J Neuropathol Exp Neurol 2019; 77:608-627. [PMID: 29788265 DOI: 10.1093/jnen/nly037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Molecular alterations compromising key metabolic pathways are poorly understood in sporadic frontotemporal lobar degeneration with TDP-43 pathology (sFTLD-TDP). Whole-transcriptome array, RT-qPCR validation, gel electrophoresis, and Western blotting, and mitochondrial electron transport chain (ETC) activity were comparatively examined in frontal cortex (area 8) of 16 sFTLD-TDP cases and 14 controls. Assessment of 111 genes by RT-qPCR showed deregulation of 81 genes linked to neurotransmission and synapses, neuronal architecture, cytoskeleton of axons and dendrites, vesicle trafficking, purines, mitochondria, and energy metabolism in sFTLD-TDP. Western blotting studies disclosed downregulation of several mitochondrial subunits encoded by genomic DNA and MT-CO1 encoded by the mitochondrial DNA. Mitochondrial ETC activity of complexes I, IV, and V was decreased in sFTLD-TDP. These findings provide robust information about downregulation of genes involved in vital biochemical pathways and in synaptic neurotransmission which may help to increase understanding about the biochemical substrates of clinical manifestations in sFTLD-TDP.
Collapse
Affiliation(s)
- Pol Andrés-Benito
- Neuropathology, Pathologic Anatomy Service, Bellvitge University Hospital, IDIBELL, Hospitalet de Llobregat, Spain.,Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Hospitalet de Llobregat, Spain
| | - Ellen Gelpi
- Neurological Tissue Bank of the Biobanc-Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Mónica Povedano
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Service of Neurology, Bellvitge University Hospital, Hospitalet de Llobregat, Spain
| | - Gabriel Santpere
- Department of Neurobiology, Yale School of Medicine, New Haven, Connecticut.,Department of Experimental and Health Sciences, IBE, Institute of Evolutionary Biology, Universitat Pompeu Fabra-CSIC, Barcelona, Spain
| | - Isidro Ferrer
- Neuropathology, Pathologic Anatomy Service, Bellvitge University Hospital, IDIBELL, Hospitalet de Llobregat, Spain.,Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute Carlos III, Hospitalet de Llobregat, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain.,Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
86
|
Birsa N, Bentham MP, Fratta P. Cytoplasmic functions of TDP-43 and FUS and their role in ALS. Semin Cell Dev Biol 2019; 99:193-201. [PMID: 31132467 DOI: 10.1016/j.semcdb.2019.05.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/06/2019] [Accepted: 05/23/2019] [Indexed: 02/08/2023]
Abstract
TAR DNA-binding protein of 43 kDa (TDP-43) and fused in sarcoma (FUS) are RNA binding proteins (RBPs) primarily located in the nucleus, and involved in numerous aspects of RNA metabolism. Both proteins can be found to be depleted from the nucleus and accumulated in cytoplasmic inclusions in two major neurodegenerative conditions, amyotrophic lateral sclerosis and frontotemporal dementia. Recent evidences suggest that, in addition to their nuclear functions, both TDP-43 and FUS are involved in multiple processes in the cytoplasm, including mRNA stability and transport, translation, the stress response, mitochondrial function and autophagy regulation. Here, we review the most recent advances in understanding their functions in the cytoplasm and how these are affected in disease.
Collapse
Affiliation(s)
- Nicol Birsa
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK.
| | - Matthew Peter Bentham
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Pietro Fratta
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, Queen Square, London, WC1N 3BG, UK; MRC Centre for Neuromuscular Disease, Queen Square, London, WC1N 3BG, UK.
