1
|
Fare CM, Rothstein JD. Nuclear pore dysfunction and disease: a complex opportunity. Nucleus 2024; 15:2314297. [PMID: 38383349 PMCID: PMC10883112 DOI: 10.1080/19491034.2024.2314297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024] Open
Abstract
The separation of genetic material from bulk cytoplasm has enabled the evolution of increasingly complex organisms, allowing for the development of sophisticated forms of life. However, this complexity has created new categories of dysfunction, including those related to the movement of material between cellular compartments. In eukaryotic cells, nucleocytoplasmic trafficking is a fundamental biological process, and cumulative disruptions to nuclear integrity and nucleocytoplasmic transport are detrimental to cell survival. This is particularly true in post-mitotic neurons, where nuclear pore injury and errors to nucleocytoplasmic trafficking are strongly associated with neurodegenerative disease. In this review, we summarize the current understanding of nuclear pore biology in physiological and pathological contexts and discuss potential therapeutic approaches for addressing nuclear pore injury and dysfunctional nucleocytoplasmic transport.
Collapse
Affiliation(s)
- Charlotte M Fare
- Department of Neurology and Brain Science Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey D Rothstein
- Department of Neurology and Brain Science Institute, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
2
|
Paris A, Lakatos A. Cell and gene therapy for amyotrophic lateral sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:217-241. [PMID: 39341656 DOI: 10.1016/b978-0-323-90120-8.00017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disorder with rapidly progressive skeletal muscle weakness, which can also cause a variable cognitive deficit. Genetic causes are only identified in approximately 10% of all cases, with complex genotype-phenotype associations, making it challenging to identify treatment targets. What further hampers therapeutic development is a broad heterogeneity in mechanisms, possible targets, and disturbances across various cell types, aside from the cortical and spinal motor neurons that lie at the heart of the pathology of ALS. Over the last decade, significant progress in biotechnologic techniques, cell and ribonucleic acid (RNA) engineering, animal models, and patient-specific human stem cell and organoid models have accelerated both mechanistic and therapeutic discoveries. The growing number of clinical trials mirrors this. This chapter reviews the current state of human preclinical models supporting trial strategies as well as recent clinical cell and gene therapy approaches.
Collapse
Affiliation(s)
- Alvar Paris
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom; Department of Neurology, Cambridge University Hospitals NHS Trust, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - András Lakatos
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom; Department of Neurology, Cambridge University Hospitals NHS Trust, Addenbrooke's Hospital, Cambridge, United Kingdom.
| |
Collapse
|
3
|
McCluskey G, Morrison KE, Donaghy C, McConville J, McCarron MO, McVerry F, Duddy W, Duguez S. Serum Neurofilaments in Motor Neuron Disease and Their Utility in Differentiating ALS, PMA and PLS. Life (Basel) 2023; 13:1301. [PMID: 37374084 DOI: 10.3390/life13061301] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Neurofilament levels are elevated in many neurodegenerative diseases and have shown promise as diagnostic and prognostic biomarkers in Amyotrophic Lateral Sclerosis (ALS), the most common form of Motor Neuron Disease (MND). This study assesses serum neurofilament light (NFL) and neurofilament heavy (NFH) chain concentrations in patients with ALS, other variants of motor neuron disease such as Progressive Muscular Atrophy (PMA) and Primary Lateral Sclerosis (PLS), and a range of other neurological diseases. It aims to evaluate the use of NFL and NFH to differentiate these conditions and for the prognosis of MND disease progression. NFL and NFH levels were quantified using electrochemiluminescence immunoassays (ECLIA). Both were elevated in 47 patients with MND compared to 34 patients with other neurological diseases and 33 healthy controls. NFL was able to differentiate patients with MND from the other groups with a Receiver Operating Characteristic (ROC) curve area under the curve (AUC) of 0.90 (p < 0.001). NFL correlated with the rate of disease progression in MND (rho 0.758, p < 0.001) and with the ALS Functional Rating Scale (rho -0.335, p = 0.021). NFL levels were higher in patients with ALS compared to both PMA (p = 0.032) and PLS (p = 0.012) and were able to distinguish ALS from both PMA and PLS with a ROC curve AUC of 0.767 (p = 0.005). These findings support the use of serum NFL to help diagnose and differentiate types of MND, in addition to providing prognostic information to patients and their families.
Collapse
Affiliation(s)
- Gavin McCluskey
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry BT47 6SB, UK
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Department of Neurology, Altnagelvin Hospital, Derry BT47 6SB, UK
| | - Karen E Morrison
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Faculty of Medicine, Health & Life Sciences, Queen's University, Belfast BT9 6AG, UK
| | - Colette Donaghy
- Department of Neurology, Altnagelvin Hospital, Derry BT47 6SB, UK
| | - John McConville
- Department of Neurology, Royal Victoria Hospital, Belfast BT12 6BA, UK
- Department of Neurology, Ulster Hospital, Belfast BT16 1RH, UK
| | - Mark O McCarron
- Department of Neurology, Altnagelvin Hospital, Derry BT47 6SB, UK
| | - Ferghal McVerry
- Department of Neurology, Altnagelvin Hospital, Derry BT47 6SB, UK
| | - William Duddy
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry BT47 6SB, UK
| | - Stephanie Duguez
- Personalised Medicine Centre, School of Medicine, Ulster University, Derry BT47 6SB, UK
| |
Collapse
|
4
|
Jiang Q, Lin J, Wei Q, Li C, Hou Y, Zhang L, Ou R, Liu K, Yang T, Xiao Y, Hadano S, Shang H. Genetic and clinical characteristics of ALS patients with NEK1 gene variants. Neurobiol Aging 2023; 123:191-199. [PMID: 36443167 DOI: 10.1016/j.neurobiolaging.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/19/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2022]
Abstract
NIMA-related kinase 1(NEK1) gene was related to amyotrophic lateral sclerosis (ALS). However, genetic spectrum and clinical characteristics of ALS patients with NEK1 variants was largely unknown. We conducted genetic analysis on 1587 Chinese ALS patients and used software to predict the pathogenicity of NEK1 missense variant. We searched the literatures in PubMed, Embase, and Web of Science. In our ALS cohort, 42 ALS patients (2.6%) carried NEK1 variants, including 10 novel loss-of-function (LoF) variant carriers and 32 missense variant carriers. 90% of the NEK1 LoF variant carriers had upper limbs onset. The median survival time of LoF variant carriers tend to be shorter than that of probably pathogenic variant carriers (23.80 vs. 42.77 months). In 16 related studies, 167 different NEK1 variants, including 62 LoF and 105 missense variants, were found in 237 reported ALS patients. It was found that the survival time of LoF variant carriers was significantly shorter than that of missense variant carriers. Our study expanded the genotype and phenotype spectrum of ALS patients with NEK1 variants.
Collapse
Affiliation(s)
- Qirui Jiang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junyu Lin
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanbing Hou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingyu Zhang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kuncheng Liu
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianmi Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Xiao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shinji Hadano
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
5
|
Meo G, Ferraro PM, Cillerai M, Gemelli C, Cabona C, Zaottini F, Roccatagliata L, Villani F, Schenone A, Caponnetto C. MND Phenotypes Differentiation: The Role of Multimodal Characterization at the Time of Diagnosis. Life (Basel) 2022; 12:life12101506. [PMID: 36294940 PMCID: PMC9604895 DOI: 10.3390/life12101506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/13/2022] [Accepted: 09/21/2022] [Indexed: 11/25/2022] Open
Abstract
Pure/predominant upper motor neuron (pUMN) and lower motor neuron (pLMN) diseases have significantly better prognosis compared to amyotrophic lateral sclerosis (ALS), but their early differentiation is often challenging. We therefore tested whether a multimodal characterization approach embedding clinical, cognitive/behavioral, genetic, and neurophysiological data may improve the differentiation of pUMN and pLMN from ALS already by the time of diagnosis. Dunn’s and chi-squared tests were used to compare data from 41 ALS, 34 pLMN, and 19 pUMN cases with diagnoses confirmed throughout a 2-year observation period. Area under the curve (AUC) analyses were implemented to identify the finest tools for phenotypes discrimination. Relative to ALS, pLMN showed greater lower limbs weakness, lower UMN burden, and progression rate (p < 0.001−0.04). PUMN showed a greater frequency of lower limbs onset, higher UMN burden, lower ALSFRS-r and MRC progression rates (p < 0.001−0.03), and greater ulnar compound muscle action potential (CMAP) amplitude and tibial central motor conduction time (CMCT) (p = 0.05−0.03). The UMN progression rate was the finest measure to identify pLMN cases (AUC = 90%), while the MRC progression rate was the finest tool to identify pUMN (AUC = 82%). Detailed clinical and neurophysiological examinations may significantly improve MNDs differentiation, facilitating prognosis estimation and ameliorating stratification strategies for clinical trials enrollment.
Collapse
Affiliation(s)
- Giuseppe Meo
- Department of Neurology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16126 Genoa, Italy
| | - Pilar M. Ferraro
- Department of Neurology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence: ; Tel.: +39-01-0353-7040
| | - Marta Cillerai
- Department of Neurology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16126 Genoa, Italy
| | - Chiara Gemelli
- Department of Neurology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Corrado Cabona
- Division of Clinical Neurophysiology and Epilepsy Center, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Federico Zaottini
- Department of Radiology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Luca Roccatagliata
- Department of Neuroradiology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, 16126 Genoa, Italy
| | - Flavio Villani
- Division of Clinical Neurophysiology and Epilepsy Center, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Angelo Schenone
- Department of Neurology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16126 Genoa, Italy
| | - Claudia Caponnetto
- Department of Neurology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
6
|
Cozzi M, Ferrari V. Autophagy Dysfunction in ALS: from Transport to Protein Degradation. J Mol Neurosci 2022; 72:1456-1481. [PMID: 35708843 PMCID: PMC9293831 DOI: 10.1007/s12031-022-02029-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/17/2022] [Indexed: 01/18/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting upper and lower motor neurons (MNs). Since the identification of the first ALS mutation in 1993, more than 40 genes have been associated with the disorder. The most frequent genetic causes of ALS are represented by mutated genes whose products challenge proteostasis, becoming unable to properly fold and consequently aggregating into inclusions that impose proteotoxic stress on affected cells. In this context, increasing evidence supports the central role played by autophagy dysfunctions in the pathogenesis of ALS. Indeed, in early stages of disease, high levels of proteins involved in autophagy are present in ALS MNs; but at the same time, with neurodegeneration progression, autophagy-mediated degradation decreases, often as a result of the accumulation of toxic protein aggregates in affected cells. Autophagy is a complex multistep pathway that has a central role in maintaining cellular homeostasis. Several proteins are involved in its tight regulation, and importantly a relevant fraction of ALS-related genes encodes products that directly take part in autophagy, further underlining the relevance of this key protein degradation system in disease onset and progression. In this review, we report the most relevant findings concerning ALS genes whose products are involved in the several steps of the autophagic pathway, from phagophore formation to autophagosome maturation and transport and finally to substrate degradation.
