201
|
Fogh I, Lin K, Tiloca C, Rooney J, Gellera C, Diekstra FP, Ratti A, Shatunov A, van Es MA, Proitsi P, Jones A, Sproviero W, Chiò A, McLaughlin RL, Sorarù G, Corrado L, Stahl D, Del Bo R, Cereda C, Castellotti B, Glass JD, Newhouse S, Dobson R, Smith BN, Topp S, van Rheenen W, Meininger V, Melki J, Morrison KE, Shaw PJ, Leigh PN, Andersen PM, Comi GP, Ticozzi N, Mazzini L, D'Alfonso S, Traynor BJ, Van Damme P, Robberecht W, Brown RH, Landers JE, Hardiman O, Lewis CM, van den Berg LH, Shaw CE, Veldink JH, Silani V, Al-Chalabi A, Powell J. Association of a Locus in the CAMTA1 Gene With Survival in Patients With Sporadic Amyotrophic Lateral Sclerosis. JAMA Neurol 2016; 73:812-20. [PMID: 27244217 PMCID: PMC5556366 DOI: 10.1001/jamaneurol.2016.1114] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
IMPORTANCE Amyotrophic lateral sclerosis (ALS) is a devastating adult-onset neurodegenerative disorder with a poor prognosis and a median survival of 3 years. However, a significant proportion of patients survive more than 10 years from symptom onset. OBJECTIVE To identify gene variants influencing survival in ALS. DESIGN, SETTING, AND PARTICIPANTS This genome-wide association study (GWAS) analyzed survival in data sets from several European countries and the United States that were collected by the Italian Consortium for the Genetics of ALS and the International Consortium on Amyotrophic Lateral Sclerosis Genetics. The study population included 4256 patients with ALS (3125 [73.4%] deceased) with genotype data extended to 7 174 392 variants by imputation analysis. Samples of DNA were collected from January 1, 1993, to December 31, 2009, and analyzed from March 1, 2014, to February 28, 2015. MAIN OUTCOMES AND MEASURES Cox proportional hazards regression under an additive model with adjustment for age at onset, sex, and the first 4 principal components of ancestry, followed by meta-analysis, were used to analyze data. Survival distributions for the most associated genetic variants were assessed by Kaplan-Meier analysis. RESULTS Among the 4256 patients included in the analysis (2589 male [60.8%] and 1667 female [39.2%]; mean [SD] age at onset, 59 [12] years), the following 2 novel loci were significantly associated with ALS survival: at 10q23 (rs139550538; P = 1.87 × 10-9) and in the CAMTA1 gene at 1p36 (rs2412208, P = 3.53 × 10-8). At locus 10q23, the adjusted hazard ratio for patients with the rs139550538 AA or AT genotype was 1.61 (95% CI, 1.38-1.89; P = 1.87 × 10-9), corresponding to an 8-month reduction in survival compared with TT carriers. For rs2412208 CAMTA1, the adjusted hazard ratio for patients with the GG or GT genotype was 1.17 (95% CI, 1.11-1.24; P = 3.53 × 10-8), corresponding to a 4-month reduction in survival compared with TT carriers. CONCLUSIONS AND RELEVANCE This GWAS robustly identified 2 loci at genome-wide levels of significance that influence survival in patients with ALS. Because ALS is a rare disease and prevention is not feasible, treatment that modifies survival is the most realistic strategy. Therefore, identification of modifier genes that might influence ALS survival could improve the understanding of the biology of the disease and suggest biological targets for pharmaceutical intervention. In addition, genetic risk scores for survival could be used as an adjunct to clinical trials to account for the genetic contribution to survival.
Collapse
Affiliation(s)
- Isabella Fogh
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Kuang Lin
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Cinzia Tiloca
- Department of Neurology and Laboratory of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milano, Italy
| | - James Rooney
- Academic Unit of Neurology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Cinzia Gellera
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Frank P Diekstra
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milano, Italy6Department of Pathophysiology and Tranplantation, Dino Ferrari Center, Università degli Studi d
| | - Aleksey Shatunov
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Michael A van Es
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Petroula Proitsi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Ashley Jones
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - William Sproviero
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Adriano Chiò
- Rita Levi Montalcini Department of Neuroscience, ALS (Amyotrophic Lateral Sclerosis) Centre, University of Torino, Turin, Italy8Azienda Ospedaliera Città della Salute e della Scienza, Torino, Italy
| | - Russell Lewis McLaughlin
- Population Genetics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padua, Italy
| | - Lucia Corrado
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, A. Avogadro University, Novara, Italy
| | - Daniel Stahl
- Department of Biostatistics, IoPPN, King's College London, London, England
| | - Roberto Del Bo
- Neurologic Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristina Cereda
- Laboratory of Experimental Neurobiology, IRCCS C. Mondino National Institute of Neurology Foundation, Pavia, Italy
| | - Barbara Castellotti
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | | | - Steven Newhouse
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health, IoPPN, King's College London, London, England17Department of Biostatistics, IoPPN, King's College London, London, England
| | - Richard Dobson
- National Institute for Health Research (NIHR) Biomedical Research Centre for Mental Health, IoPPN, King's College London, London, England18NIHR Biomedical Research Unit in Dementia, King's College London, London, England
| | - Bradley N Smith
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Simon Topp
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Wouter van Rheenen
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Vincent Meininger
- Département des Maladies du Système Nerveux, Assistance Publique-Hôpitaux de Paris, Réseau SLA (Sclérose Latérale) Île de France, Hôpital Pitié-Salpêtrière, Paris, France
| | - Judith Melki
- Institut National de la Santé et de la Recherche Medicale Unité Mixte de Recherché-788 and University of Paris 11, Bicetre Hospital, Paris, France
| | - Karen E Morrison
- School of Clinical and Experimental Medicine, College of Medicine and Dentistry, University of Birmingham, Birmingham, England22Neurosciences Division, University Hospitals Birmingham National Health Service Foundation Trust, Birmingham, England
| | - Pamela J Shaw
- Academic Neurology Unit, Department of Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, England
| | - P Nigel Leigh
- Section of Neurology, Division of Medicine, Brighton and Sussex Medical School, Trafford Centre for Biomedical Research, University of Sussex, East Sussex, England
| | - Peter M Andersen
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany26Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Giacomo P Comi
- Neurologic Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Nicola Ticozzi
- Department of Neurology and Laboratory of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milano, Italy6Department of Pathophysiology and Tranplantation, Dino Ferrari Center, Università degli Studi d
| | - Letizia Mazzini
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, A. Avogadro University, Novara, Italy27ALS Center Department of Neurology, Maggiore della Carità University Hospital, Novara, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases, A. Avogadro University, Novara, Italy
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology, Flanders Instititue for Biotechnology, Vesalius Research Center, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium30Department of Neurology, University Hospitals Leuven, Leuven