| |
Collapse
|
87
|
Logroscino G, Piccininni M, Binetti G, Zecca C, Turrone R, Capozzo R, Tortelli R, Battista P, Bagoj E, Barone R, Fostinelli S, Benussi L, Ghidoni R, Padovani A, Cappa SF, Alberici A, Borroni B. Incidence of frontotemporal lobar degeneration in Italy: The Salento-Brescia Registry study. Neurology 2019; 92:e2355-e2363. [PMID: 30979859 DOI: 10.1212/wnl.0000000000007498] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 01/19/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE The goal of the present work, based on a collaborative research registry in Italy (the Salento-Brescia Registry), was to assess the incidence of frontotemporal lobar degeneration (FTLD) and to define the frequencies of different FTLD phenotypes in the general population. METHODS The study was conducted from January 1, 2017, to December 31, 2017, in 2 Italian provinces: Lecce (in Puglia) in the south (area 2,799.07 km2, inhabitants 802,082) and Brescia (in Lombardy) in the north (area 4,785.62 km2, inhabitants 1,262,678). During the study period, all new cases of FTLD (incident FTLD) were counted, and all patients' records were reviewed. The incidence was standardized to the Italian general population in 2017. RESULTS In the 2 provinces, 63 patients with FTLD were diagnosed. The incidence rate for FTLD was 3.05 (95% confidence interval [CI] 2.34-3.90) per 100,000 person-years (py), while the age-sex standardized incidence rate was 3.09 (95% CI 2.95-3.23) per 100,000 py. In the Italian population, the lifetime risk was 1:400. There was a progressive increase in FTLD incidence across age groups, reaching its peak in the 75- to 79-year-old group, with an incidence rate of 15.97 (95% CI 8.94-26.33) per 100,000 py. The behavioral variant of frontotemporal dementia was the most common phenotype (37%). No difference in crude incidence rate between the 2 provinces was observed. CONCLUSION FTLD is a more common form of dementia than previously recognized, with a risk spanning in a wide age range and with maximum incidence in the mid-70s. Improved knowledge of FTLD epidemiology will help to provide appropriate public health service policies.
Collapse
Affiliation(s)
- Giancarlo Logroscino
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy.
| | - Marco Piccininni
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Giuliano Binetti
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Chiara Zecca
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Rosanna Turrone
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Rosa Capozzo
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Rosanna Tortelli
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Petronilla Battista
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Eriola Bagoj
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Roberta Barone
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Silvia Fostinelli
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Luisa Benussi
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Roberta Ghidoni
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Alessandro Padovani
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Stefano F Cappa
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Antonella Alberici
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| | - Barbara Borroni
- From the Center for Neurodegenerative Diseases and the Aging Brain (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Department of Clinical Research in Neurology, and Department of Basic Medical Sciences, Neuroscience and Sense Organs (G.L., M.P., P.B.), University of Bari "Aldo Moro"; "Pia Fondazione Cardinale G. Panico" (G.L., M.P., C.Z., R.C., R. Tortelli, P.B., R.B.), Tricase, Lecce; IRCCS Centro San Giovanni di Dio Fatebenefratelli (G.B., S.F., L.B., R.G., S.F.C.); Department of Clinical and Experimental Sciences (R. Turrone, A.P., A.A., B.B.), Neurology Unit, University of Brescia; Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA) (P.B.), University of Firenze; and Department of Neurology (E.B.), Ospedali Riuniti, Foggia, Italy
| |
Collapse
|
88
|
Andrés-Benito P, Gelpi E, Povedano M, Ausín K, Fernández-Irigoyen J, Santamaría E, Ferrer I. Combined Transcriptomics and Proteomics in Frontal Cortex Area 8 in Frontotemporal Lobar Degeneration Linked to C9ORF72 Expansion. J Alzheimers Dis 2019; 68:1287-1307. [DOI: 10.3233/jad-181123] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Pol Andrés-Benito
- Neuropathology, Pathologic Anatomy Service, Bellvitge University Hospital - Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Hospitalet de Llobregat, Spain
| | - Ellen Gelpi