Collapse
Affiliation(s)
- Marta Cozzi
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, Università Degli Studi Di Milano, 20133, Milan, Italy.
| | - Veronica Ferrari
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, Università Degli Studi Di Milano, 20133, Milan, Italy.
| |
Collapse
|
7
|
Alteration of the Neuromuscular Junction and Modifications of Muscle Metabolism in Response to Neuron-Restricted Expression of the CHMP2Bintron5 Mutant in a Mouse Model of ALS-FTD Syndrome. Biomolecules 2022; 12:biom12040497. [PMID: 35454086 PMCID: PMC9025139 DOI: 10.3390/biom12040497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/12/2022] [Accepted: 03/21/2022] [Indexed: 02/04/2023] Open
Abstract
CHMP2B is a protein that coordinates membrane scission events as a core component of the ESCRT machinery. Mutations in CHMP2B are an uncommon cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two neurodegenerative diseases with clinical, genetic, and pathological overlap. Different mutations have now been identified across the ALS-FTD spectrum. Disruption of the neuromuscular junction is an early pathogenic event in ALS. Currently, the links between neuromuscular junction functionality and ALS-associated genes, such as CHMP2B, remain poorly understood. We have previously shown that CHMP2B transgenic mice expressing the CHMP2Bintron5 mutant specifically in neurons develop a progressive motor phenotype reminiscent of ALS. In this study, we used complementary approaches (behavior, histology, electroneuromyography, and biochemistry) to determine the extent to which neuron-specific expression of CHMP2Bintron5 could impact the skeletal muscle characteristics. We show that neuronal expression of the CHMP2Bintron5 mutant is sufficient to trigger progressive gait impairment associated with structural and functional changes in the neuromuscular junction. Indeed, CHMP2Bintron5 alters the pre-synaptic terminal organization and the synaptic transmission that ultimately lead to a switch of fast-twitch glycolytic muscle fibers to more oxidative slow-twitch muscle fibers. Taken together these data indicate that neuronal expression of CHMP2Bintron5 is sufficient to induce a synaptopathy with molecular and functional changes in the motor unit reminiscent of those found in ALS patients.
Collapse
|
8
|
Todd TW, Petrucelli L. Modelling amyotrophic lateral sclerosis in rodents. Nat Rev Neurosci 2022; 23:231-251. [PMID: 35260846 DOI: 10.1038/s41583-022-00564-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2022] [Indexed: 12/11/2022]
Abstract
The efficient study of human disease requires the proper tools, one of the most crucial of which is an accurate animal model that faithfully recapitulates the human condition. The study of amyotrophic lateral sclerosis (ALS) is no exception. Although the majority of ALS cases are considered sporadic, most animal models of this disease rely on genetic mutations identified in familial cases. Over the past decade, the number of genes associated with ALS has risen dramatically and, with each new genetic variant, there is a drive to develop associated animal models. Rodent models are of particular importance as they allow for the study of ALS in the context of a living mammal with a comparable CNS. Such models not only help to verify the pathogenicity of novel mutations but also provide critical insight into disease mechanisms and are crucial for the testing of new therapeutics. In this Review, we aim to summarize the full spectrum of ALS rodent models developed to date.
Collapse
Affiliation(s)
- Tiffany W Todd
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic Jacksonville, Jacksonville, FL, USA.
| |
Collapse
|
9
|
Chandra S, Lusk CP. Emerging Connections between Nuclear Pore Complex Homeostasis and ALS. Int J Mol Sci 2022; 23:1329. [PMID: 35163252 PMCID: PMC8835831 DOI: 10.3390/ijms23031329] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 12/26/2022] Open
Abstract
Developing effective treatments for neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) requires understanding of the underlying pathomechanisms that contribute to the motor neuron loss that defines the disease. As it causes the largest fraction of familial ALS cases, considerable effort has focused on hexanucleotide repeat expansions in the C9ORF72 gene, which encode toxic repeat RNA and dipeptide repeat (DPR) proteins. Both the repeat RNA and DPRs interact with and perturb multiple elements of the nuclear transport machinery, including shuttling nuclear transport receptors, the Ran GTPase and the nucleoporin proteins (nups) that build the nuclear pore complex (NPC). Here, we consider recent work that describes changes to the molecular composition of the NPC in C9ORF72 model and patient neurons in the context of quality control mechanisms that function at the nuclear envelope (NE). For example, changes to NPC structure may be caused by the dysregulation of a conserved NE surveillance pathway mediated by the endosomal sorting complexes required for the transport protein, CHMP7. Thus, these studies are introducing NE and NPC quality control pathways as key elements in a pathological cascade that leads to C9ORF72 ALS, opening entirely new experimental avenues and possibilities for targeted therapeutic intervention.
Collapse
Affiliation(s)
| | - C. Patrick Lusk
- Department of Cell Biology, Yale School of Medicine, 295 Congress Ave, New Haven, CT 06520, USA;
| |
Collapse
|
10
|
Lotti F, Przedborski S. Motoneuron Diseases. ADVANCES IN NEUROBIOLOGY 2022; 28:323-352. [PMID: 36066831 DOI: 10.1007/978-3-031-07167-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Motoneuron diseases (MNDs) represent a heterogeneous group of progressive paralytic disorders, mainly characterized by the loss of upper (corticospinal) motoneurons, lower (spinal) motoneurons or, often both. MNDs can occur from birth to adulthood and have a highly variable clinical presentation, even within gene-positive forms, suggesting the existence of environmental and genetic modifiers. A combination of cell autonomous and non-cell autonomous mechanisms contributes to motoneuron degeneration in MNDs, suggesting multifactorial pathogenic processes.
Collapse
Affiliation(s)
- Francesco Lotti
- Departments of Neurology, Pathology & Cell Biology, and Neuroscience, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Serge Przedborski
- Departments of Neurology, Pathology & Cell Biology, and Neuroscience, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
11
|
Spinelli EG, Ghirelli A, Basaia S, Cividini C, Riva N, Canu E, Castelnovo V, Domi T, Magnani G, Caso F, Caroppo P, Prioni S, Rossi G, Tremolizzo L, Appollonio I, Silani V, Carrera P, Filippi M, Agosta F. Structural MRI Signatures in Genetic Presentations of the Frontotemporal Dementia/Motor Neuron Disease Spectrum. Neurology 2021; 97:e1594-e1607. [PMID: 34544819 PMCID: PMC8548958 DOI: 10.1212/wnl.0000000000012702] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To assess cortical, subcortical, and cerebellar gray matter (GM) atrophy using MRI in patients with disorders of the frontotemporal lobar degeneration (FTLD) spectrum with known genetic mutations. METHODS Sixty-six patients carrying FTLD-related mutations were enrolled, including 44 with pure motor neuron disease (MND) and 22 with frontotemporal dementia (FTD). Sixty-one patients with sporadic FTLD (sFTLD) matched for age, sex, and disease severity with genetic FTLD (gFTLD) were also included, as well as 52 healthy controls. A whole-brain voxel-based morphometry (VBM) analysis was performed. GM volumes of subcortical and cerebellar structures were obtained. RESULTS Compared with controls, GM atrophy on VBM was greater and more diffuse in genetic FTD, followed by sporadic FTD and genetic MND cases, whereas patients with sporadic MND (sMND) showed focal motor cortical atrophy. Patients carrying C9orf72 and GRN mutations showed the most widespread cortical volume loss, in contrast with GM sparing in SOD1 and TARDBP. Globally, patients with gFTLD showed greater atrophy of parietal cortices and thalami compared with sFTLD. In volumetric analysis, patients with gFTLD showed volume loss compared with sFTLD in the caudate nuclei and thalami, in particular comparing C9-MND with sMND cases. In the cerebellum, patients with gFTLD showed greater atrophy of the right lobule VIIb than sFTLD. Thalamic volumes of patients with gFTLD with a C9orf72 mutation showed an inverse correlation with Frontal Behavioral Inventory scores. DISCUSSION Measures of deep GM and cerebellar structural involvement may be useful markers of gFTLD, particularly C9orf72-related disorders, regardless of the clinical presentation within the FTLD spectrum.
Collapse
Affiliation(s)
- Edoardo Gioele Spinelli
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Alma Ghirelli
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Silvia Basaia
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Camilla Cividini
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Nilo Riva
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Elisa Canu
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Veronica Castelnovo
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Teuta Domi
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Magnani
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Francesca Caso
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Paola Caroppo
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Sara Prioni
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Giacomina Rossi
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Lucio Tremolizzo
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Ildebrando Appollonio
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Vincenzo Silani
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Paola Carrera
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Massimo Filippi
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy
| | - Federica Agosta
- From the Neuroimaging Research Unit (E.G.S., A.G., S.B., C.C., E.C., V.C., M.F., F.A.) and Experimental Neuropathology Unit (N.R., T.D.), Division of Neuroscience, Neurorehabilitation Unit (N.R., M.F.), Neurology Unit (E.G.S., G.M., F.C., M.F., F.A.), Laboratory of Clinical Molecular Biology (P. Carrera), and Neurophysiology Service (M.F.), IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University (E.G.S., A.G., C.C., V.C., M.F., F.A.); Unit of Neurology 5-Neuropathology (P. Caroppo, S.P., G.R.), Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan; Neurology Unit (L.T., I.A.), "San Gerardo" Hospital and University of Milano-Bicocca, Monza; Department of Neurology and Laboratory of Neuroscience (V.S.), IRCCS Istituto Auxologico Italiano; and "Dino Ferrari" Center, Department of Pathophysiology and Transplantation (V.S.), Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
12
|
Barceló MA, Povedano M, Vázquez-Costa JF, Franquet Á, Solans M, Saez M. Estimation of the prevalence and incidence of motor neuron diseases in two Spanish regions: Catalonia and Valencia. Sci Rep 2021; 11:6207. [PMID: 33737526 PMCID: PMC7973725 DOI: 10.1038/s41598-021-85395-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 02/23/2021] [Indexed: 11/18/2022] Open
Abstract
According to the degree of upper and lower motor neuron degeneration, motor neuron diseases (MND) can be categorized into amyotrophic lateral sclerosis (ALS), primary lateral sclerosis (PLS) or progressive muscular atrophy (PMA). Although several studies have addressed the prevalence and incidence of ALS, there is a high heterogeneity in their results. Besides this, neither concept has been previously studied in PLS or PMA. Thus, the objective of this study was to estimate the prevalence and incidence of MND, (distinguishing ALS, PLS and PMA), in the Spanish regions of Catalonia and Valencia in the period 2011-2019. Two population-based Spanish cohorts were used, one from Catalonia and the other from Valencia. Given that the samples that comprised both cohorts were not random, i.e., leading to a selection bias, we used a two-part model in which both the individual and contextual observed and unobserved confounding variables are controlled for, along with the spatial and temporal dependence. The prevalence of MND was estimated to be between 3.990 and 6.334 per 100,000 inhabitants (ALS between 3.248 and 5.120; PMA between 0.065 and 0.634; and PLS between 0.046 and 1.896), and the incidence between 1.682 and 2.165 per 100,000 person-years for MND (ALS between 1.351 and 1.754; PMA between 0.225 and 0.628; and PLS between 0.409-0.544). Results were similar in the two regions and did not differ from those previously reported for ALS, suggesting that the proposed method is robust and that neither region presents differential risk or protective factors.