| | - Wim Robberecht
- Department of Neurosciences, Experimental Neurology, Flanders Instititue for Biotechnology, Vesalius Research Center, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester
| | - Orla Hardiman
- Population Genetics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Cathryn M Lewis
- IoPPN Genomics and Biomarker Core, Translational Genetics Group, Medical Research Council Social, Genetic and Developmental Psychiatry Centre, King's College London, London, England33Department of Medical and Molecular Genetics, King's College London, Lon
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Christopher E Shaw
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Auxologico Italiano, Milano, Italy6Department of Pathophysiology and Tranplantation, Dino Ferrari Center, Università degli Studi d
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| | - John Powell
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience (IoPPN), King's College London, London, England
| |
Collapse
|
202
|
Zufiría M, Gil-Bea FJ, Fernández-Torrón R, Poza JJ, Muñoz-Blanco JL, Rojas-García R, Riancho J, López de Munain A. ALS: A bucket of genes, environment, metabolism and unknown ingredients. Prog Neurobiol 2016; 142:104-129. [DOI: 10.1016/j.pneurobio.2016.05.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 03/22/2016] [Accepted: 05/09/2016] [Indexed: 12/11/2022]
|
203
|
De Marco G, Lomartire A, Calvo A, Risso A, De Luca E, Mostert M, Mandrioli J, Caponnetto C, Borghero G, Manera U, Canosa A, Moglia C, Restagno G, Fini N, Tarella C, Giordana MT, Rinaudo MT, Chiò A. Monocytes of patients with amyotrophic lateral sclerosis linked to gene mutations display altered TDP-43 subcellular distribution. Neuropathol Appl Neurobiol 2016; 43:133-153. [PMID: 27178390 DOI: 10.1111/nan.12328] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/20/2016] [Accepted: 05/14/2016] [Indexed: 12/12/2022]
Abstract
AIMS Cytoplasmic accumulation of the nuclear protein transactive response DNA-binding protein 43 (TDP-43) is an early determinant of motor neuron degeneration in most amyotrophic lateral sclerosis (ALS) cases. We previously disclosed this accumulation in circulating lymphomonocytes (CLM) of ALS patients with mutant TARDBP, the TDP-43-coding gene, as well as of a healthy individual carrying the parental TARDBP mutation. Here, we investigate TDP-43 subcellular localization in CLM and in the constituent cells, lymphocytes and monocytes, of patients with various ALS-linked mutant genes. METHODS TDP-43 subcellular localization was analysed with western immunoblotting and immunocytofluorescence in CLM of healthy controls (n = 10), patients with mutant TARDBP (n = 4, 1 homozygous), valosin-containing protein (VCP; n = 2), fused in sarcoma/translocated in liposarcoma (FUS; n = 2), Cu/Zn superoxide dismutase 1 (SOD1; n = 6), chromosome 9 open reading frame 72 (C9ORF72; n = 4), without mutations (n = 5) and neurologically unaffected subjects with mutant TARDBP (n = 2). RESULTS TDP-43 cytoplasmic accumulation was found (P < 0.05 vs. controls) in CLM of patients with mutant TARDBP or VCP, but not FUS, in line with TDP-43 subcellular localization described for motor neurons of corresponding groups. Accumulation also characterized CLM of the healthy individuals with mutant TARDBP and of some patients with mutant SOD1 or C9ORF72. In 5 patients, belonging to categories described to carry TDP-43 mislocalization in motor neurons (3 C9ORF72, 1 TARDBP and 1 without mutations), TDP-43 cytoplasmic accumulation was not detected in CLM or in lymphocytes but was in monocytes. CONCLUSIONS In ALS forms characterized by TDP-43 mislocalization in motor neurons, monocytes display this alteration, even when not manifest in CLM. Monocytes may be used to support diagnosis, as well as to identify subjects at risk, of ALS and to develop/monitor targeted treatments.
Collapse
Affiliation(s)
- G De Marco
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - A Lomartire
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - A Calvo
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,ALS Center, University of Turin and AOU Città della Salute e della Scienza, Turin, Italy
| | - A Risso
- Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - E De Luca
- Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - M Mostert
- Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - J Mandrioli
- Department of Neuroscience, Sant'Agostino Estense Hospital, University of Modena, Modena, Italy
| | - C Caponnetto
- Department of Neurosciences, Ophthalmology, Genetics, Rehabilitation and Child Health, IRCCS AOU San Martino IST, University of Genoa, Genoa, Italy
| | - G Borghero
- Department of Neurology, AOU and University of Cagliari, Cagliari, Italy
| | - U Manera
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,ALS Center, University of Turin and AOU Città della Salute e della Scienza, Turin, Italy
| | - A Canosa
- ALS Center, University of Turin and AOU Città della Salute e della Scienza, Turin, Italy.,Department of Neurosciences, Ophthalmology, Genetics, Rehabilitation and Child Health, IRCCS AOU San Martino IST, University of Genoa, Genoa, Italy
| | - C Moglia
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,ALS Center, University of Turin and AOU Città della Salute e della Scienza, Turin, Italy
| | - G Restagno
- Molecular Genetics Unit, Department of Clinical Pathology, AOU Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - N Fini
- Department of Neuroscience, Sant'Agostino Estense Hospital, University of Modena, Modena, Italy
| | - C Tarella
- Clinical Hemato-Oncology, European Institute of Oncology (IEO), Milan, Italy
| | - M T Giordana
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - M T Rinaudo
- Department of Oncology, University of Turin, Turin, Italy
| | - A Chiò
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy.,ALS Center, University of Turin and AOU Città della Salute e della Scienza, Turin, Italy
| |
Collapse
|
204
|
Webster CP, Smith EF, Bauer CS, Moller A, Hautbergue GM, Ferraiuolo L, Myszczynska MA, Higginbottom A, Walsh MJ, Whitworth AJ, Kaspar BK, Meyer K, Shaw PJ, Grierson AJ, De Vos KJ. The C9orf72 protein interacts with Rab1a and the ULK1 complex to regulate initiation of autophagy. EMBO J 2016; 35:1656-76. [PMID: 27334615 PMCID: PMC4969571 DOI: 10.15252/embj.201694401] [Citation(s) in RCA: 295] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/03/2016] [Indexed: 12/12/2022] Open
Abstract
A GGGGCC hexanucleotide repeat expansion in the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD). C9orf72 encodes two C9orf72 protein isoforms of unclear function. Reduced levels of C9orf72 expression have been reported in C9ALS/FTD patients, and although C9orf72 haploinsufficiency has been proposed to contribute to C9ALS/FTD, its significance is not yet clear. Here, we report that C9orf72 interacts with Rab1a and the Unc‐51‐like kinase 1 (ULK1) autophagy initiation complex. As a Rab1a effector, C9orf72 controls initiation of autophagy by regulating the Rab1a‐dependent trafficking of the ULK1 autophagy initiation complex to the phagophore. Accordingly, reduction of C9orf72 expression in cell lines and primary neurons attenuated autophagy and caused accumulation of p62‐positive puncta reminiscent of the p62 pathology observed in C9ALS/FTD patients. Finally, basal levels of autophagy were markedly reduced in C9ALS/FTD patient‐derived iNeurons. Thus, our data identify C9orf72 as a novel Rab1a effector in the regulation of autophagy and indicate that C9orf72 haploinsufficiency and associated reductions in autophagy might be the underlying cause of C9ALS/FTD‐associated p62 pathology.