- Neurological Tissue Bank of the Biobanc-Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Mónica Povedano
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Service of Neurology, Bellvitge University Hospital, Hospitalet de Llobregat, Spain
| | - Karina Ausín
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain
- Clinical Neuroproteomics group and Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Department of Health, Public University of Navarra, Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain
- Clinical Neuroproteomics group and Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Department of Health, Public University of Navarra, Pamplona, Spain
| | - Enrique Santamaría
- IDISNA, Navarra Institute for Health Research, Pamplona, Spain
- Clinical Neuroproteomics group and Proteored-ISCIII, Proteomics Unit, Navarrabiomed, Department of Health, Public University of Navarra, Pamplona, Spain
| | - Isidro Ferrer
- Neuropathology, Pathologic Anatomy Service, Bellvitge University Hospital - Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
- Biomedical Network Research Center on Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Hospitalet de Llobregat, Spain
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Hospitalet de Llobregat, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| |
Collapse
|
89
|
van der Ende EL, Meeter LH, Stingl C, van Rooij JGJ, Stoop MP, Nijholt DAT, Sanchez-Valle R, Graff C, Öijerstedt L, Grossman M, McMillan C, Pijnenburg YAL, Laforce R, Binetti G, Benussi L, Ghidoni R, Luider TM, Seelaar H, van Swieten JC. Novel CSF biomarkers in genetic frontotemporal dementia identified by proteomics. Ann Clin Transl Neurol 2019; 6:698-707. [PMID: 31019994 PMCID: PMC6469343 DOI: 10.1002/acn3.745] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To identify novel CSF biomarkers in GRN-associated frontotemporal dementia (FTD) by proteomics using mass spectrometry (MS). METHODS Unbiased MS was applied to CSF samples from 19 presymptomatic and 9 symptomatic GRN mutation carriers and 24 noncarriers. Protein abundances were compared between these groups. Proteins were then selected for validation if identified by ≥4 peptides and if fold change was ≤0.5 or ≥2.0. Validation and absolute quantification by parallel reaction monitoring (PRM), a high-resolution targeted MS method, was performed on an international cohort (n = 210) of presymptomatic and symptomatic GRN, C9orf72 and MAPT mutation carriers. RESULTS Unbiased MS revealed 20 differentially abundant proteins between symptomatic mutation carriers and noncarriers and nine between symptomatic and presymptomatic carriers. Seven of these proteins fulfilled our criteria for validation. PRM analyses revealed that symptomatic GRN mutation carriers had significantly lower levels of neuronal pentraxin receptor (NPTXR), receptor-type tyrosine-protein phosphatase N2 (PTPRN2), neurosecretory protein VGF, chromogranin-A (CHGA), and V-set and transmembrane domain-containing protein 2B (VSTM2B) than presymptomatic carriers and noncarriers. Symptomatic C9orf72 mutation carriers had lower levels of NPTXR, PTPRN2, CHGA, and VSTM2B than noncarriers, while symptomatic MAPT mutation carriers had lower levels of NPTXR and CHGA than noncarriers. INTERPRETATION We identified and validated five novel CSF biomarkers in GRN-associated FTD. Our results show that synaptic, secretory vesicle, and inflammatory proteins are dysregulated in the symptomatic stage and may provide new insights into the pathophysiology of genetic FTD. Further validation is needed to investigate their clinical applicability as diagnostic or monitoring biomarkers.
Collapse
Affiliation(s)
- Emma L van der Ende
- Department of Neurology Erasmus Medical Center PO Box 2040 3015 GD Rotterdam The Netherlands
| | - Lieke H Meeter
- Department of Neurology Erasmus Medical Center PO Box 2040 3015 GD Rotterdam The Netherlands
| | - Christoph Stingl
- Laboratory of Neuro-oncology Clinical and Cancer Proteomics Department of Neurology Erasmus Medical Center PO Box 2040 3000 CA Rotterdam The Netherlands
| | - Jeroen G J van Rooij
- Department of Neurology Erasmus Medical Center PO Box 2040 3015 GD Rotterdam The Netherlands
- Department of Internal Medicine Erasmus Medical Center PO Box 2040 3015 GD Rotterdam The Netherlands
| | - Marcel P Stoop
- Laboratory of Neuro-oncology Clinical and Cancer Proteomics Department of Neurology Erasmus Medical Center PO Box 2040 3000 CA Rotterdam The Netherlands
| | - Diana A T Nijholt
- Laboratory of Neuro-oncology Clinical and Cancer Proteomics Department of Neurology Erasmus Medical Center PO Box 2040 3000 CA Rotterdam The Netherlands