Collapse
Affiliation(s)
- Maria A Barceló
- Research Group On Statistics, Econometrics and Health (GRECS), University of Girona, Carrer de la Universitat de Girona 10, Campus de Montilivi, 17003, Girona, Spain.
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain.
- Unidad Funcional de Motoneurona (UFMN), Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospital Universitario de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain.
| | - Mònica Povedano
- Unidad Funcional de Motoneurona (UFMN), Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospital Universitario de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Juan F Vázquez-Costa
- Unidad Funcional de Motoneurona (UFMN), Instituto de Investigación Sanitaria La Fe (IIS La Fe), Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
- Medicine Department, Facultad de Medicina, University of Valencia, Valencia, Spain
| | - Álvaro Franquet
- Research Group On Statistics, Econometrics and Health (GRECS), University of Girona, Carrer de la Universitat de Girona 10, Campus de Montilivi, 17003, Girona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Fundació Salut Empordà, Figueres, Spain
| | - Marta Solans
- Research Group On Statistics, Econometrics and Health (GRECS), University of Girona, Carrer de la Universitat de Girona 10, Campus de Montilivi, 17003, Girona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Marc Saez
- Research Group On Statistics, Econometrics and Health (GRECS), University of Girona, Carrer de la Universitat de Girona 10, Campus de Montilivi, 17003, Girona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
13
|
Lessons learned from CHMP2B, implications for frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis 2020; 147:105144. [PMID: 33144171 DOI: 10.1016/j.nbd.2020.105144] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) and Amyotrophic Lateral Sclerosis (ALS) are two neurodegenerative diseases with clinical, genetic and pathological overlap. As such, they are commonly regarded as a single spectrum disorder, with pure FTD and pure ALS representing distinct ends of a continuum. Dysfunctional endo-lysosomal and autophagic trafficking, leading to impaired proteostasis is common across the FTD-ALS spectrum. These pathways are, in part, mediated by CHMP2B, a protein that coordinates membrane scission events as a core component of the ESCRT machinery. Here we review how ALS and FTD disease causing mutations in CHMP2B have greatly contributed to our understanding of how endosomal-lysosomal and autophagic dysfunction contribute to neurodegeneration, and how in vitro and in vivo models have helped elucidate novel candidates for potential therapeutic intervention with implications across the FTD-ALS spectrum.
Collapse
|
14
|
West RJH, Ugbode C, Fort-Aznar L, Sweeney ST. Neuroprotective activity of ursodeoxycholic acid in CHMP2B Intron5 models of frontotemporal dementia. Neurobiol Dis 2020; 144:105047. [PMID: 32801000 PMCID: PMC7491204 DOI: 10.1016/j.nbd.2020.105047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/16/2020] [Accepted: 08/08/2020] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) is one of the most prevalent forms of early-onset dementia. It represents part of the FTD-Amyotrophic Lateral Sclerosis (ALS) spectrum, a continuum of genetically and pathologically overlapping disorders. FTD-causing mutations in CHMP2B, a gene encoding a core component of the heteromeric ESCRT-III Complex, lead to perturbed endosomal-lysosomal and autophagic trafficking with impaired proteostasis. While CHMP2B mutations are rare, dysfunctional endosomal-lysosomal signalling is common across the FTD-ALS spectrum. Using our established Drosophila and mammalian models of CHMP2BIntron5 induced FTD we demonstrate that the FDA-approved compound Ursodeoxycholic Acid (UDCA) conveys neuroprotection, downstream of endosomal-lysosomal dysfunction in both Drosophila and primary mammalian neurons. UDCA exhibited a dose dependent rescue of neuronal structure and function in Drosophila pan-neuronally expressing CHMP2BIntron5. Rescue of CHMP2BIntron5 dependent dendritic collapse and apoptosis with UDCA in rat primary neurons was also observed. UDCA failed to ameliorate aberrant accumulation of endosomal and autophagic organelles or ubiquitinated neuronal inclusions in both models. We demonstrate the neuroprotective activity of UDCA downstream of endosomal-lysosomal and autophagic dysfunction, delineating the molecular mode of action of UDCA and highlighting its potential as a therapeutic for the treatment of FTD-ALS spectrum disorders.
Collapse
Affiliation(s)
- Ryan J H West
- Sheffield Institute for Translational Neuroscience, University of Sheffield, S10 2HQ, UK; Neuroscience Institute, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Chris Ugbode
- Department of Biology, University of York, York YO10 5DD, UK
| | | | - Sean T Sweeney
- Department of Biology, University of York, York YO10 5DD, UK.
| |
Collapse
|
15
|
Fort-Aznar L, Ugbode C, Sweeney ST. Retrovirus reactivation in CHMP2BIntron5 models of frontotemporal dementia. Hum Mol Genet 2020; 29:2637-2646. [PMID: 32628265 PMCID: PMC7530534 DOI: 10.1093/hmg/ddaa142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Frontotemporal dementia (FTD) is the second most prevalent form of pre-senile dementia after Alzheimer's disease. Amyotrophic lateral sclerosis (ALS) can overlap genetically, pathologically and clinically with FTD indicating the two conditions are ends of a spectrum and may share common pathological mechanisms. FTD-ALS causing mutations are known to be involved in endosomal trafficking and RNA regulation. Using an unbiased genome-wide genetic screen to identify mutations affecting an FTD-ALS-related phenotype in Drosophila caused by CHMP2BIntron5 expression, we have uncovered repressors of retrovirus (RV) activity as modifiers of CHMP2BIntron5 toxicity. We report that neuronal expression of CHMP2BIntron5 causes an increase in the activity of the endogenous Drosophila RV, gypsy, in the nervous system. Genetically blocking Drosophila gypsy activation and pharmacologically inhibiting viral reverse transcriptase activity prevents degenerative phenotypes observed in fly and rat neurons. These findings directly link endosomal dysfunction to RV de-repression in an FTD-ALS model without TDP-43 pathology. These observations may contribute an understanding to previous discoveries of RV activation in ALS affected patients.
Collapse
Affiliation(s)
- Laura Fort-Aznar
- Department of Biology, University of York, York YO10 5DD, UK
- York Biomedical Research Institute, University of York, York YO10 5DD, UK
| | - Chris Ugbode
- Department of Biology, University of York, York YO10 5DD, UK
- York Biomedical Research Institute, University of York, York YO10 5DD, UK
| | - Sean T Sweeney
- Department of Biology, University of York, York YO10 5DD, UK
- York Biomedical Research Institute, University of York, York YO10 5DD, UK
| |
Collapse
|
16
|
Lu Y, West RJH, Pons M, Sweeney ST, Gao FB. Ik2/TBK1 and Hook/Dynein, an adaptor complex for early endosome transport, are genetic modifiers of FTD-associated mutant CHMP2B toxicity in Drosophila. Sci Rep 2020; 10:14221. [PMID: 32848189 PMCID: PMC7450086 DOI: 10.1038/s41598-020-71097-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mutations in CHMP2B, encoding a protein in the endosomal sorting complexes required for transport (ESCRT) machinery, causes frontotemporal dementia linked to chromosome 3 (FTD3). FTD, the second most common form of pre-senile dementia, can also be caused by genetic mutations in other genes, including TANK-binding kinase 1 (TBK1). How FTD-causing disease genes interact is largely unknown. We found that partial loss function of Ik2, the fly homologue of TBK1 also known as I-kappaB kinase ε (IKKε), enhanced the toxicity of mutant CHMP2B in the fly eye and that Ik2 overexpression suppressed the effect of mutant CHMP2B in neurons. Partial loss of function of Spn-F, a downstream phosphorylation target of Ik2, greatly enhanced the mutant CHMP2B phenotype. An interactome analysis to understand cellular processes regulated by Spn-F identified a network of interacting proteins including Spn-F, Ik2, dynein light chain, and Hook, an adaptor protein in early endosome transport. Partial loss of function of dynein light chain or Hook also enhanced mutant CHMP2B toxicity. These findings identify several evolutionarily conserved genes, including ik2/TBK1, cut up (encoding dynein light chain) and hook, as genetic modifiers of FTD3-associated mutant CHMP2B toxicity and implicate early endosome transport as a potential contributing pathway in FTD.
Collapse
Affiliation(s)
- Yubing Lu
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Department of Pathology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ryan J H West
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Marine Pons
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Sean T Sweeney
- Department of Biology, University of York, York, YO10 5DD, UK
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
17
|
Targeted next-generation sequencing study in familial ALS-FTD Portuguese patients negative for C9orf72 HRE. J Neurol 2020; 267:3578-3592. [DOI: 10.1007/s00415-020-10042-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 12/18/2022]
|
18
|
Aluri KC, Salisbury JP, Prehn JHM, Agar JN. Loss of angiogenin function is related to earlier ALS onset and a paradoxical increase in ALS duration. Sci Rep 2020; 10:3715. [PMID: 32111867 PMCID: PMC7048737 DOI: 10.1038/s41598-020-60431-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 02/12/2020] [Indexed: 12/11/2022] Open
Abstract
0.5-1% of ALS (Amyotrophic Lateral Sclerosis) and Parkinson's disease (PD) are associated with mutations in the angiogenin (ANG). These mutations are thought to cause disease through a loss of ANG function, but this hypothesis has not been evaluated statistically. In addition, the potential for ANG to promote disease has not been considered. With the goal of better defining the etiology of ANG-ALS, we assembled all clinical onset and disease duration data and determined if these were correlated with biochemical properties of ANG variants. Loss of ANG stability and ribonuclease activity were found to correlate with early ALS onset, confirming an aspect of the prevailing model of ANG-ALS. Conversely, loss of ANG stability and ribonuclease activity correlated with longer survival following diagnosis, which is inconsistent with the prevailing model. These results indicate that functional ANG appears to decrease the risk of developing ALS but exacerbate ALS once in progress. These findings are rationalized in terms of studies demonstrating that distinct mechanisms contribute to ALS onset and progression and propose that ANG replacement or stabilization would benefit pre-symptomatic ANG-ALS patients. However, this study challenges the prevailing hypothesis that augmenting ANG will benefit symptomatic ANG-ALS patients. Instead, our results suggest that silencing of ANG activity may be beneficial for symptomatic ALS patients. This study will serve as a call-to-arms for neurologists to consistently publish ALS and PD patient's clinical data-if all ANG-ALS patients' data were available our findings could be tested with considerable statistical power.