Collapse
Affiliation(s)
- Christopher P Webster
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Emma F Smith
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Claudia S Bauer
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Annekathrin Moller
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Monika A Myszczynska
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Matthew J Walsh
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | | | - Brian K Kaspar
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Kathrin Meyer
- The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Andrew J Grierson
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| | - Kurt J De Vos
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience University of Sheffield, Sheffield, UK
| |
Collapse
|
205
|
Todd TW, Petrucelli L. Insights into the pathogenic mechanisms of Chromosome 9 open reading frame 72 (C9orf72) repeat expansions. J Neurochem 2016; 138 Suppl 1:145-62. [PMID: 27016280 DOI: 10.1111/jnc.13623] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/04/2016] [Accepted: 03/21/2016] [Indexed: 12/12/2022]
Abstract
The identification of a hexanucleotide repeat expansion in a non-coding region of C9orf72 as a major cause of both frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) drastically changed the field of research on both of these conditions. Yet, despite the vast amount of work aimed at elucidating the molecular mechanisms underlying the role of this repeat in disease, the exact pathomechanisms are still unclear. A reduction in the expression of the C9orf72 gene is observed in patients, but a gain-of-function model is now preferred. The hexanucleotide repeat expansion forms RNA foci in the central nervous system (CNS) of repeat-positive FTD and ALS patients, and these foci are believed to sequester RNA-binding proteins (RBPs) and impair their function in RNA processing. At the same time, the repeat undergoes repeat-associated non-ATG translation to produce dipeptide repeat proteins that also form inclusions in the patient CNS. Studies from cells and flies suggest that these proteins may also be an important factor in the disease. Finally, the hexanucleotide repeat also induces the mislocalization and aggregation of TAR DNA-binding protein 43 (TDP-43) through an as yet unknown mechanism. This review covers the different potential pathogenic factors that have been put forth for C9orf72-repeat-associated FTD and ALS (C9-FTD/ALS), while highlighting some remaining questions. A repeat expansion in C9orf72 is a common cause of both frontal temporal dementia and amyotrophic lateral sclerosis. Although there is a decrease in C9orf72 expression in patients, this repeat is believed to induce disease primarily through an unknown gain-of-function mechanism involving the RNA, repeat-associated non-AUG translation, or both. This review summarizes and discusses current knowledge on C9orf72 repeat-associated pathophysiology. This article is part of the Frontotemporal Dementia special issue.
Collapse
Affiliation(s)
- Tiffany W Todd
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
206
|
Geevasinga N, Menon P, Ng K, Van Den Bos M, Byth K, Kiernan MC, Vucic S. Riluzole exerts transient modulating effects on cortical and axonal hyperexcitability in ALS. Amyotroph Lateral Scler Frontotemporal Degener 2016; 17:580-588. [DOI: 10.1080/21678421.2016.1188961] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | | | - Karl Ng
- Department of Neurology, Royal North Shore Hospital, St. Leonards, University of Sydney, Sydney,
| | | | - Karen Byth
- Westmead Hospital, Research and Education Network, Sydney, Australia NHMRC Clinical Trials Centre, University of Sydney, Sydney, and
| | | | - Steve Vucic
- Western Clinical School, University of Sydney, Sydney,
| |
Collapse
|
207
|
Menke RAL, Proudfoot M, Wuu J, Andersen PM, Talbot K, Benatar M, Turner MR. Increased functional connectivity common to symptomatic amyotrophic lateral sclerosis and those at genetic risk. J Neurol Neurosurg Psychiatry 2016; 87:580-8. [PMID: 26733601 PMCID: PMC4893149 DOI: 10.1136/jnnp-2015-311945] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/18/2015] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To discern presymptomatic changes in brain structure or function using advanced MRI in carriers of mutations predisposing to amyotrophic lateral sclerosis (ALS). METHODS T1-weighted, diffusion weighted and resting state functional MRI data were acquired at 3 T for 12 asymptomatic mutation carriers (psALS), 12 age-matched controls and affected patients with ALS. Cortical thickness analysis, voxel-based morphometry, volumetric and shape analyses of subcortical structures, tract-based spatial statistics of metrics derived from the diffusion tensor, and resting state functional connectivity (FC) analyses were performed. RESULTS Grey matter cortical thickness and shape analysis revealed significant atrophy in patients with ALS (but not psALS) compared with controls in the right primary motor cortex and right caudate. Comparison of diffusion tensor metrics showed widespread fractional anisotropy and radial diffusivity differences in patients with ALS compared to controls and the psALS group, encompassing parts of the corpus callosum, corticospinal tracts and superior longitudinal fasciculus. While FC in the resting-state sensorimotor network was similar in psALS and controls, FC between the cerebellum and a network comprising the precuneus, cingulate & middle frontal lobe was significantly higher in psALS and affected ALS compared to controls. CONCLUSIONS Rather than structural brain changes, increased FC may be among the earliest detectable brain abnormalities in asymptomatic carriers of ALS-causing gene mutations. With replication and significant refinement, this technique has potential in the future assessment of neuroprotective strategies.
Collapse
Affiliation(s)
- Ricarda A L Menke
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK FMRIB Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Malcolm Proudfoot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Joanne Wuu
- Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Peter M Andersen
- Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Michael Benatar
- Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK FMRIB Centre, John Radcliffe Hospital, University of Oxford, Oxford, UK
| |
Collapse
|
208
|
Gelpi E. How neuropathology can contribute to the understanding of dementia. Neurodegener Dis Manag 2016; 6:183-6. [PMID: 27230123 DOI: 10.2217/nmt-2016-0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Ellen Gelpi
- Neurological Tissue Bank of the Biobank-Hospital Clinic-Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
209
|
Halliday GM, Kiernan MC, Kril JJ, Mito R, Masuda-Suzukake M, Hasegawa M, McCann H, Bartley L, Dobson-Stone C, Kwok JBJ, Hornberger M, Hodges JR, Tan RH. TDP-43 in the hypoglossal nucleus identifies amyotrophic lateral sclerosis in behavioral variant frontotemporal dementia. J Neurol Sci 2016; 366:197-201. [PMID: 27288806 DOI: 10.1016/j.jns.2016.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 04/22/2016] [Accepted: 05/03/2016] [Indexed: 11/16/2022]
Abstract
The hypoglossal nucleus was recently identified as a key brain region in which the presence of TDP-43 pathology could accurately discriminate TDP-43 proteinopathy cases with clinical amyotrophic lateral sclerosis (ALS). The objective of the present study was to assess the hypoglossal nucleus in behavioral variant frontotemporal dementia (bvFTD), and determine whether TDP-43 in this region is associated with clinical ALS. Twenty-nine cases with neuropathological FTLD-TDP and clinical bvFTD that had not been previously assessed for hypoglossal TDP-43 pathology were included in this study. Of these 29 cases, 41% (n=12) had a dual diagnosis of bvFTD-ALS at presentation, all 100% (n=12) of which demonstrated hypoglossal TDP-43 pathology. Of the 59% (n=17) cohort that presented with pure bvFTD, 35% (n=6) were identified with hypoglossal TDP-43 pathology. Review of the case files of all pure bvFTD cases revealed evidence of possible or probable ALS in 5 of the 6 hypoglossal-positive cases (83%) towards the end of disease, and this was absent from all cases without such pathology. In conclusion, the present study validates grading the presence of TDP-43 in the hypoglossal nucleus for the pathological identification of bvFTD cases with clinical ALS, and extends this to include the identification of cases with possible ALS at end-stage.