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit Department of Neurology Hospital Clínic Institut d'Investigació Biomèdica August Pi i Sunyer Villarroel, 170 08036 Barcelona Spain
| | - Caroline Graff
- Division of Neurogeriatrics Department NVS Karolinska Institutet Center for Alzheimer Research Visionsgatan 4 171 64 Solna Stockholm Sweden
- Unit for Hereditary Dementias Theme Aging Karolinska University Hospital-Solna 171 64 Stockholm Sweden
| | - Linn Öijerstedt
- Division of Neurogeriatrics Department NVS Karolinska Institutet Center for Alzheimer Research Visionsgatan 4 171 64 Solna Stockholm Sweden
- Unit for Hereditary Dementias Theme Aging Karolinska University Hospital-Solna 171 64 Stockholm Sweden
| | - Murray Grossman
- Department of Neurology Penn Frontotemporal Degeneration Center University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Corey McMillan
- Department of Neurology Penn Frontotemporal Degeneration Center University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Yolande A L Pijnenburg
- Alzheimer Center and Department of Neurology Neuroscience Campus Amsterdam VU University Medical Center PO Box 7057 1007 MB Amsterdam The Netherlands
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire (CIME) CHU de Québec Département des Sciences Neurologiques Université Laval Québec Québec Canada
| | - Giuliano Binetti
- Molecular Markers Laboratory IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli via Pilastroni 4 Brescia 25125 Italy
- MAC Memory Clinic IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli via Pilastroni 4 Brescia 25125 Italy
| | - Luisa Benussi
- Molecular Markers Laboratory IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli via Pilastroni 4 Brescia 25125 Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli via Pilastroni 4 Brescia 25125 Italy
| | - Theo M Luider
- Laboratory of Neuro-oncology Clinical and Cancer Proteomics Department of Neurology Erasmus Medical Center PO Box 2040 3000 CA Rotterdam The Netherlands
| | - Harro Seelaar
- Department of Neurology Erasmus Medical Center PO Box 2040 3015 GD Rotterdam The Netherlands
| | - John C van Swieten
- Department of Neurology Erasmus Medical Center PO Box 2040 3015 GD Rotterdam The Netherlands
| |
Collapse
|
90
|
Öijerstedt L, Chiang HH, Björkström J, Forsell C, Lilius L, Lindström AK, Thonberg H, Graff C. Confirmation of high frequency of C9orf72 mutations in patients with frontotemporal dementia from Sweden. Neurobiol Aging 2019; 84:241.e21-241.e25. [PMID: 30992141 DOI: 10.1016/j.neurobiolaging.2019.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/05/2019] [Accepted: 03/19/2019] [Indexed: 12/27/2022]
Abstract
Frontotemporal dementia (FTD) is the second most common early-onset dementia. Up to half of the cases are familial, and several mutations have been identified as pathogenic. Repeat expansion mutations in C9orf72 are the most common genetic cause of FTD and are particularly frequent in Sweden and Finland. We aimed to determine the mutation frequency in patients with FTD ascertained at a memory clinic in Sweden and assess the inheritance pattern in the families. We screened 132 patients with FTD for mutations in C9orf72, GRN, and MAPT, and the frequency was 34.1%. Two novel variations, not previously published, were found; a pathogenic GRN mutation and a MAPT variation in intron 9 that we report as VUS. The likelihood of finding a mutation was highest in patients with a clear family history of dementia or motor neuron disease (76%), but mutations were also found in apparent sporadic cases. This confirms that FTD cohorts from Sweden have a relatively higher risk of an underlying mutation in all risk categories compared with other reported cohorts.
Collapse
Affiliation(s)
- Linn Öijerstedt
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Karolinska University Hospital, Theme Aging, Unit for Hereditary Dementias QA12, Stockholm, Sweden
| | - Huei-Hsin Chiang
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Jenny Björkström
- Karolinska University Hospital, Theme Aging, Unit for Hereditary Dementias QA12, Stockholm, Sweden
| | - Charlotte Forsell
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Lena Lilius
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Anna-Karin Lindström
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Thonberg
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Karolinska University Hospital, Theme Aging, Unit for Hereditary Dementias QA12, Stockholm, Sweden
| | - Caroline Graff
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Karolinska University Hospital, Theme Aging, Unit for Hereditary Dementias QA12, Stockholm, Sweden.