Collapse
Affiliation(s)
- Krishna C Aluri
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, USA
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts, 02115, United States
| | - Joseph P Salisbury
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, USA
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts, 02115, United States
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, SFI Future-Neuro Centre, Royal College of Surgeons in Ireland, Dublin, 2, Ireland
| | - Jeffrey N Agar
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, USA.
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts, 02115, United States.
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts, 02115, United States.
| |
Collapse
|
19
|
Waegaert R, Dirrig-Grosch S, Parisot F, Keime C, Henriques A, Loeffler JP, René F. Longitudinal transcriptomic analysis of altered pathways in a CHMP2B intron5-based model of ALS-FTD. Neurobiol Dis 2019; 136:104710. [PMID: 31837425 DOI: 10.1016/j.nbd.2019.104710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 10/28/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia are two neurodegenerative diseases with currently no cure. These two diseases share a clinical continuum with overlapping genetic causes. Mutations in the CHMP2B gene are found in patients with ALS, FTD and ALS-FTD. To highlight deregulated mechanisms occurring in ALS-FTD linked to the CHMP2B gene, we performed a whole transcriptomic study on lumbar spinal cord from CHMP2Bintron5 mice, a model that develops progressive motor alterations associated with dementia symptoms reminiscent of both ALS and FTD. To gain insight into the transcriptomic changes taking place during disease progression this study was performed at three stages: asymptomatic, symptomatic and end stage. We showed that before appearance of motor symptoms, the major disrupted mechanisms were linked with the immune system/inflammatory response and lipid metabolism. These processes were progressively replaced by alterations of neuronal electric activity as motor symptoms appeared, alterations that could lead to motor neuron dysfunction. To investigate overlapping alterations in gene expression between two ALS-causing genes, we then compared the transcriptome of symptomatic CHMP2Bintron5 mice with the one of symptomatic SOD1G86R mice and found the same families deregulated providing further insights into common underlying dysfunction of biological pathways, disrupted or disturbed in ALS. Altogether, this study provides a database to explore potential new candidate genes involved in the CHMP2Bintron5-based pathogenesis of ALS, and provides molecular clues to further understand the functional consequences that diseased neurons expressing CHMP2B mutant may have on their neighbor cells.
Collapse
Affiliation(s)
- Robin Waegaert
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Sylvie Dirrig-Grosch
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Florian Parisot
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258, CNRS, UMR7104, Université de Strasbourg, 1 Rue Laurent Fries, 67400 Illkirch-Graffenstaden, France
| | - Alexandre Henriques
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Jean-Philippe Loeffler
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France
| | - Frédérique René
- INSERM U1118 Mécanismes centraux et périphériques de la neurodégénérescence, Université de Strasbourg, 11 rue Humann, Strasbourg, France.
| |
Collapse
|
20
|
Tripolszki K, Danis J, Padhi AK, Gomes J, Bozó R, Nagy ZF, Nagy D, Klivényi P, Engelhardt JI, Széll M. Angiogenin mutations in Hungarian patients with amyotrophic lateral sclerosis: Clinical, genetic, computational, and functional analyses. Brain Behav 2019; 9:e01293. [PMID: 31025543 PMCID: PMC6576160 DOI: 10.1002/brb3.1293] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Mutations in the angiogenin (ANG) gene are known to be associated with both familial and sporadic amyotrophic lateral sclerosis (ALS). The majority of disease-causing mutations of ANG result in loss of either ribonucleolytic activity, nuclear translocation activity or both. METHODS We sequenced ANG gene from a total of 136 sporadic ALS patients and 112 healthy controls of Hungarian origin. To elucidate the role of the R33W mutation in the disease mechanism, computational, and functional analyses were performed. RESULTS Mutation screening revealed a mutation located in the signal peptide (M-24I) and two mutations that affect the mature protein (R33W, V103I). The R33W mutation, which has not been previously detected in ALS patients, affects the key amino acid of the nuclear translocation signal of the ANG protein. Molecular dynamics simulations suggested that the R33W mutation results in partial loss of ribonucleolytic activity and reduced nuclear translocation activity. The ribonucleolytic assay and nuclear translocation assay of the R33W ANG protein confirmed the molecular dynamics results. CONCLUSIONS In the Hungarian ALS population, the observed frequency of ANG mutations was 2.9%, which is higher than previously reported for sporadic cohorts. The evidence from computational and functional analyses support the deleterious effect of the novel R33W variant detected in this study.
Collapse
Affiliation(s)
| | - Judit Danis
- MTA-SZTE Dermatological Research Group, Szeged, Hungary
| | - Aditya K Padhi
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, India
| | - James Gomes
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, India
| | - Renáta Bozó
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Zsófia F Nagy
- Department of Medical Genetics, University of Szeged, Szeged, Hungary
| | - Dóra Nagy
- Department of Medical Genetics, University of Szeged, Szeged, Hungary
| | - Péter Klivényi
- Department of Neurology, University of Szeged, Szeged, Hungary
| | | | - Márta Széll
- Department of Medical Genetics, University of Szeged, Szeged, Hungary.,MTA-SZTE Dermatological Research Group, Szeged, Hungary
| |
Collapse
|
21
|
de Vries BS, Rustemeijer LMM, Bakker LA, Schröder CD, Veldink JH, van den Berg LH, Nijboer TCW, van Es MA. Cognitive and behavioural changes in PLS and PMA:challenging the concept of restricted phenotypes. J Neurol Neurosurg Psychiatry 2019; 90:141-147. [PMID: 30076267 DOI: 10.1136/jnnp-2018-318788] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Cognitive and behavioural changes within the spectrum of frontotemporal dementia (FTD) are observed frequently in patients with amyotrophic lateral sclerosis (ALS). Whether these changes also occur in other forms of motor neuron disease (MND) is not well studied. We therefore systemically screened a large cohort of patients with primary lateral sclerosis (PLS) and progressive muscular atrophy (PMA) for cognitive and behavioural changes, and subsequently compared our findings with a cohort of patients with ALS. METHODS Using a set of screening instruments (Edinburgh Cognitive and Behavioural ALS Screen, ALS and Frontotemporal Dementia Questionnaire, Frontal Assessment Battery, and Hospital Anxiety and Depression Scale), the presence of cognitive and behavioural changes as well as anxiety and depression in 277 patients with ALS, 75 patients with PLS and 143 patients with PMA was evaluated retrospectively. RESULTS We found a high frequency of cognitive and behavioural abnormalities with similar profiles in all three groups. Subjects with behavioural variant FTD were identified in all groups. CONCLUSIONS The percentage of patients with PLS and PMA with cognitive dysfunction was similar to patients with ALS, emphasising the importance for cognitive screening as part of routine clinical care in all three patient groups. With a similar cognitive profile, in line with genetic and clinical overlap between the MNDs, the view of PLS as an MND exclusively affecting upper motor neurons and PMA exclusively affecting lower motor neurons cannot be held. Therefore, our findings are in contrast to the recently revised El Escorial criteria of 2015, where PLS and PMA are described as restricted phenotypes. Our study favours a view of PLS and PMA as multidomain diseases similar to ALS.
Collapse
Affiliation(s)
- Bálint S de Vries
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Laura M M Rustemeijer
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leonhard A Bakker
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Center of Excellence for Rehabilitation Medicine, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University and De Hoogstraat Rehabilitation, Utrecht, The Netherlands
| | - Carin D Schröder
- Center of Excellence for Rehabilitation Medicine, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University and De Hoogstraat Rehabilitation, Utrecht, The Netherlands.,Ecare4you, Amersfoort, The Netherlands
| | - Jan H Veldink
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tanja C W Nijboer
- Center of Excellence for Rehabilitation Medicine, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University and De Hoogstraat Rehabilitation, Utrecht, The Netherlands.,Department of Experimental Psychology, Helmholtz Institute, Utrecht University, Utrecht, The Netherlands
| | - Michael A van Es
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
22
|
Iyer S, Subramanian V, Acharya KR. C9orf72, a protein associated with amyotrophic lateral sclerosis (ALS) is a guanine nucleotide exchange factor. PeerJ 2018; 6:e5815. [PMID: 30356970 PMCID: PMC6195791 DOI: 10.7717/peerj.5815] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/22/2018] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two late onset neurodegenerative diseases, have been shown to share overlapping cellular pathologies and genetic origins. Studies suggest that a hexanucleotide repeat expansion in the first intron of the C9orf72 gene is the most common cause of familial FTD and ALS pathology. The C9orf72 protein is predicted to be a differentially expressed in normal and neoplastic cells domain protein implying that C9orf72 functions as a guanine nucleotide exchange factor (GEF) to regulate specific Rab GTPases. Reported studies thus far point to a putative role for C9orf72 in lysosome biogenesis, vesicular trafficking, autophagy and mechanistic target of rapamycin complex1 (mTORC1) signaling. Here we report the expression, purification and biochemical characterization of C9orf72 protein. We conclusively show that C9orf72 is a GEF. The distinctive presence of both Rab- and Rho-GTPase GEF activities suggests that C9orf72 may function as a dual exchange factor coupling physiological functions such as cytoskeleton modulation and autophagy with endocytosis.
Collapse
Affiliation(s)
- Shalini Iyer
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| | | | - K Ravi Acharya
- Department of Biology and Biochemistry, University of Bath, Bath, UK
| |
Collapse
|
23
|
Weil R, Laplantine E, Curic S, Génin P. Role of Optineurin in the Mitochondrial Dysfunction: Potential Implications in Neurodegenerative Diseases and Cancer. Front Immunol 2018; 9:1243. [PMID: 29971063 PMCID: PMC6018216 DOI: 10.3389/fimmu.2018.01243] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/17/2018] [Indexed: 12/11/2022] Open
Abstract
Optineurin (Optn) is a 577 aa protein encoded by the Optn gene. Mutations of Optn are associated with normal tension glaucoma and amyotrophic lateral sclerosis, and its gene has also been linked to the development of Paget’s disease of bone and Crohn’s disease. Optn is involved in diverse cellular functions, including NF-κB regulation, membrane trafficking, exocytosis, vesicle transport, reorganization of actin and microtubules, cell cycle control, and autophagy. Besides its role in xenophagy and autophagy of aggregates, Optn has been identified as a primary autophagy receptor, among the five adaptors that translocate to mitochondria during mitophagy. Mitophagy is a selective macroautophagy process during which irreparable mitochondria are degraded, preventing accumulation of defective mitochondria and limiting the release of reactive oxygen species and proapoptotic factors. Mitochondrial quality control via mitophagy is central to the health of cells. One of the important surveillance pathways of mitochondrial health is the recently defined signal transduction pathway involving the mitochondrial PTEN-induced putative kinase 1 (PINK1) protein and the cytosolic RING-between-RING ubiquitin ligase Parkin. Both of these proteins, when mutated, have been identified in certain forms of Parkinson’s disease. By targeting ubiquitinated mitochondria to autophagosomes through its association with autophagy related proteins, Optn is responsible for a critical step in mitophagy. This review reports recent discoveries on the role of Optn in mitophagy and provides insight into its link with neurodegenerative diseases. We will also discuss the involvement of Optn in other pathologies in which mitophagy dysfunctions are involved including cancer.