Collapse
Affiliation(s)
- Glenda M Halliday
- Neuroscience Research Australia, Sydney, Australia; School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, Sydney Medical School, The University of Sydney, Australia
| | - Jillian J Kril
- Discipline of Pathology, Sydney Medical School, The University of Sydney, Australia
| | - Remika Mito
- Discipline of Pathology, Sydney Medical School, The University of Sydney, Australia
| | - Masami Masuda-Suzukake
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Japan
| | - Masato Hasegawa
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Japan
| | | | | | - Carol Dobson-Stone
- Neuroscience Research Australia, Sydney, Australia; School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - John B J Kwok
- Neuroscience Research Australia, Sydney, Australia; School of Medical Sciences, University of New South Wales, Sydney, Australia
| | | | - John R Hodges
- Neuroscience Research Australia, Sydney, Australia; School of Medical Sciences, University of New South Wales, Sydney, Australia; ARC Centre of Excellence in Cognition and Its Disorders, Sydney, Australia
| | - Rachel H Tan
- Neuroscience Research Australia, Sydney, Australia; School of Medical Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
210
|
C9orf72 BAC Mouse Model with Motor Deficits and Neurodegenerative Features of ALS/FTD. Neuron 2016; 90:521-34. [DOI: 10.1016/j.neuron.2016.04.005] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/01/2016] [Accepted: 03/29/2016] [Indexed: 12/12/2022]
|
211
|
Sellier C, Campanari ML, Julie Corbier C, Gaucherot A, Kolb-Cheynel I, Oulad-Abdelghani M, Ruffenach F, Page A, Ciura S, Kabashi E, Charlet-Berguerand N. Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J 2016; 35:1276-97. [PMID: 27103069 DOI: 10.15252/embj.201593350] [Citation(s) in RCA: 308] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 03/15/2016] [Indexed: 12/12/2022] Open
Abstract
An intronic expansion of GGGGCC repeats within the C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD). Ataxin-2 with intermediate length of polyglutamine expansions (Ataxin-2 Q30x) is a genetic modifier of the disease. Here, we found that C9ORF72 forms a complex with the WDR41 and SMCR8 proteins to act as a GDP/GTP exchange factor for RAB8a and RAB39b and to thereby control autophagic flux. Depletion of C9orf72 in neurons partly impairs autophagy and leads to accumulation of aggregates of TDP-43 and P62 proteins, which are histopathological hallmarks of ALS-FTD SMCR8 is phosphorylated by TBK1 and depletion of TBK1 can be rescued by phosphomimetic mutants of SMCR8 or by constitutively active RAB39b, suggesting that TBK1, SMCR8, C9ORF72, and RAB39b belong to a common pathway regulating autophagy. While depletion of C9ORF72 only has a partial deleterious effect on neuron survival, it synergizes with Ataxin-2 Q30x toxicity to induce motor neuron dysfunction and neuronal cell death. These results indicate that partial loss of function of C9ORF72 is not deleterious by itself but synergizes with Ataxin-2 toxicity, suggesting a double-hit pathological mechanism in ALS-FTD.
Collapse
Affiliation(s)
- Chantal Sellier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Maria-Letizia Campanari
- Sorbonne Université, Université Pierre et Marie Curie (UPMC), Université de Paris 06, Unité Mixte 75, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1127, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225 Institut du Cerveau et de la Moelle Épinière (ICM), 75013, Paris, France
| | - Camille Julie Corbier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Angeline Gaucherot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Isabelle Kolb-Cheynel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Mustapha Oulad-Abdelghani
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Frank Ruffenach
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Adeline Page
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Sorana Ciura
- Sorbonne Université, Université Pierre et Marie Curie (UPMC), Université de Paris 06, Unité Mixte 75, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1127, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225 Institut du Cerveau et de la Moelle Épinière (ICM), 75013, Paris, France
| | - Edor Kabashi
- Sorbonne Université, Université Pierre et Marie Curie (UPMC), Université de Paris 06, Unité Mixte 75, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1127, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche 7225 Institut du Cerveau et de la Moelle Épinière (ICM), 75013, Paris, France
| | - Nicolas Charlet-Berguerand
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| |
Collapse
|
212
|
Bede P, Iyer PM, Schuster C, Elamin M, Mclaughlin RL, Kenna K, Hardiman O. The selective anatomical vulnerability of ALS: 'disease-defining' and 'disease-defying' brain regions. Amyotroph Lateral Scler Frontotemporal Degener 2016; 17:561-570. [PMID: 27087114 DOI: 10.3109/21678421.2016.1173702] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A large multiparametric MRI study has been undertaken to evaluate anatomical patterns of basal ganglia, white matter and cortical grey matter involvement in ALS. Unaffected brain regions are mapped in patients with significant disability. Multiple white matter diffusivity measures, cortical grey matter density alterations, basal ganglia volumes and subcortical grey matter atrophy patterns are evaluated. Results demonstrated a strikingly selective anatomical vulnerability pattern in ALS that preferentially affects specific grey matter structures, commissural white matter tracts and basal ganglia regions, suggestive of networkwise neurodegeneration in ALS. In conclusion, ALS pathology exhibits predilection for selective and inter-connected anatomical sites that can be comprehensively characterized in vivo by multiparametric neuroimaging. The systematic characterization of unaffected brain regions in ALS has implications for the development of classifier analyses and elucidation of disease biology. The involvement and sparing of contiguous brain regions raises important pathophysiological, phylogenetic and ontogenetic questions regarding ALS pathogenesis and disease spread.