| |
Collapse
|
91
|
Riemslagh FW, Lans H, Seelaar H, Severijnen LAWFM, Melhem S, Vermeulen W, Aronica E, Pasterkamp RJ, van Swieten JC, Willemsen R. HR23B pathology preferentially co-localizes with p62, pTDP-43 and poly-GA in C9ORF72-linked frontotemporal dementia and amyotrophic lateral sclerosis. Acta Neuropathol Commun 2019; 7:39. [PMID: 30867060 PMCID: PMC6416930 DOI: 10.1186/s40478-019-0694-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022] Open
Abstract
Human homologue of yeast UV excision repair protein Rad23b (HR23B) inclusions are found in a number of neurodegenerative diseases, including frontotemporal dementia (FTD), Huntington's disease (HD), spinocerebellar ataxia type 3 and 7 (SCA3/7), fragile X associated tremor/ataxia syndrome (FXTAS) and Parkinson's disease (PD). Here, we describe HR23B pathology in C9ORF72 linked FTD and amyotrophic lateral sclerosis (ALS) cases. HR23B presented in neuropils, intranuclear inclusions and cytoplasmic and perinuclear inclusions and was predominantly found in cortices (frontal, temporal and motor), spinal cord and hippocampal dentate gyrus. HR23B co-localized with poly-GA-, pTDP-43- and p62-positive inclusions in frontal cortex and in hippocampal dentate gyrus, the latter showing higher co-localization percentages. HR23B binding partners XPC, 20S and ataxin-3, which are involved in nucleotide excision repair (NER) and the ubiquitin-proteasome system (UPS), did not show an aberrant distribution. However, C9ORF72 fibroblasts were more sensitive for UV-C damage than healthy control fibroblasts, even though all factors involved in NER localized normally to DNA damage and the efficiency of DNA repair was not reduced. HR23Bs other binding partner NGly1/PNGase, involved in ER-associated degradation (ERAD) of misfolded proteins, was not expressed in the majority of neurons in C9FTD/ALS brain sections compared to non-demented controls. Our results suggest a difference in HR23B aggregation and co-localization pattern with DPRs, pTDP-43 and p62 between different brain areas from C9FTD/ALS cases. We hypothesize that HR23B may play a role in C9ORF72 pathogenesis, possibly by aberrant ERAD functioning.
Collapse
|
92
|
Gittings LM, Foti SC, Benson BC, Gami-Patel P, Isaacs AM, Lashley T. Heterogeneous nuclear ribonucleoproteins R and Q accumulate in pathological inclusions in FTLD-FUS. Acta Neuropathol Commun 2019; 7:18. [PMID: 30755280 PMCID: PMC6371513 DOI: 10.1186/s40478-019-0673-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/03/2019] [Indexed: 12/14/2022] Open
Abstract
Frontotemporal lobar degeneration (FTLD) is pathologically subdivided based on the presence of particular pathological proteins that are identified in inclusion bodies observed post-mortem. The FTLD-FUS subgroup is defined by the presence of the fused in sarcoma protein (FUS) in pathological inclusions. FUS is a heterogeneous nuclear ribonucleoprotein (hnRNP) protein and a member of the FET (FUS, EWS, TAF15) protein family. It shuttles between the nucleus and cytoplasm, and has been implicated in many cellular functions including translation, splicing, and RNA transport. EWS, TAF15 and the nuclear import receptor transportin have been shown to co-accumulate with FUS in neuronal inclusions specifically in FTLD-FUS, with transportin-positive inclusions most frequently observed. Here, we report the identification of hnRNP R and hnRNP Q in neuronal cytoplasmic and intranuclear inclusions in the frontal cortex and hippocampus of FTLD-FUS patients, as frequently as transportin. hnRNP R and hnRNP Q were not found in the characteristic pathological inclusions observed in FTLD-TDP (subtypes A-C). Additionally, we studied the expression of hnRNP R in the frontal and temporal cortices from patients with FTLD and found significantly increased expression of the heterogeneous nuclear ribonucleoprotein R in several FTLD disease groups. Our identification of the frequent presence of hnRNP R and hnRNP Q in FTLD-FUS inclusions suggests a potential role for these hnRNPs in FTLD-FUS pathogenesis and supports the role of dysfunctional RNA metabolism in FTLD.
Collapse
|
93
|
Amygdala subnuclei are differentially affected in the different genetic and pathological forms of frontotemporal dementia. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2019; 11:136-141. [PMID: 30788410 PMCID: PMC6369146 DOI: 10.1016/j.dadm.2018.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction Frontotemporal dementia (FTD) is a heterogeneous neurodegenerative disorder with multiple genetic and pathological causes. It is characterized by both cortical and subcortical atrophies, with previous studies showing early involvement of the amygdala. However, no prior study has specifically investigated the atrophy of different subnuclei of the amygdala. Methods Using an automated segmentation tool for T1-weighted volumetric magnetic resonance imaging, we investigated amygdalar subnuclei (AS) involvement in a cohort of 132 patients with genetic or pathologically confirmed FTD (age: mean = 61 years (standard deviation = 8); disease duration: 5 (3) years) compared with 107 age-matched controls. Results AS were affected in all genetic and pathological forms of FTD. MAPT mutations/FTDP-17, Pick's disease, and transactive response DNA binding protein 43 kDa type C were the forms with the smallest amygdala (35%–50% smaller than controls in the most affected hemisphere, P < .0005). In most FTD groups, medial subnuclei (particularly the superficial, accessory basal and basal/paralaminar subnuclei) tended to be affected more than the lateral subnuclei, except for the progressive supranuclear palsy group, in which the corticoamygdaloid transition area was the least-affected area. Discussion Differential involvement of the AS was seen in the different genetic and pathological forms of FTD. In general, the most affected subnuclei were the superficial, accessory basal and basal/paralaminar subnuclei, which form part of a network of regions that control reward and emotion regulation, functions known to be particularly affected in FTD. Amygdalar subnuclei are affected in all genetic and pathological forms of frontotemporal dementia. Amygdalar subnuclei are differently involved among genetic and pathological forms. MAPT/FTDP-17, Pick's disease, and TDP-43 type C showed the smallest amygdala. The medial subnuclei tended to be affected more than the lateral subnuclei.