Collapse
Affiliation(s)
- Robert Weil
- Laboratory of Signaling and Pathogenesis, Institut Pasteur, CNRS UMR3691, Paris, France
| | - Emmanuel Laplantine
- Laboratory of Signaling and Pathogenesis, Institut Pasteur, CNRS UMR3691, Paris, France
| | - Shannel Curic
- Laboratory of Signaling and Pathogenesis, Institut Pasteur, CNRS UMR3691, Paris, France
| | - Pierre Génin
- Laboratory of Signaling and Pathogenesis, Institut Pasteur, CNRS UMR3691, Paris, France
| |
Collapse
|
24
|
Ryan TA, Tumbarello DA. Optineurin: A Coordinator of Membrane-Associated Cargo Trafficking and Autophagy. Front Immunol 2018; 9:1024. [PMID: 29867991 PMCID: PMC5962687 DOI: 10.3389/fimmu.2018.01024] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/24/2018] [Indexed: 12/13/2022] Open
Abstract
Optineurin is a multifunctional adaptor protein intimately involved in various vesicular trafficking pathways. Through interactions with an array of proteins, such as myosin VI, huntingtin, Rab8, and Tank-binding kinase 1, as well as via its oligomerisation, optineurin has the ability to act as an adaptor, scaffold, or signal regulator to coordinate many cellular processes associated with the trafficking of membrane-delivered cargo. Due to its diverse interactions and its distinct functions, optineurin is an essential component in a number of homeostatic pathways, such as protein trafficking and organelle maintenance. Through the binding of polyubiquitinated cargoes via its ubiquitin-binding domain, optineurin also serves as a selective autophagic receptor for the removal of a wide range of substrates. Alternatively, it can act in an ubiquitin-independent manner to mediate the clearance of protein aggregates. Regarding its disease associations, mutations in the optineurin gene are associated with glaucoma and have more recently been found to correlate with Paget’s disease of bone and amyotrophic lateral sclerosis (ALS). Indeed, ALS-associated mutations in optineurin result in defects in neuronal vesicular localisation, autophagosome–lysosome fusion, and secretory pathway function. More recent molecular and functional analysis has shown that it also plays a role in mitophagy, thus linking it to a number of other neurodegenerative conditions, such as Parkinson’s. Here, we review the role of optineurin in intracellular membrane trafficking, with a focus on autophagy, and describe how upstream signalling cascades are critical to its regulation. Current data and contradicting reports would suggest that optineurin is an important and selective autophagy receptor under specific conditions, whereby interplay, synergy, and functional redundancy with other receptors occurs. We will also discuss how dysfunction in optineurin-mediated pathways may lead to perturbation of critical cellular processes, which can drive the pathologies of number of diseases. Therefore, further understanding of optineurin function, its target specificity, and its mechanism of action will be critical in fully delineating its role in human disease.
Collapse
Affiliation(s)
- Thomas A Ryan
- Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - David A Tumbarello
- Biological Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
25
|
Jutzi D, Akinyi MV, Mechtersheimer J, Frilander MJ, Ruepp MD. The emerging role of minor intron splicing in neurological disorders. Cell Stress 2018; 2:40-54. [PMID: 31225466 PMCID: PMC6558932 DOI: 10.15698/cst2018.03.126] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pre-mRNA splicing is an essential step in eukaryotic gene expression. Mutations in cis-acting sequence elements within pre-mRNA molecules or trans-acting factors involved in pre-mRNA processing have both been linked to splicing dysfunction that give rise to a large number of human diseases. These mutations typically affect the major splicing pathway, which excises more than 99% of all introns in humans. However, approximately 700-800 human introns feature divergent intron consensus sequences at their 5' and 3' ends and are recognized by a separate pre-mRNA processing machinery denoted as the minor spliceosome. This spliceosome has been studied less than its major counterpart, but has received increasing attention during the last few years as a novel pathomechanistic player on the stage in neurodevelopmental and neurodegenerative diseases. Here, we review the current knowledge on minor spliceosome function and discuss its potential pathomechanistic role and impact in neurodegeneration.
Collapse
Affiliation(s)
- Daniel Jutzi
- Department of Chemistry and Biochemistry, University of Bern, CH-3012 Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Maureen V Akinyi
- Institute of Biotechnology, University of Helsinki, FI-00014, Finland
| | - Jonas Mechtersheimer
- Department of Chemistry and Biochemistry, University of Bern, CH-3012 Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Mikko J Frilander
- Institute of Biotechnology, University of Helsinki, FI-00014, Finland
| | - Marc-David Ruepp
- Department of Chemistry and Biochemistry, University of Bern, CH-3012 Bern, Switzerland.,United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 9NU London, UK
| |
Collapse
|
26
|
Starr A, Sattler R. Synaptic dysfunction and altered excitability in C9ORF72 ALS/FTD. Brain Res 2018; 1693:98-108. [PMID: 29453960 DOI: 10.1016/j.brainres.2018.02.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/06/2018] [Accepted: 02/10/2018] [Indexed: 02/08/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by a progressive degeneration of upper and lower motor neurons, resulting in fatal paralysis due to denervation of the muscle. Due to genetic, pathological and symptomatic overlap, ALS is now considered a spectrum disease together with frontotemporal dementia (FTD), the second most common cause of dementia in individuals under the age of 65. Interestingly, in both diseases, there is a large prevalence of RNA binding proteins (RBPs) that are mutated and considered disease-causing, or whose dysfunction contribute to disease pathogenesis. The most common shared genetic mutation in ALS/FTD is a hexanucleuotide repeat expansion within intron 1 of C9ORF72 (C9). Three potentially overlapping, putative toxic mechanisms have been proposed: loss of function due to haploinsufficient expression of the C9ORF72 mRNA, gain of function of the repeat RNA aggregates, or RNA foci, and repeat-associated non-ATG-initiated translation (RAN) of the repeat RNA into toxic dipeptide repeats (DPRs). Regardless of the causative mechanism, disease symptoms are ultimately caused by a failure of neurotransmission in three regions: the brain, the spinal cord, and the neuromuscular junction. Here, we review C9 ALS/FTD-associated synaptic dysfunction and aberrant neuronal excitability in these three key regions, focusing on changes in morphology and synapse formation, excitability, and excitotoxicity in patients, animal models, and in vitro models. We compare these deficits to those seen in other forms of ALS and FTD in search of shared pathways, and discuss the potential targeting of synaptic dysfunctions for therapeutic intervention in ALS and FTD patients.
Collapse
Affiliation(s)
- Alexander Starr
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, United States
| | - Rita Sattler
- Division of Neurobiology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013, United States.
| |
Collapse
|
27
|
van Es MA, Hardiman O, Chio A, Al-Chalabi A, Pasterkamp RJ, Veldink JH, van den Berg LH. Amyotrophic lateral sclerosis. Lancet 2017; 390:2084-2098. [PMID: 28552366 DOI: 10.1016/s0140-6736(17)31287-4] [Citation(s) in RCA: 809] [Impact Index Per Article: 115.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 03/13/2017] [Accepted: 03/20/2017] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis is characterised by the progressive loss of motor neurons in the brain and spinal cord. This neurodegenerative syndrome shares pathobiological features with frontotemporal dementia and, indeed, many patients show features of both diseases. Many different genes and pathophysiological processes contribute to the disease, and it will be necessary to understand this heterogeneity to find effective treatments. In this Seminar, we discuss clinical and diagnostic approaches as well as scientific advances in the research fields of genetics, disease modelling, biomarkers, and therapeutic strategies.
Collapse
Affiliation(s)
- Michael A van Es
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, the Netherlands
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland; Department of Neurology, Beaumont Hospital, Beaumont, Ireland
| | - Adriano Chio
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy; Azienda Ospedaliero Universitaria Citta della Salute e della Scienza di Torino, Turin, Italy; Neuroscience Institute of Turin, Turin, Italy
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK; NIHR Dementia Biomedical Research Unit, King's College London, London, UK
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, the Netherlands
| | - Jan H Veldink
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, the Netherlands
| | - Leonard H van den Berg
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, the Netherlands.
| |
Collapse
|
28
|
Sadoul R, Laporte MH, Chassefeyre R, Chi KI, Goldberg Y, Chatellard C, Hemming FJ, Fraboulet S. The role of ESCRT during development and functioning of the nervous system. Semin Cell Dev Biol 2017; 74:40-49. [PMID: 28811263 DOI: 10.1016/j.semcdb.2017.08.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/21/2017] [Accepted: 08/04/2017] [Indexed: 12/12/2022]
Abstract
The endosomal sorting complex required for transport (ESCRT) is made of subcomplexes (ESCRT 0-III), crucial to membrane remodelling at endosomes, nuclear envelope and cell surface. ESCRT-III shapes membranes and in most cases cooperates with the ATPase VPS4 to mediate fission of membrane necks from the inside. The first ESCRT complexes mainly serve to catalyse the formation of ESCRT-III but can be bypassed by accessory proteins like the Alg-2 interacting protein-X (ALIX). In the nervous system, ALIX/ESCRT controls the survival of embryonic neural progenitors and later on the outgrowth and pruning of axons and dendrites, all necessary steps to establish a functional brain. In the adult brain, ESCRTs allow the endosomal turn over of synaptic vesicle proteins while stable ESCRT complexes might serve as scaffolds for the postsynaptic parts. The necessity of ESCRT for the harmonious function of the brain has its pathological counterpart, the mutations in CHMP2B of ESCRT-III giving rise to several neurodegenerative diseases.