Collapse
Affiliation(s)
- Peter Bede
- a Quantitative Neuroimaging Group, Academic Unit of Neurology , Biomedical Sciences Institute, Trinity College Dublin , Ireland
| | - Parameswaran M Iyer
- a Quantitative Neuroimaging Group, Academic Unit of Neurology , Biomedical Sciences Institute, Trinity College Dublin , Ireland
| | - Christina Schuster
- a Quantitative Neuroimaging Group, Academic Unit of Neurology , Biomedical Sciences Institute, Trinity College Dublin , Ireland
| | - Marwa Elamin
- a Quantitative Neuroimaging Group, Academic Unit of Neurology , Biomedical Sciences Institute, Trinity College Dublin , Ireland
| | - Russell L Mclaughlin
- a Quantitative Neuroimaging Group, Academic Unit of Neurology , Biomedical Sciences Institute, Trinity College Dublin , Ireland
| | - Kevin Kenna
- a Quantitative Neuroimaging Group, Academic Unit of Neurology , Biomedical Sciences Institute, Trinity College Dublin , Ireland
| | - Orla Hardiman
- a Quantitative Neuroimaging Group, Academic Unit of Neurology , Biomedical Sciences Institute, Trinity College Dublin , Ireland
| |
Collapse
|
213
|
Protein folding alterations in amyotrophic lateral sclerosis. Brain Res 2016; 1648:633-649. [PMID: 27064076 DOI: 10.1016/j.brainres.2016.04.010] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/04/2016] [Accepted: 04/05/2016] [Indexed: 02/07/2023]
Abstract
Protein misfolding leads to the formation of aggregated proteins and protein inclusions, which are associated with synaptic loss and neuronal death in neurodegenerative diseases. Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that targets motor neurons in the brain, brainstem and spinal cord. Several proteins misfold and are associated either genetically or pathologically in ALS, including superoxide dismutase 1 (SOD1), Tar DNA binding protein-43 (TDP-43), Ubiquilin-2, p62, VCP, and dipeptide repeat proteins produced by unconventional repeat associated non-ATG translation of the repeat expansion in C9ORF72. Chaperone proteins, including heat shock proteins (Hsp׳s) and the protein disulphide isomerase (PDI) family, assist in protein folding and therefore can prevent protein misfolding, and have been implicated as being protective in ALS. In this review we provide an overview of the current literature regarding the molecular mechanisms of protein misfolding and aggregation in ALS, and the role of chaperones as potential targets for therapeutic intervention. This article is part of a Special Issue entitled SI:ER stress.
Collapse
|
214
|
Green KM, Linsalata AE, Todd PK. RAN translation-What makes it run? Brain Res 2016; 1647:30-42. [PMID: 27060770 DOI: 10.1016/j.brainres.2016.04.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/24/2016] [Accepted: 04/01/2016] [Indexed: 12/14/2022]
Abstract
Nucleotide-repeat expansions underlie a heterogeneous group of neurodegenerative and neuromuscular disorders for which there are currently no effective therapies. Recently, it was discovered that such repetitive RNA motifs can support translation initiation in the absence of an AUG start codon across a wide variety of sequence contexts, and that the products of these atypical translation initiation events contribute to neuronal toxicity. This review examines what we currently know and do not know about repeat associated non-AUG (RAN) translation in the context of established canonical and non-canonical mechanisms of translation initiation. We highlight recent findings related to RAN translation in three repeat expansion disorders: CGG repeats in fragile X-associated tremor ataxia syndrome (FXTAS), GGGGCC repeats in C9orf72 associated amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) and CAG repeats in Huntington disease. These studies suggest that mechanistic differences may exist for RAN translation dependent on repeat type, repeat reading frame, and the surrounding sequence context, but that for at least some repeats, RAN translation retains a dependence on some of the canonical translational initiation machinery. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.
Collapse
Affiliation(s)
- Katelyn M Green
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Alexander E Linsalata
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Peter K Todd
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Veterans Affairs Medical Center, Ann Arbor, MI, United States.
| |
Collapse
|
215
|
Geevasinga N, Menon P, Nicholson GA, Ng K, Howells J, Kril JJ, Yiannikas C, Kiernan MC, Vucic S. Cortical Function in Asymptomatic Carriers and Patients With C9orf72 Amyotrophic Lateral Sclerosis. JAMA Neurol 2016; 72:1268-74. [PMID: 26348842 DOI: 10.1001/jamaneurol.2015.1872] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IMPORTANCE The identification of the chromosome 9 open reading frame 72 (c9orf72) gene hexanucleotide repeat expansion represents a major advance in the understanding of amyotrophic lateral sclerosis (ALS) pathogenesis. The pathophysiological mechanism by which the c9orf72 gene expansion leads to neurodegeneration is not yet elucidated. Cortical hyperexcitability is potentially an important pathophysiological process in sporadic ALS and familial ALS (FALS). OBJECTIVE To investigate whether cortical hyperexcitability forms the pathophysiological basis of c9orf72 FALS using the threshold-tracking transcranial magnetic stimulation technique. DESIGN, SETTING, AND PARTICIPANTS Prospective case-control single-center study that took place at hospitals and outpatient clinics from January 1, 2013, to January 1, 2015. Clinical and functional assessments along with transcranial magnetic stimulation studies were taken on 15 patients with c9orf72 FALS and 11 asymptomatic expansion carriers of c9orf72 who were longitudinally followed up for 3 years. Results were compared with 73 patients with sporadic ALS and 74 healthy control participants. MAIN OUTCOMES AND MEASURES Cortical excitability variables, including short-interval intracortical inhibition, were measured in patients with c9orf72 FALS and results were compared with asymptomatic c9orf72 carriers, patients with sporadic ALS, and healthy control participants. RESULTS Mean (SD) short-interval intracortical inhibition was significantly reduced in patients with c9orf72 FALS (1.2% [1.8%]) and sporadic ALS (1.6% [1.2%]) compared with asymptomatic c9orf72 expansion carriers (10.2% [1.8%]; F = 16.1; P < .001) and healthy control participants (11.8% [1.0%]; F = 16.1; P < .001). The reduction of short-interval intracortical inhibition was accompanied by an increase in intracortical facilitation (P < .01) and motor-evoked potential amplitude (P < .05) as well as a reduction in the resting motor threshold (P < .05) and cortical silent period duration (P < .001). CONCLUSIONS AND RELEVANCE This study establishes cortical hyperexcitability as an intrinsic feature of symptomatic c9orf72 expansion-related ALS but not asymptomatic expansion carriers.
Collapse
Affiliation(s)
- Nimeshan Geevasinga
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Parvathi Menon
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Garth A Nicholson
- Northcott Neuroscience Laboratory, ANZAC Research Institute, Sydney, New South Wales, Australia
| | - Karl Ng
- Department of Neurology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - James Howells
- Brain and Mind Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Jillian J Kril
- Disciplines of Medicine and Pathology, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Con Yiannikas
- Department of Neurology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Matthew C Kiernan
- Brain and Mind Research Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Steve Vucic
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
216
|
Ishiguro A, Kimura N, Watanabe Y, Watanabe S, Ishihama A. TDP-43 binds and transports G-quadruplex-containing mRNAs into neurites for local translation. Genes Cells 2016; 21:466-81. [PMID: 26915990 DOI: 10.1111/gtc.12352] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 02/01/2016] [Indexed: 12/14/2022]
Abstract
Growth and differentiation of the neurites depends on long-distance transport of a specific set of mRNAs to restricted area and their local translation. Here, we found that a TAR DNA-binding protein of 43 kDa in size (TDP-43) plays an essential role in intracellular transport of mRNA. For identification of target RNAs recognized by TDP-43, we purified TDP-43 in soluble dimer form and subjected to in vitro systematic evolution of ligands by exponential enrichment (SELEX) screening. All the TDP-43-bound RNAs were found to contain G-quadruplex (G4). Using a double-fluorescent probe system, G4-containing RNAs were found to be transported, together with TDP-43, into the distal neurites. Two lines of evidence indicated that loss of function of TDP-43 results in the neurodegenerative disorder: (i) amyotrophic lateral sclerosis (ALS)-linked mutant TDP-43M337V lacks the activity of binding and transport of G4-containing mRNAs; and (ii) RNA containing G4-forming GGGGCC repeat expansion from the ALS-linked C9orf72 gene absorbs TDP-43, thereby reducing the intracellular pool of functional TDP-43. Taken together, we propose that TDP-43 within neurons plays an essential role of mRNA transport into distal neurites for local translation, and thus, dysfunctions of TDP-43 cause neural diseases such as ALS and frontotemporal lobar degeneration.