Collapse
|
94
|
Forrest SL, Halliday GM, McCann H, McGeachie AB, McGinley CV, Hodges JR, Piguet O, Kwok JB, Spillantini MG, Kril JJ. Heritability in frontotemporal tauopathies. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2019; 11:115-124. [PMID: 30723775 PMCID: PMC6351353 DOI: 10.1016/j.dadm.2018.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction Exploring the degree of heritability in a large cohort of frontotemporal lobar degeneration with tau-immunopositive inclusions (FTLD-tau) and determining if different FTLD-tau subtypes are associated with stronger heritability will provide important insight into disease pathogenesis. Methods Using modified Goldman pedigree classifications, heritability was examined in pathologically proven FTLD-tau cases with dementia at any time (n = 124) from the Sydney-Cambridge collection. Results Thirteen percent of the FTLD-tau cohort have a suggested autosomal dominant pattern of inheritance, 25% have some family history, and 62% apparently sporadic. MAPT mutations were found in 9% of cases. Globular glial tauopathy was associated with the strongest heritability with 40% having a suggested autosomal dominant pattern of inheritance followed by corticobasal degeneration (19%), Pick's disease (8%), and progressive supranuclear palsy (6%). Discussion Similar to clinical frontotemporal dementia syndromes, heritability varies between pathological subtypes. Further identification of a genetic link in cases with strong heritability await discovery.
Collapse
Affiliation(s)
- Shelley L Forrest
- Faculty of Medicine and Health, Charles Perkins Centre and Discipline of Pathology, University of Sydney, Sydney, Australia
| | - Glenda M Halliday
- Faculty of Medicine and Health, Brain and Mind Centre and Central Clinical School, University of Sydney, Sydney, Australia.,Neuroscience Research Australia, Sydney, Australia.,School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | | | | - Ciara V McGinley
- Faculty of Medicine and Health, Charles Perkins Centre and Discipline of Pathology, University of Sydney, Sydney, Australia
| | - John R Hodges
- Faculty of Medicine and Health, Brain and Mind Centre and Central Clinical School, University of Sydney, Sydney, Australia.,Neuroscience Research Australia, Sydney, Australia.,School of Medical Sciences, University of New South Wales, Sydney, Australia.,ARC Centre of Excellence in Cognition and its Disorders, Sydney, Australia
| | - Olivier Piguet
- Neuroscience Research Australia, Sydney, Australia.,ARC Centre of Excellence in Cognition and its Disorders, Sydney, Australia.,Brain and Mind Centre and School of Psychology, University of Sydney, Sydney, Australia
| | - John B Kwok
- Faculty of Medicine and Health, Brain and Mind Centre and Central Clinical School, University of Sydney, Sydney, Australia.,Neuroscience Research Australia, Sydney, Australia.,School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Maria G Spillantini
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Jillian J Kril
- Faculty of Medicine and Health, Charles Perkins Centre and Discipline of Pathology, University of Sydney, Sydney, Australia
| |
Collapse
|
95
|
Revesz T, Mann DM, Lashley T. Frontotemporal lobar degenerations: from basic science to clinical manifestations. Neuropathol Appl Neurobiol 2019; 45:3-5. [PMID: 30666692 DOI: 10.1111/nan.12537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- T Revesz
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - D M Mann
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Salford Royal Hospital, Salford, UK
| | - T Lashley
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.,Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
96
|
TDP-43 extracted from frontotemporal lobar degeneration subject brains displays distinct aggregate assemblies and neurotoxic effects reflecting disease progression rates. Nat Neurosci 2018; 22:65-77. [PMID: 30559480 DOI: 10.1038/s41593-018-0294-y] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022]
Abstract
Accumulation of abnormally phosphorylated TDP-43 (pTDP-43) is the main pathology in affected neurons of people with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Morphological diversity and neuroanatomical distribution of pTDP-43 accumulations allowed classification of FTLD cases into at least four subtypes, which are correlated with clinical presentations and genetic causes. To understand the molecular basis of this heterogeneity, we developed SarkoSpin, a new method for biochemical isolation of pathological TDP-43. By combining SarkoSpin with mass spectrometry, we revealed proteins beyond TDP-43 that become abnormally insoluble in a disease subtype-specific manner. We show that pTDP-43 extracted from brain forms stable assemblies of distinct densities and morphologies that are associated with disease subtypes. Importantly, biochemically extracted pTDP-43 assemblies showed differential neurotoxicity and seeding that were correlated with disease duration of FTLD subjects. Our data are consistent with the notion that disease heterogeneity could originate from alternate pathological TDP-43 conformations, which are reminiscent of prion strains.