Collapse
Affiliation(s)
- Rémy Sadoul
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France.
| | - Marine H Laporte
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Romain Chassefeyre
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Kwang Il Chi
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Yves Goldberg
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Christine Chatellard
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Fiona J Hemming
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| | - Sandrine Fraboulet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1216, F-38042 Grenoble, France; Université Grenoble Alpes, Institut des Neurosciences, F-38042 Grenoble, France
| |
Collapse
|
29
|
Tard C, Defebvre L, Moreau C, Devos D, Danel-Brunaud V. Clinical features of amyotrophic lateral sclerosis and their prognostic value. Rev Neurol (Paris) 2017; 173:263-272. [DOI: 10.1016/j.neurol.2017.03.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 03/27/2017] [Indexed: 12/29/2022]
|
30
|
Comparison of the clinical and cognitive features of genetically positive ALS patients from the largest tertiary center in Serbia. J Neurol 2017; 264:1091-1098. [DOI: 10.1007/s00415-017-8495-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 11/26/2022]
|
31
|
Sheehan P, Waites CL. Coordination of synaptic vesicle trafficking and turnover by the Rab35 signaling network. Small GTPases 2017; 10:54-63. [PMID: 28129039 PMCID: PMC6343537 DOI: 10.1080/21541248.2016.1270392] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rab35 and the Rab35 network of GAPs, GEFs, and effectors are important regulators of membrane trafficking for a variety of cellular processes, from cytokinesis and phagocytosis to neurite outgrowth. In the past five years, components of this signaling network have also been implicated as critical mediators of synaptic vesicle (SV) recycling and protein homeostasis. Recent studies by several groups, including our own, have demonstrated that Rab35-mediated endosomal sorting is required for the degradation of SV proteins via the ESCRT pathway, thereby eliminating old or damaged proteins from the SV pool. This sorting process is regulated by Rab35 activation in response to neuronal activity, and potentially by an antagonistic signaling relationship between Rab35 and the small GTPase Arf6 that directs SVs into distinct recycling pathways depending on neuronal activity levels. Furthermore, mutations in genes encoding Rab35 regulatory proteins are emerging as causative factors in human neurologic and neurodegenerative diseases, consistent with their important roles in synaptic and neuronal health. Here, we review these recent findings and offer our perspective on how the Rab35 signaling network functions to maintain neurotransmission and synaptic fitness.
Collapse
Affiliation(s)
- Patricia Sheehan
- a Department of Pathology and Cell Biology , Columbia University Medical Center , New York , NY , USA
| | - Clarissa L Waites
- a Department of Pathology and Cell Biology , Columbia University Medical Center , New York , NY , USA.,b Department of Neuroscience , Columbia University Medical Center , New York , NY , USA
| |
Collapse
|
32
|
Saberi S, Stauffer JE, Schulte DJ, Ravits J. Neuropathology of Amyotrophic Lateral Sclerosis and Its Variants. Neurol Clin 2016; 33:855-76. [PMID: 26515626 DOI: 10.1016/j.ncl.2015.07.012] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The neuropathologic molecular signature common to almost all sporadic amyotrophic lateral sclerosis (ALS) and most familial ALS is TDP-43 immunoreactive neuronal cytoplasmic inclusions. The neuropathologic and molecular neuropathologic features of ALS variants, primarily lateral sclerosis and progressive muscular atrophy, are less certain but also seem to share the primary features of ALS. Genetic causes, including mutations in SOD1, TDP-43, FUS, and C9orf72, all have distinctive molecular neuropathologic signatures. Neuropathology will continue to play an increasingly key role in solving the puzzle of ALS pathogenesis.
Collapse
Affiliation(s)
- Shahram Saberi
- Department of Neurosciences, ALS Translational Research, University of California (San Diego), 9500 Gilman Drive, MC0624, La Jolla, CA 92093, USA
| | - Jennifer E Stauffer
- Department of Neurosciences, ALS Translational Research, University of California (San Diego), 9500 Gilman Drive, MC0624, La Jolla, CA 92093, USA
| | - Derek J Schulte
- Department of Neurosciences, ALS Translational Research, University of California (San Diego), 9500 Gilman Drive, MC0624, La Jolla, CA 92093, USA
| | - John Ravits
- Department of Neurosciences, ALS Translational Research, University of California (San Diego), 9500 Gilman Drive, MC0624, La Jolla, CA 92093, USA.
| |
Collapse
|
33
|
Abstract
Progressive muscular atrophy (PMA) is a rare, sporadic, adult-onset motor neuron disease, clinically characterized by isolated lower motor neuron features; however, clinically evident upper motor neuron signs may emerge in some patients. Subclinical upper motor neuron involvement is identified pathologically, radiologically, and neurophysiologically in a substantial number of patients with PMA. Patients with subclinical upper motor neuron involvement do not fulfill the revised El Escorial criteria to participate in amyotrophic lateral sclerosis clinical trials. Intravenous immunoglobulin therapy is only marginally beneficial in a small subgroup of patients with lower motor neuron syndrome without conduction block.
Collapse
Affiliation(s)
- Teerin Liewluck
- Department of Neurology, University of Colorado School of Medicine, Anschutz Medical Campus, 12631 East 17th Avenue, Mail Stop B-185, Aurora, CO 80045, USA; Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | - David S Saperstein
- Phoenix Neurological Associates, University of Arizona College of Medicine, 5090 North 40th Street, Suite 250, Phoenix, AZ 85018, USA
| |
Collapse
|
34
|
Vernay A, Therreau L, Blot B, Risson V, Dirrig-Grosch S, Waegaert R, Lequeu T, Sellal F, Schaeffer L, Sadoul R, Loeffler JP, René F. A transgenic mouse expressing CHMP2Bintron5 mutant in neurons develops histological and behavioural features of amyotrophic lateral sclerosis and frontotemporal dementia. Hum Mol Genet 2016; 25:3341-3360. [PMID: 27329763 DOI: 10.1093/hmg/ddw182] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022] Open
Abstract
Mutations in the charged multivesicular body protein 2B (CHMP2B) are associated with frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS), and with a mixed ALS-FTD syndrome. To model this syndrome, we generated a transgenic mouse line expressing the human CHMP2Bintron5 mutant in a neuron-specific manner. These mice developed a dose-dependent disease phenotype. A longitudinal study revealed progressive gait abnormalities, reduced muscle strength and decreased motor coordination. CHMP2Bintron5 mice died due to generalized paralysis. When paralyzed, signs of denervation were present as attested by altered electromyographic profiles, by decreased number of fully innervated neuromuscular junctions, by reduction in size of motor endplates and by a decrease of sciatic nerve axons area. However, spinal motor neurons cell bodies were preserved until death. In addition to the motor dysfunctions, CHMP2Bintron5 mice progressively developed FTD-relevant behavioural modifications such as disinhibition, stereotypies, decrease in social interactions, compulsivity and change in dietary preferences. Furthermore, neurons in the affected spinal cord and brain regions showed accumulation of p62-positive cytoplasmic inclusions associated or not with ubiquitin and CHMP2Bintron5 As observed in FTD3 patients, these inclusions were negative for TDP-43 and FUS. Moreover, astrogliosis and microgliosis developed with age. Altogether, these data indicate that the neuronal expression of human CHMP2Bintron5 in areas involved in motor and cognitive functions induces progressive motor alterations associated with dementia symptoms and with histopathological hallmarks reminiscent of both ALS and FTD.
Collapse
Affiliation(s)
- Aurélia Vernay
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Ludivine Therreau
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Béatrice Blot
- INSERM U836, Grenoble Institut des Neurosciences, Université Joseph Fourier, F-38700 La Tronche, France
| | - Valérie Risson
- Laboratoire de Biologie Moléculaire de la Cellule, UMR5239 CNRS/ENS Lyon/UCBL/HCL Ecole normale supérieure de Lyon, F-69364 Lyon Cedex 07, France
| | - Sylvie Dirrig-Grosch
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Robin Waegaert
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Thiebault Lequeu
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - François Sellal
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Neurology department, Hôpitaux civils and CMRR, F-68000 Colmar, France
| | - Laurent Schaeffer
- Laboratoire de Biologie Moléculaire de la Cellule, UMR5239 CNRS/ENS Lyon/UCBL/HCL Ecole normale supérieure de Lyon, F-69364 Lyon Cedex 07, France
| | - Rémy Sadoul
- INSERM U836, Grenoble Institut des Neurosciences, Université Joseph Fourier, F-38700 La Tronche, France
| | - Jean-Philippe Loeffler
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France.,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| | - Frédérique René
- INSERM, U1118, Mécanismes Centraux et Périphériques de la Neurodégénérescence, F-67000 Strasbourg, France .,Université de Strasbourg, UMRS1118, Faculté de Médecine, Fédération de Médecine Translationelle de Strasbourg, F-67000 Strasbourg, France
| |
Collapse
|
35
|
Variants within the SP110 nuclear body protein modify risk of canine degenerative myelopathy. Proc Natl Acad Sci U S A 2016; 113:E3091-100. [PMID: 27185954 DOI: 10.1073/pnas.1600084113] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Canine degenerative myelopathy (DM) is a naturally occurring neurodegenerative disease with similarities to some forms of amyotrophic lateral sclerosis (ALS). Most dogs that develop DM are homozygous for a common superoxide dismutase 1 gene (SOD1) mutation. However, not all dogs homozygous for this mutation develop disease. We performed a genome-wide association analysis in the Pembroke Welsh Corgi (PWC) breed comparing DM-affected and -unaffected dogs homozygous for the SOD1 mutation. The analysis revealed a modifier locus on canine chromosome 25. A haplotype within the SP110 nuclear body protein (SP110) was present in 40% of affected compared with 4% of unaffected dogs (P = 1.5 × 10(-5)), and was associated with increased probability of developing DM (P = 4.8 × 10(-6)) and earlier onset of disease (P = 1.7 × 10(-5)). SP110 is a nuclear body protein involved in the regulation of gene transcription. Our findings suggest that variations in SP110-mediated gene transcription may underlie, at least in part, the variability in risk for developing DM among PWCs that are homozygous for the disease-related SOD1 mutation. Further studies are warranted to clarify the effect of this modifier across dog breeds.
Collapse
|
36
|
Tortelli R, Copetti M, Panza F, Fontana A, Cortese R, Capozzo R, Introna A, D'Errico E, Zoccolella S, Arcuti S, Seripa D, Simone IL, Logroscino G. Time to generalization and prediction of survival in patients with amyotrophic lateral sclerosis: a retrospective observational study. Eur J Neurol 2016; 23:1117-25. [PMID: 27016147 DOI: 10.1111/ene.12994] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/02/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND PURPOSE A strong association between time to generalization (TTG), considered as the time of spreading of the clinical signs from spinal or bulbar localization to both, and survival was recently identified in patients with amyotrophic lateral sclerosis (ALS). Thus, TTG may be used as an early to intermediate end-point in survival studies. The aim of the present study was to test TTG as a predictor of survival in ALS. METHODS This was an observational retrospective study of ALS patients from a tertiary referral centre over a 5-year follow-up period. RESULTS In 212 ALS patients, TTG was associated with time to death/tracheostomy [R 0.62, 95% confidence interval (CI) 0.53-0.70; P < 0.001]. In a time-to-event analysis, longer TTG resulted in lower risk to reach a composite outcome (death or tracheostomy) both in univariate [hazard ratio (HR) 0.98, 95% CI 0.97-0.99] and multivariate Cox analyses (HR 0.98, 95% CI 0.96-0.99). TTG predicted death/tracheostomy at 4 years (C-statistic 0.58; 95% CI 0.53-0.63) and at 5 years (C-statistic 0.58; 95% CI 0.53-0.62). CONCLUSIONS Based on the present results from a large clinical cohort, TTG may be used as a new early to intermediate end-point to describe the ALS natural history. TTG may be potentially useful as a new primary outcome measure for clinical trials.