Collapse
Affiliation(s)
- Akira Ishiguro
- Research Center for Micro-Nano Technology, University of Hosei, Midori-cho 3-11-15, Koganei, Tokyo, 184-0003, Japan
| | - Nobuyuki Kimura
- Section of Cell Biology and Pathology, Department of Alzheimer's Disease Research, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Morioka 7-430, Obu, Aichi, 474-8511, Japan
| | - Yuto Watanabe
- Department of Biophysics and Biochemistry, Graduate School of Science, The University of Tokyo, Yayoi 2-11-16, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Sumiko Watanabe
- Department of Molecular and Developmental Biology, Institute of Medical Science, The University of Tokyo, Shirokane-dai 4-6-1, Minatoku-ku, Tokyo, 108-8639, Japan
| | - Akira Ishihama
- Research Center for Micro-Nano Technology, University of Hosei, Midori-cho 3-11-15, Koganei, Tokyo, 184-0003, Japan
| |
Collapse
|
217
|
An amyloid-like cascade hypothesis for C9orf72 ALS/FTD. Curr Opin Neurobiol 2016; 36:99-106. [DOI: 10.1016/j.conb.2015.10.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/08/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022]
|
218
|
Chang YJ, Jeng US, Chiang YL, Hwang IS, Chen YR. The Glycine-Alanine Dipeptide Repeat from C9orf72 Hexanucleotide Expansions Forms Toxic Amyloids Possessing Cell-to-Cell Transmission Properties. J Biol Chem 2016; 291:4903-11. [PMID: 26769963 DOI: 10.1074/jbc.m115.694273] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Indexed: 11/06/2022] Open
Abstract
Hexanucleotide expansions, GGGGCC, in the non-coding regions of the C9orf72 gene were found in major frontotemporal lobar dementia and amyotrophic lateral sclerosis patients (C9FTD/ALS). In addition to possible RNA toxicity, several dipeptide repeats (DPRs) are translated through repeat-associated non-ATG-initiated translation. The DPRs, including poly(GA), poly(GR), poly(GP), poly(PR), and poly(PA), were found in the brains and spinal cords of C9FTD/ALS patients. Among the DPRs, poly(GA) is highly susceptible to form cytoplasmic inclusions, which is a characteristic of C9FTD/ALS. To elucidate DPR aggregation, we used synthetic (GA)15 DPR as a model system to examine the aggregation and structural properties in vitro. We found that (GA)15 with 15 repeats fibrillates rapidly and ultimately forms flat, ribbon-type fibrils evidenced by transmission electron microscopy and atomic force microscopy. The fibrils are capable of amyloid dye binding and contain a characteristic cross-β sheet structure, as revealed by x-ray scattering. Furthermore, using neuroblastoma cells, we demonstrated the neurotoxicity and cell-to-cell transmission property of (GA)15 DPR. Overall, our results show the structural and toxicity properties of GA DPR to facilitate future DPR-related therapeutic development.
Collapse
Affiliation(s)
| | - U-Ser Jeng
- the National Synchrotron Radiation Research Center, 101 Hsin-Ann Road, Science Park, Hsinchu 30076, Taiwan, and the Chemical Engineering Department and
| | - Ya-Ling Chiang
- Institute of Physics, Academia Sinica, Academia Road, Section 2, Nankang District, Taipei 115, Taiwan, Department of Material Science and Engineering, National Tsing-Hua University, Hsinchu 30013, Taiwan
| | - Ing-Shouh Hwang
- Institute of Physics, Academia Sinica, Academia Road, Section 2, Nankang District, Taipei 115, Taiwan, Department of Material Science and Engineering, National Tsing-Hua University, Hsinchu 30013, Taiwan
| | | |
Collapse
|
219
|
Livesey MR, Magnani D, Cleary EM, Vasistha NA, James OT, Selvaraj BT, Burr K, Story D, Shaw CE, Kind PC, Hardingham GE, Wyllie DJA, Chandran S. Maturation and electrophysiological properties of human pluripotent stem cell-derived oligodendrocytes. Stem Cells 2016; 34:1040-53. [PMID: 26763608 PMCID: PMC4840312 DOI: 10.1002/stem.2273] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 10/14/2015] [Accepted: 11/05/2015] [Indexed: 12/12/2022]
Abstract
Rodent‐based studies have shown that the membrane properties of oligodendrocytes play prominent roles in their physiology and shift markedly during their maturation from the oligodendrocyte precursor cell (OPC) stage. However, the conservation of these properties and maturation processes in human oligodendrocytes remains unknown, despite their dysfunction being implicated in human neurodegenerative diseases such as multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS). Here, we have defined the membrane properties of human oligodendrocytes derived from pluripotent stem cells as they mature from the OPC stage, and have identified strong conservation of maturation‐specific physiological characteristics reported in rodent systems. We find that as human oligodendrocytes develop and express maturation markers, they exhibit a progressive decrease in voltage‐gated sodium and potassium channels and a loss of tetrodotoxin‐sensitive spiking activity. Concomitant with this is an increase in inwardly rectifying potassium channel activity, as well as a characteristic switch in AMPA receptor composition. All these steps mirror the developmental trajectory observed in rodent systems. Oligodendrocytes derived from mutant C9ORF72‐carryng ALS patient induced pluripotent stem cells did not exhibit impairment to maturation and maintain viability with respect to control lines despite the presence of RNA foci, suggesting that maturation defects may not be a primary feature of this mutation. Thus, we have established that the development of human oligodendroglia membrane properties closely resemble those found in rodent cells and have generated a platform to enable the impact of human neurodegenerative disease‐causing mutations on oligodendrocyte maturation to be studied. Stem Cells2016;34:1040–1053
Collapse
Affiliation(s)
- Matthew R Livesey
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom.,Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Dario Magnani
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.,MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Elaine M Cleary
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.,MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Navneet A Vasistha
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Owain T James
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, Karnataka, India
| | - Bhuvaneish T Selvaraj
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Karen Burr
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David Story
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.,MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Christopher E Shaw
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, United Kingdom
| | - Peter C Kind
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, Karnataka, India
| | - Giles E Hardingham
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - David J A Wyllie
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom.,Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, Karnataka, India