Collapse
|
97
|
Pattamatta A, Cleary JD, Ranum LPW. All in the Family: Repeats and ALS/FTD. Trends Neurosci 2018; 41:247-250. [PMID: 29703376 DOI: 10.1016/j.tins.2018.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 12/12/2022]
Abstract
In 2011, an intronic (G4C2)•(G2C4) expansion was shown to cause the most common forms of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). This discovery linked ALS with a clinically distinct form of dementia and a larger group of microsatellite repeat diseases, and catalyzed basic and translational research.
Collapse
Affiliation(s)
- Amrutha Pattamatta
- Center for Neurogenetics, College of Medicine, University of Florida, Gainesville, FL, USA; Departments of Molecular Genetics and Neurology, College of Medicine, University of Florida, Gainesville, FA, USA
| | - John D Cleary
- Center for Neurogenetics, College of Medicine, University of Florida, Gainesville, FL, USA; Departments of Molecular Genetics and Neurology, College of Medicine, University of Florida, Gainesville, FA, USA
| | - Laura P W Ranum
- Center for Neurogenetics, College of Medicine, University of Florida, Gainesville, FL, USA; Departments of Molecular Genetics and Neurology, College of Medicine, University of Florida, Gainesville, FA, USA; McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Genetics Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
98
|
Sun Y, Medina Cruz A, Hadley KC, Galant NJ, Law R, Vernon RM, Morris VK, Robertson J, Chakrabartty A. Physiologically Important Electrolytes as Regulators of TDP-43 Aggregation and Droplet-Phase Behavior. Biochemistry 2018; 58:590-607. [DOI: 10.1021/acs.biochem.8b00842] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yulong Sun
- University Health Network, Princess Margaret Cancer Centre, University of Toronto, TMDT 4-305, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Alison Medina Cruz
- Department of Medical Biophysics, Princess Margaret Cancer Centre, University of Toronto, TMDT 4-305, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Kevin C. Hadley
- University Health Network, Princess Margaret Cancer Centre, University of Toronto, TMDT 4-305, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Natalie J. Galant
- University Health Network, Princess Margaret Cancer Centre, University of Toronto, TMDT 4-305, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Ryan Law
- Department of Medical Biophysics, Princess Margaret Cancer Centre, University of Toronto, TMDT 4-305, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Robert M. Vernon
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Vanessa K. Morris
- University Health Network, Princess Margaret Cancer Centre, University of Toronto, TMDT 4-305, 101 College Street, Toronto, ON M5G 1L7, Canada
- School of Biological Sciences, University of Canterbury, Ilam, Christchurch 8041, New Zealand
| | - Janice Robertson
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5T 2S8, Canada
| | - Avijit Chakrabartty
- University Health Network, Princess Margaret Cancer Centre, University of Toronto, TMDT 4-305, 101 College Street, Toronto, ON M5G 1L7, Canada
- Department of Medical Biophysics, Princess Margaret Cancer Centre, University of Toronto, TMDT 4-305, 101 College Street, Toronto, ON M5G 1L7, Canada
- Department of Biochemistry, University of Toronto, TMDT 4-305, 101 College Street, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
99
|