Collapse
Affiliation(s)
- R Tortelli
- Unit of Neurodegenerative Diseases, Department of Clinical Research in Neurology, University of Bari 'Aldo Moro', Tricase, Lecce, Italy
| | - M Copetti
- Unit of Biostatistics, IRCCS 'Casa Sollievo della Sofferenza', San Giovanni Rotondo, Foggia, Italy
| | - F Panza
- Unit of Neurodegenerative Diseases, Department of Clinical Research in Neurology, University of Bari 'Aldo Moro', Tricase, Lecce, Italy.,Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy.,Geriatric Unit and Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS 'Casa Sollievo della Sofferenza', San Giovanni Rotondo, Foggia, Italy
| | - A Fontana
- Unit of Biostatistics, IRCCS 'Casa Sollievo della Sofferenza', San Giovanni Rotondo, Foggia, Italy
| | - R Cortese
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - R Capozzo
- Unit of Neurodegenerative Diseases, Department of Clinical Research in Neurology, University of Bari 'Aldo Moro', Tricase, Lecce, Italy
| | - A Introna
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - E D'Errico
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - S Zoccolella
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - S Arcuti
- Unit of Neurodegenerative Diseases, Department of Clinical Research in Neurology, University of Bari 'Aldo Moro', Tricase, Lecce, Italy
| | - D Seripa
- Geriatric Unit and Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS 'Casa Sollievo della Sofferenza', San Giovanni Rotondo, Foggia, Italy
| | - I L Simone
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| | - G Logroscino
- Unit of Neurodegenerative Diseases, Department of Clinical Research in Neurology, University of Bari 'Aldo Moro', Tricase, Lecce, Italy.,Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari 'Aldo Moro', Bari, Italy
| |
Collapse
|
37
|
Li HF, Wu ZY. Genotype-phenotype correlations of amyotrophic lateral sclerosis. Transl Neurodegener 2016; 5:3. [PMID: 26843957 PMCID: PMC4738789 DOI: 10.1186/s40035-016-0050-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 01/27/2016] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by progressive neuronal loss and degeneration of upper motor neuron (UMN) and lower motor neuron (LMN). The clinical presentations of ALS are heterogeneous and there is no single test or procedure to establish the diagnosis of ALS. Most cases are diagnosed based on symptoms, physical signs, progression, EMG, and tests to exclude the overlapping conditions. Familial ALS represents about 5 ~ 10 % of ALS cases, whereas the vast majority of patients are sporadic. To date, more than 20 causative genes have been identified in hereditary ALS. Detecting the pathogenic mutations or risk variants for each ALS individual is challenging. However, ALS patients carrying some specific mutations or variant may exhibit subtly distinct clinical features. Unraveling the respective genotype-phenotype correlation has important implications for the genetic explanations. In this review, we will delineate the clinical features of ALS, outline the major ALS-related genes, and summarize the possible genotype-phenotype correlations of ALS.
Collapse
Affiliation(s)
- Hong-Fu Li
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009 China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology in Second Affiliated Hospital, and the Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009 China
| |
Collapse
|
38
|
Large-scale screening in sporadic amyotrophic lateral sclerosis identifies genetic modifiers in C9orf72 repeat carriers. Neurobiol Aging 2015; 39:220.e9-15. [PMID: 26777436 DOI: 10.1016/j.neurobiolaging.2015.12.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/18/2015] [Accepted: 12/20/2015] [Indexed: 12/11/2022]
Abstract
Sporadic amyotrophic lateral sclerosis (ALS) is considered to be a complex disease with multiple genetic risk factors contributing to the pathogenesis. Identification of genetic risk factors that co-occur frequently could provide relevant insight into underlying mechanisms of motor neuron degeneration. To dissect the genetic architecture of sporadic ALS, we undertook a large sequencing study in 755 apparently sporadic ALS cases and 959 controls, analyzing 10 ALS genes: SOD1, C9orf72, TARDBP, FUS, ANG, CHMP2B, ATXN2, NIPA1, SMN1, and UNC13A. We observed sporadic cases with multiple genetic risk variants in 4.1% compared with 1.3% in controls. The overall difference was not in excess of what is to be expected by chance (binomial test, p = 0.59). We did, however, observe a higher frequency than expected of C9orf72 repeat carriers with co-occurring susceptibility variants (ATXN2, NIPA1, and SMN1; p = 0.001), which is mainly because of the co-occurrence of NIPA1 repeats in 15% of C9orf72 repeat carriers (p = 0.006).
Collapse
|
39
|
Abstract
Around 10-15% of patients diagnosed with frontotemporal dementia (FTD) have a
positive family history for FTD with an autosomal dominant pattern of
inheritance. Since the identification of mutations in MAPT
(microtubule-associated protein tau gene) in 1998, over 10 other genes have been
associated with FTD spectrum disorders, discussed in this review. Along with
MAPT, mutations in GRN (progranulin) and
C9orf72 (chromosome 9 open reading frame 72) are the most
commonly identified in FTD cohorts. The association of FTD and motor neuron
disease (MND) can be caused by mutations in C9orf72 and other
genes, such as TARDBP (TAR DNA-binding protein),
FUS (fused in sarcoma), UBQLN2 (ubiquilin
2). Multisystem proteinopathy is a complex phenotype that includes FTD, Paget
disease of the bone, inclusion body myopathy and MND, and can be due to
mutations in VCP (valosing containing protein) and other
recently identified genes.
Collapse
Affiliation(s)
- Leonel T Takada
- MD, PhD, Cognitive and Behavioral Neurology Unit, Department of Neurology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Brazil
| |
Collapse
|
40
|
West RJH, Lu Y, Marie B, Gao FB, Sweeney ST. Rab8, POSH, and TAK1 regulate synaptic growth in a Drosophila model of frontotemporal dementia. ACTA ACUST UNITED AC 2015; 208:931-47. [PMID: 25800055 PMCID: PMC4384727 DOI: 10.1083/jcb.201404066] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mutations in genes essential for protein homeostasis have been identified in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) patients. Why mature neurons should be particularly sensitive to such perturbations is unclear. We identified mutations in Rab8 in a genetic screen for enhancement of an FTD phenotype associated with ESCRT-III dysfunction. Examination of Rab8 mutants or motor neurons expressing a mutant ESCRT-III subunit, CHMP2B(Intron5), at the Drosophila melanogaster neuromuscular junction synapse revealed synaptic overgrowth and endosomal dysfunction. Expression of Rab8 rescued overgrowth phenotypes generated by CHMP2B(Intron5). In Rab8 mutant synapses, c-Jun N-terminal kinase (JNK)/activator protein-1 and TGF-β signaling were overactivated and acted synergistically to potentiate synaptic growth. We identify novel roles for endosomal JNK-scaffold POSH (Plenty-of-SH3s) and a JNK kinase kinase, TAK1, in regulating growth activation in Rab8 mutants. Our data uncover Rab8, POSH, and TAK1 as regulators of synaptic growth responses and point to recycling endosome as a key compartment for synaptic growth regulation during neurodegenerative processes.
Collapse
Affiliation(s)
- Ryan J H West
- Department of Biology and Hull York Medical School, University of York, Heslington, York YO10 5DD, England, UK Department of Biology and Hull York Medical School, University of York, Heslington, York YO10 5DD, England, UK
| | - Yubing Lu
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Bruno Marie
- Institute of Neurobiology, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico 00901
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605
| | - Sean T Sweeney
- Department of Biology and Hull York Medical School, University of York, Heslington, York YO10 5DD, England, UK Department of Biology and Hull York Medical School, University of York, Heslington, York YO10 5DD, England, UK
| |
Collapse
|
41
|
Chassefeyre R, Martínez-Hernández J, Bertaso F, Bouquier N, Blot B, Laporte M, Fraboulet S, Couté Y, Devoy A, Isaacs AM, Pernet-Gallay K, Sadoul R, Fagni L, Goldberg Y. Regulation of postsynaptic function by the dementia-related ESCRT-III subunit CHMP2B. J Neurosci 2015; 35:3155-73. [PMID: 25698751 PMCID: PMC4331633 DOI: 10.1523/jneurosci.0586-14.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 12/23/2014] [Accepted: 01/06/2015] [Indexed: 12/14/2022] Open
Abstract
The charged multivesicular body proteins (Chmp1-7) are an evolutionarily conserved family of cytosolic proteins that transiently assembles into helical polymers that change the curvature of cellular membrane domains. Mutations in human CHMP2B cause frontotemporal dementia, suggesting that this protein may normally control some neuron-specific process. Here, we examined the function, localization, and interactions of neuronal Chmp2b. The protein was highly expressed in mouse brain and could be readily detected in neuronal dendrites and spines. Depletion of endogenous Chmp2b reduced dendritic branching of cultured hippocampal neurons, decreased excitatory synapse density in vitro and in vivo, and abolished activity-induced spine enlargement and synaptic potentiation. To understand the synaptic effects of Chmp2b, we determined its ultrastructural distribution by quantitative immuno-electron microscopy and its biochemical interactions by coimmunoprecipitation and mass spectrometry. In the hippocampus in situ, a subset of neuronal Chmp2b was shown to concentrate beneath the perisynaptic membrane of dendritic spines. In synaptoneurosome lysates, Chmp2b was stably bound to a large complex containing other members of the Chmp family, as well as postsynaptic scaffolds. The supramolecular Chmp assembly detected here corresponds to a stable form of the endosomal sorting complex required for transport-III (ESCRT-III), a ubiquitous cytoplasmic protein complex known to play a central role in remodeling of lipid membranes. We conclude that Chmp2b-containing ESCRT-III complexes are also present at dendritic spines, where they regulate synaptic plasticity. We propose that synaptic ESCRT-III filaments may function as a novel element of the submembrane cytoskeleton of spines.