| | - Siddharthan Chandran
- Euan MacDonald Centre for MND Research, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom.,MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, Karnataka, India
| |
Collapse
|
220
|
Almeida MR, Letra L, Pires P, Santos A, Rebelo O, Guerreiro R, van der Zee J, Van Broeckhoven C, Santana I. Characterization of an FTLD-PDB family with the coexistence of SQSTM1 mutation and hexanucleotide (G₄C₂) repeat expansion in C9orf72 gene. Neurobiol Aging 2015; 40:191.e1-191.e8. [PMID: 26839080 DOI: 10.1016/j.neurobiolaging.2015.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/14/2015] [Accepted: 12/23/2015] [Indexed: 12/13/2022]
Abstract
The C9orf72 expansion is considered a major genetic cause of familial frontotemporal dementia (FTD) in several patients' cohorts. Interestingly, C9orf72 expansion carriers, present also abundant neuronal p62-positive inclusions. Although p62/SQSTM1 mutations were initially associated with Paget disease of bone (PDB), they have been also identified in FTD. We describe an FTD-PDB family in which the proband presented with behavioral FTD phenotype and concomitant Paget disease. The molecular genetic analysis revealed the co-occurrence of 2 mutations; the pathogenic C9orf72 expansion and p.P392L heterozygous missense mutation in SQSTM1 gene. Amongst the 6 family members analyzed, the p.P392L SQSTM1 mutation segregated as expected with PDB, whereas the C9orf72 expansion segregated with frontal cognitive impairment or dementia in all but one carrier. The coexistence of these conditions could be underestimated since neither patients with FTD nor patients with PDB undergo bone scintigraphy or cognitive assessment, respectively. The number of cases with double mutations could also be over looked as the molecular strategy adopted in most laboratories ends with the identification of one pathogenic mutation in one of the known causative genes. Therefore, we advocate for further clinical and molecular evaluation in suspect cases.
Collapse
Affiliation(s)
- Maria Rosário Almeida
- CNC-Center for Neuroscience and Cell Biology, Neurogenetics Department, University of Coimbra, Portugal.
| | - Liliana Letra
- Neurology Department, Coimbra University Hospital, Coimbra, Portugal
| | - Paula Pires
- Neurology Department, Hospital de Santo Espírito de Angra do Heroísmo, Azores, Portugal
| | - Ana Santos
- CNC-Center for Neuroscience and Cell Biology, Neurogenetics Department, University of Coimbra, Portugal
| | - Olinda Rebelo
- Neuropathology Laboratory, Neurology Department, Coimbra University Hospital, Portugal
| | - Rita Guerreiro
- Department of Molecular Neuroscience, Institute of Neurology, UCL, London, UK
| | - Julie van der Zee
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium; Institute Born-Bunge, Neurogenetics Department, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, Department of Molecular Genetics, VIB, Antwerp, Belgium; Institute Born-Bunge, Neurogenetics Department, University of Antwerp, Antwerp, Belgium
| | - Isabel Santana
- CNC-Center for Neuroscience and Cell Biology, Neurogenetics Department, University of Coimbra, Portugal; Neurology Department, Coimbra University Hospital, Coimbra, Portugal
| |
Collapse
|
221
|
Whitehouse A, Doherty K, Yeh HH, Robinson AC, Rollinson S, Pickering-Brown S, Snowden J, Thompson JC, Davidson YS, Mann DMA. Histone deacetylases (HDACs) in frontotemporal lobar degeneration. Neuropathol Appl Neurobiol 2015; 41:245-57. [PMID: 24861260 DOI: 10.1111/nan.12153] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 04/15/2014] [Indexed: 11/29/2022]
Abstract
AIMS Frontotemporal lobar degeneration (FTLD) is clinically and pathologically heterogeneous. Although associated with variations in MAPT, GRN and C9ORF72, the pathogenesis of these, and of other nongenetic, forms of FTLD, remains unknown. Epigenetic factors such as histone regulation by histone deacetylases (HDAC) may play a role in the dysregulation of transcriptional activity, thought to underpin the neurodegenerative process. METHODS The distribution and intensity of HDACs 4, 5 and 6 was assessed semi-quantitatively in immunostained sections of temporal cortex with hippocampus, and cerebellum, from 33 pathologically confirmed cases of FTLD and 27 controls. RESULTS We found a significantly greater intensity of cytoplasmic immunostaining for HDAC4 and HDAC6 in granule cells of the dentate gyrus in cases of FTLD overall compared with controls, and specifically in cases of FTLD tau-Picks compared with FTLD tau-MAPT and controls. No differences were noted between FTLD-TDP subtypes, or between the different genetic and nongenetic forms of FTLD. No changes were seen in HDAC5 in any FTLD or control cases. CONCLUSIONS Dysregulation of HDAC4 and/or HDAC6 could play a role in the pathogenesis of FTLD-tau associated with Pick bodies, although their lack of immunostaining implies that such changes do not contribute directly to the formation of Pick bodies.
Collapse
Affiliation(s)
- Amy Whitehouse
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Couratier P, Corcia P, Lautrette G, Nicol M, Preux PM, Marin B. Epidemiology of amyotrophic lateral sclerosis: A review of literature. Rev Neurol (Paris) 2015; 172:37-45. [PMID: 26727307 DOI: 10.1016/j.neurol.2015.11.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/10/2015] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease of motor neurons, resulting in worsening weakness of voluntary muscles until death occurs from respiratory failure. The incidence of ALS in European populations is two to three people per year per 100,000 of the general population. In Europe, crude prevalences range from 1.1/100,000 population in Yugoslavia to 8.2/100,000 in the Faroe Islands. Major advances have been made in our understanding of the genetic causes of ALS, whereas the contribution of environmental factors has been more difficult to assess and large-scale studies have not yet revealed a replicable, definitive environmental risk factor. The only established risk factors to date are older age, male gender and a family history of ALS. Median survival time from onset to death is usually 3 years from the first appearance of symptoms. Older age and bulbar onset are consistently reported to have poorer outcomes. However, there are conflicting data regarding gender, diagnostic delay and El Escorial criteria. The rate of symptom progression has been revealed to be an independent prognostic factor. Psychosocial factors and impaired cognitive function are negatively related to ALS outcome, while nutritional status and respiratory function are also related to ALS prognosis. The effect of enteral nutrition on survival is still unclear, although noninvasive positive pressure ventilation (NIPPV) has been found to improve survival. These findings have relevant implications for the design of future trials.