Prioni S, Redaelli V, Soliveri P, Fetoni V, Barocco F, Caffarra P, Scaglioni A, Tramacere I, Girotti F. Stereotypic behaviours in frontotemporal dementia and progressive supranuclear palsy. Cortex 2018; 109:272-278. [PMID: 30399478 DOI: 10.1016/j.cortex.2018.09.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 06/04/2018] [Accepted: 09/27/2018] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The behavioural variant of frontotemporal dementia (bvFTD), and the Richardson variant of progressive supranuclear palsy (PSP-RS) share several clinical signs and symptoms. Since stereotypic behaviours are fairly common in bvFTD, and are also described in other degenerative dementias including Alzheimer's disease, and parkinsonisms with dementia, we aimed to examine the extent to which stereotypies also characterise PSP-RS. METHODS We compared 53 bvFTD patients with 40 demented PSP-RS patients, seen consecutively as outpatients at four Italian Hospitals. Patients were assessed by the Neuropsychiatric Inventory (NPI); Mini-Mental State Examination (MMSE) and Frontal Assessment Battery (FAB) for cognitive functions; Stereotypy Rating Inventory (SRI) for stereotypies; Unified Parkinson's Disease Rating Scale (UPDRS) for motor function; and Activities of Daily Living (ADL) to assess autonomy in daily life. RESULTS The groups did not differ for age, illness duration, cognitive functions or total NPI score; PSP-RS had significantly more depressive symptoms and greater motor and autonomy compromise than bvFTD. The groups did not differ significantly on total SRI score, but bvFTD had significantly more cooking and eating stereotypies. Twenty-three (57.5%) PSP-RS and 43 (81%) bvFTD patients had at least one stereotypy; 16/23 (69.5%) PSP-RS and 9/43 (20.9%) bvFTD patients appeared aware of their stereotypies. CONCLUSION Stereotypies were common in our demented PSP-RS patients. Further studies on earlier stage non-demented PSP patients are required to ascertain whether stereotypies are characteristic of PSP in general or are confined to PSP-RS, and whether they may be used to suggest a PSP diagnosis early in disease course.
Collapse
Affiliation(s)
- Sara Prioni
- Neurology Department, C. Besta Neurological Institute and Foundation (IRCCS), Milano, Italy
| | - Veronica Redaelli
- Neurology Department, C. Besta Neurological Institute and Foundation (IRCCS), Milano, Italy
| | - Paola Soliveri
- Neurology Department, C. Besta Neurological Institute and Foundation (IRCCS), Milano, Italy
| | | | | | | | | | - Irene Tramacere
- Research and Clinical Development Department, C. Besta Neurological Institute and Foundation (IRCCS), Milano, Italy
| | - Floriano Girotti
- Neurology Department, C. Besta Neurological Institute and Foundation (IRCCS), Milano, Italy.
| |
Collapse
|
100
|
Steinacker P, Barschke P, Otto M. Biomarkers for diseases with TDP-43 pathology. Mol Cell Neurosci 2018; 97:43-59. [PMID: 30399416 DOI: 10.1016/j.mcn.2018.10.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/26/2018] [Accepted: 10/29/2018] [Indexed: 01/01/2023] Open
Abstract
The discovery that aggregated transactive response DNA-binding protein 43 kDa (TDP-43) is the major component of pathological ubiquitinated inclusions in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) caused seminal progress in the unveiling of the genetic bases and molecular characteristics of these now so-called TDP-43 proteinopathies. Substantial increase in the knowledge of clinic-pathological coherencies, especially for FTLD variants, could be made in the last decade, but also revealed a considerable complexity of TDP-43 pathology and often a poor correlation of clinical and molecular disease characteristics. To date, an underlying TDP-43 pathology can be predicted only for patients with mutations in the genes C9orf72 and GRN, but is dependent on neuropathological verification in patients without family history, which represent the majority of cases. As etiology-specific therapies for neurodegenerative proteinopathies are emerging, methods to forecast TDP-43 pathology at patients' lifetime are highly required. Here, we review the current status of research pursued to identify specific indicators to predict or exclude TDP-43 pathology in the ALS-FTLD spectrum disorders and findings on candidates for prognosis and monitoring of disease progression in TDP-43 proteinopathies with a focus on TDP-43 with its pathological forms, neurochemical and imaging biomarkers.
Collapse
Affiliation(s)
| | - Peggy Barschke
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany.
| |
Collapse
|