Collapse
Affiliation(s)
- Romain Chassefeyre
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - José Martínez-Hernández
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Federica Bertaso
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34094 Montpellier, France, Universités de Montpellier 1 & 2, UMR-5203, F-34094 Montpellier, France, INSERM, Unité 661, F-34094 Montpellier, France
| | - Nathalie Bouquier
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34094 Montpellier, France, Universités de Montpellier 1 & 2, UMR-5203, F-34094 Montpellier, France, INSERM, Unité 661, F-34094 Montpellier, France
| | - Béatrice Blot
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Marine Laporte
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Sandrine Fraboulet
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Yohann Couté
- INSERM, Unité 1038, F-38054 Grenoble, France, Commissariat à l'Energie Atomique (CEA), Institut de Recherches en Technologies et Sciences pour le Vivant (iRTSV), Laboratoire de Biologie à Grande Echelle, F-38054 Grenoble, France
| | - Anny Devoy
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London WC1N 3BG, United Kingdom, and
| | - Adrian M Isaacs
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London WC1N 3BG, United Kingdom, and
| | - Karin Pernet-Gallay
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France
| | - Rémy Sadoul
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France,
| | - Laurent Fagni
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34094 Montpellier, France, Universités de Montpellier 1 & 2, UMR-5203, F-34094 Montpellier, France, INSERM, Unité 661, F-34094 Montpellier, France
| | - Yves Goldberg
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 836, F-38042 Grenoble, France, Université Grenoble Alpes, Grenoble Institut des Neurosciences (GIN), F-38042 Grenoble, France, CEA, iRTSV, Groupe Physiopathologie du Cytosquelette (GPC), F-38054 Grenoble, France
| |
Collapse
|
42
|
Buratti E. Functional Significance of TDP-43 Mutations in Disease. ADVANCES IN GENETICS 2015; 91:1-53. [DOI: 10.1016/bs.adgen.2015.07.001] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
43
|
Agosta F, Al-Chalabi A, Filippi M, Hardiman O, Kaji R, Meininger V, Nakano I, Shaw P, Shefner J, van den Berg LH, Ludolph A. The El Escorial criteria: strengths and weaknesses. Amyotroph Lateral Scler Frontotemporal Degener 2014; 16:1-7. [PMID: 25482030 DOI: 10.3109/21678421.2014.964258] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The El Escorial criteria for the diagnosis of amyotrophic lateral sclerosis (ALS) were established 20 years ago and have been used as inclusion criteria for clinical trials. However, concerns have been raised concerning their use as diagnostic criteria in clinical practice. Moreover, as modern genetics have shed new light on the heterogeneity of ALS and the close relationship between ALS and frontotemporal dementia (FTD) recognized, the World Federation of Neurology Research Group on ALS/MND has initiated discussions to amend and update the criteria, while preserving the essential components for clinical trial enrolment purposes.
Collapse
Affiliation(s)
- Federica Agosta
- San Raffaele Scientific Institute and Vita-Salute San Raffaele University , Milan , Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Deivasigamani S, Verma HK, Ueda R, Ratnaparkhi A, Ratnaparkhi GS. A genetic screen identifies Tor as an interactor of VAPB in a Drosophila model of amyotrophic lateral sclerosis. Biol Open 2014; 3:1127-38. [PMID: 25361581 PMCID: PMC4232771 DOI: 10.1242/bio.201410066] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a progressive neurodegenerative disorder characterized by selective death of motor neurons. In 5–10% of the familial cases, the disease is inherited because of mutations. One such mutation, P56S, was identified in human VAPB that behaves in a dominant negative manner, sequestering wild type protein into cytoplasmic inclusions. We have conducted a reverse genetic screen to identify interactors of Drosophila VAPB. We screened 2635 genes and identified 103 interactors, of which 45 were enhancers and 58 were suppressors of VAPB function. Interestingly, the screen identified known ALS loci – TBPH, alsin2 and SOD1. Also identified were genes involved in cellular energetics and homeostasis which were used to build a gene regulatory network of VAPB modifiers. One key modifier identified was Tor, whose knockdown reversed the large bouton phenotype associated with VAP(P58S) expression in neurons. A similar reversal was seen by over-expressing Tuberous Sclerosis Complex (Tsc1,2) that negatively regulates TOR signaling as also by reduction of S6K activity. In comparison, the small bouton phenotype associated with VAP(wt) expression was reversed with Tsc1 knock down as well as S6K-CA expression. Tor therefore interacts with both VAP(wt) and VAP(P58S), but in a contrasting manner. Reversal of VAP(P58S) bouton phenotypes in larvae fed with the TOR inhibitor Rapamycin suggests upregulation of TOR signaling in response to VAP(P58S) expression. The VAPB network and further mechanistic understanding of interactions with key pathways, such as the TOR cassette, will pave the way for a better understanding of the mechanisms of onset and progression of motor neuron disease.
Collapse
Affiliation(s)
| | | | - Ryu Ueda
- National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | | | | |
Collapse
|
45
|
Abstract
Our understanding of amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease, is expanding rapidly as its genetic causes are uncovered. The pace of new gene discovery over the last 5 years has accelerated, providing new insights into the pathogenesis of disease and highlighting biological pathways as targets for therapeutic development. This article reviews our current understanding of the heritability of ALS and provides an overview of each of the major ALS genes, highlighting their phenotypic characteristics and frequencies as a guide for clinicians evaluating patients with ALS.
Collapse
Affiliation(s)
- Matthew B Harms
- Neuromuscular Division, Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO 63110, USA.
| | | |
Collapse
|
46
|
Chiò A, Battistini S, Calvo A, Caponnetto C, Conforti FL, Corbo M, Giannini F, Mandrioli J, Mora G, Sabatelli M, Ajmone C, Mastro E, Pain D, Mandich P, Penco S, Restagno G, Zollino M, Surbone A, Lunetta C, Pintor GL, Salvi F, Bartolomei I, Quattrone A, Gambardella A, Logroscino G, Simone I, Pisano F, Spataro R, La Bella V, Colletti T, Mancardi G, Origone P, Sola P, Borghero G, Marrosu F, Marrosu MG, Murru MR, Floris G, Cannas A, Piras V, Costantino E, Pani C, Sotgiu MA, Pugliatti M, Parish LD, Cossu P, Ticca A, Rodolico C, Portaro S, Ricci C, Moglia C, Ossola I, Brunetti M, Barberis M, Canosa A, Cammarosano S, Bertuzzo D, Fuda G, Ilardi A, Manera U, Pastore I, Sproviero W, Logullo F, Tanel R, Ajmone C, Mastro E, Pain D, Mandich P, Penco S, Restagno G, Zollino M, Surbone A. Genetic counselling in ALS: facts, uncertainties and clinical suggestions. J Neurol Neurosurg Psychiatry 2014; 85:478-85. [PMID: 23833266 DOI: 10.1136/jnnp-2013-305546] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The clinical approach to patients with amyotrophic lateral sclerosis (ALS) has been largely modified by the identification of novel genes, the detection of gene mutations in apparently sporadic patients, and the discovery of the strict genetic and clinical relation between ALS and frontotemporal dementia (FTD). As a consequence, clinicians are increasingly facing the dilemma on how to handle genetic counselling and testing both for ALS patients and their relatives. On the basis of existing literature on genetics of ALS and of other late-onset life-threatening disorders, we propose clinical suggestions to enable neurologists to provide optimal clinical and genetic counselling to patients and families. Genetic testing should be offered to ALS patients who have a first-degree or second-degree relative with ALS, FTD or both, and should be discussed with, but not offered to, all other ALS patients, with special emphasis on its major uncertainties. Presently, genetic testing should not be proposed to asymptomatic at-risk subjects, unless they request it or are enrolled in research programmes. Genetic counselling in ALS should take into account the uncertainties about the pathogenicity and penetrance of some genetic mutations; the possible presence of mutations of different genes in the same individual; the poor genotypic/phenotypic correlation in most ALS genes; and the phenotypic pleiotropy of some genes. Though psychological, social and ethical implications of genetic testing are still relatively unexplored in ALS, we recommend multidisciplinary counselling that addresses all relevant issues, including disclosure of tests results to family members and the risk for genetic discrimination.
Collapse
Affiliation(s)
- Adriano Chiò
- Department of Neuroscience, ALS Center, 'Rita Levi Montalcini', University of Torino, Torino, and Azienda Ospedaliera Città della Salute e della Scienza, , Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
van Blitterswijk M, Rademakers R, van den Berg LH. Clinical variability and additional mutations in amyotrophic lateral sclerosis patients with p.N352S mutations inTARDBP. Neuropathol Appl Neurobiol 2014; 40:356-8. [DOI: 10.1111/nan.12099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 10/30/2013] [Indexed: 12/14/2022]
Affiliation(s)
| | - R. Rademakers
- Department of Neuroscience; Mayo Clinic; Jacksonville FL USA
| | - L. H. van den Berg
- Department of Neurology; Rudolf Magnus Institute of Neuroscience; University Medical Center Utrecht; Utrecht The Netherlands
| |
Collapse
|
48
|
Recent progress in the genetics of motor neuron disease. Eur J Med Genet 2014; 57:103-12. [DOI: 10.1016/j.ejmg.2014.01.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 01/14/2014] [Indexed: 01/07/2023]
|
49
|
Homma T, Nagaoka U, Kawata A, Mochizuki Y, Kawakami H, Maruyama H, Matsubara S, Komori T. Neuropathological features of Japanese familial amyotrophic lateral sclerosis with p.N352S mutation inTARDBP. Neuropathol Appl Neurobiol 2014; 40:231-6. [DOI: 10.1111/nan.12090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/02/2013] [Indexed: 12/13/2022]
Affiliation(s)
- T. Homma
- Department of Laboratory Medicine and Pathology (Neuropathology); Tokyo Metropolitan Neurological Hospital; Tokyo Japan
- Department of Pathology; Saitama Medical University; Saitama Japan
| | - U. Nagaoka
- Department of Neurology; Tokyo Metropolitan Neurological Hospital; Tokyo Japan
| | - A. Kawata
- Department of Neurology; Tokyo Metropolitan Neurological Hospital; Tokyo Japan
| | - Y. Mochizuki
- Department of Laboratory Medicine and Pathology (Neuropathology); Tokyo Metropolitan Neurological Hospital; Tokyo Japan
- Department of Neurology; Tokyo Metropolitan Kita Medical and Rehabilitation Center for the Disabled; Tokyo Japan
| | - H. Kawakami
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine; Hiroshima University; Hiroshima Japan
| | - H. Maruyama
- Department of Epidemiology, Research Institute for Radiation Biology and Medicine; Hiroshima University; Hiroshima Japan
| | - S. Matsubara
- Department of Neurology; Tokyo Metropolitan Neurological Hospital; Tokyo Japan
| | - T. Komori
- Department of Laboratory Medicine and Pathology (Neuropathology); Tokyo Metropolitan Neurological Hospital; Tokyo Japan
| |
Collapse
|
50
|
Homma T, Nagaoka U, Kawata A, Mochizuki Y, Kawakami H, Maruyama H, Matsubara S, Komori T. Authors' reply to Drs M van Blitterswijk, R Rademakers and LH van den Berg. Neuropathol Appl Neurobiol 2013; 40:359-60. [PMID: 24304300 DOI: 10.1111/nan.12105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/02/2013] [Indexed: 11/28/2022]
Affiliation(s)
- T Homma
- Department of Laboratory Medicine and Pathology (Neuropathology), Tokyo Metropolitan Neurological Hospital, Tokyo, Japan; Department of Pathology, Saitama Medical University, Saitama, Japan
| | | | | | | | | | | | | | | |
Collapse
|