Collapse
Affiliation(s)
- P Couratier
- Centre de compétence SLA-fédération Tours-Limoges, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France; Inserm UMR1094, neuroépidémiologie tropicale, université de Limoges, 2, rue du Dr.-Marcland, 87025 Limoges cedex, France.
| | - P Corcia
- Centre de compétence SLA-fédération Tours-Limoges, CHU de Tours, 37044 Tours cedex 9, France
| | - G Lautrette
- Centre de compétence SLA-fédération Tours-Limoges, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France
| | - M Nicol
- Centre de compétence SLA-fédération Tours-Limoges, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France
| | - P-M Preux
- Inserm UMR1094, neuroépidémiologie tropicale, université de Limoges, 2, rue du Dr.-Marcland, 87025 Limoges cedex, France
| | - B Marin
- Inserm UMR1094, neuroépidémiologie tropicale, université de Limoges, 2, rue du Dr.-Marcland, 87025 Limoges cedex, France
| |
Collapse
|
223
|
Haase G, Rabouille C. Golgi Fragmentation in ALS Motor Neurons. New Mechanisms Targeting Microtubules, Tethers, and Transport Vesicles. Front Neurosci 2015; 9:448. [PMID: 26696811 PMCID: PMC4672084 DOI: 10.3389/fnins.2015.00448] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/13/2015] [Indexed: 12/12/2022] Open
Abstract
Pathological alterations of the Golgi apparatus, such as its fragmentation represent an early pre-clinical feature of many neurodegenerative diseases and have been widely studied in the motor neuron disease amyotrophic lateral sclerosis (ALS). Yet, the underlying molecular mechanisms have remained cryptic. In principle, Golgi fragmentation may result from defects in three major classes of proteins: structural Golgi proteins, cytoskeletal proteins and molecular motors, as well as proteins mediating transport to and through the Golgi. Here, we present the different mechanisms that may underlie Golgi fragmentation in animal and cellular models of ALS linked to mutations in SOD1, TARDBP (TDP-43), VAPB, and C9Orf72 and we propose a novel one based on findings in progressive motor neuronopathy (pmn) mice. These mice are mutated in the TBCE gene encoding the cis-Golgi localized tubulin-binding cofactor E, one of five chaperones that assist in tubulin folding and microtubule polymerization. Loss of TBCE leads to alterations in Golgi microtubules, which in turn impedes on the maintenance of the Golgi architecture. This is due to down-regulation of COPI coat components, dispersion of Golgi tethers and strong accumulation of ER-Golgi SNAREs. These effects are partially rescued by the GTPase ARF1 through recruitment of TBCE to the Golgi. We hypothesize that defects in COPI vesicles, microtubules and their interaction may also underlie Golgi fragmentation in human ALS linked to other mutations, spinal muscular atrophy (SMA), and related motor neuron diseases. We also discuss the functional relevance of pathological Golgi alterations, in particular their potential causative, contributory, or compensatory role in the degeneration of motor neuron cell bodies, axons and synapses.
Collapse
Affiliation(s)
- Georg Haase
- Centre National de la Recherche Scientifique and Aix-Marseille Université UMR 7289, Institut de Neurosciences de la Timone Marseille, France
| | - Catherine Rabouille
- The Department of Cell Biology, Hubrecht Institute of the Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
224
|
Corcia P, Vourc'h P, Guennoc AM, Del Mar Amador M, Blasco H, Andres C, Couratier P, Gordon PH, Meininger V. Pure cerebellar ataxia linked to large C9orf72 repeat expansion. Amyotroph Lateral Scler Frontotemporal Degener 2015; 17:301-3. [PMID: 26609732 DOI: 10.3109/21678421.2015.1113298] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Philippe Corcia
- a Centre SLA , CHU Tours .,b UMR INSERM U930 , Université François-Rabelais de Tours , Tours
| | - Patrick Vourc'h
- b UMR INSERM U930 , Université François-Rabelais de Tours , Tours .,c Service de Biochimie et Biologie Moléculaire , CHRU de Tours , Tours
| | | | - Maria Del Mar Amador
- f Fédération des Maladies du Système Nerveux , Hôpital de la Salpêtrière , Paris , France
| | - Hélène Blasco
- b UMR INSERM U930 , Université François-Rabelais de Tours , Tours .,c Service de Biochimie et Biologie Moléculaire , CHRU de Tours , Tours
| | - Christian Andres
- b UMR INSERM U930 , Université François-Rabelais de Tours , Tours .,c Service de Biochimie et Biologie Moléculaire , CHRU de Tours , Tours
| | | | - Paul H Gordon
- e Northern Navajo Medical Center , Shiprock , NM , USA , and
| | | |
Collapse
|
225
|
Walker AK, Tripathy K, Restrepo CR, Ge G, Xu Y, Kwong LK, Trojanowski JQ, Lee VMY. An insoluble frontotemporal lobar degeneration-associated TDP-43 C-terminal fragment causes neurodegeneration and hippocampus pathology in transgenic mice. Hum Mol Genet 2015; 24:7241-54. [PMID: 26476406 PMCID: PMC5007600 DOI: 10.1093/hmg/ddv424] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/05/2015] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) causes progressive personality, behavior and/or language disturbances and represents the second most common form of dementia under the age of 65. Over half of all FTD cases are classified pathologically as frontotemporal lobar degeneration (FTLD) with TAR DNA-binding protein of 43 kDa (TDP-43) pathology (FTLD-TDP). In FTLD-TDP brains, TDP-43 is phosphorylated, C-terminally cleaved, lost from the nucleus and accumulates in the cytoplasm and processes of neurons and glia. However, the contribution of TDP-43 C-terminal fragments (CTFs) to pathogenesis remains poorly understood. Here, we developed transgenic (Tg) mice with forebrain Camk2a-controlled doxycycline-suppressible expression of a TDP-43 CTF (amino acids 208-414, designated 208 TDP-43 CTF), previously identified in FTLD-TDP brains. In these 208 TDP-43 Tg mice, detergent-insoluble 208 TDP-43 CTF was present in a diffuse punctate pattern in neuronal cytoplasm and dendrites without forming large cytoplasmic inclusions. Remarkably, the hippocampus showed progressive neuron loss and astrogliosis in the dentate gyrus (DG). This was accompanied by phosphorylated TDP-43 in the CA1 subfield, and ubiquitin and mitochondria accumulations in the stratum lacunosum moleculare (SLM) layer, without loss of endogenous nuclear TDP-43. Importantly, 208 TDP-43 CTF and phosphorylated TDP-43 were rapidly cleared when CTF expression was suppressed in aged Tg mice, which ameliorated neuron loss in the DG despite persistence of ubiquitin accumulation in the SLM. Our results demonstrate that Camk2a-directed 208 TDP-43 CTF overexpression is sufficient to cause hippocampal pathology and neurodegeneration in vivo, suggesting an active role for TDP-43 CTFs in the pathogenesis of FTLD-TDP and related TDP-43 proteinopathies.
Collapse
Affiliation(s)
- Adam K Walker
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research and
| | - Kalyan Tripathy
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research and
| | - Clark R Restrepo
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research and
| | - Guanghui Ge
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research and
| | - Yan Xu
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research and
| | - Linda K Kwong
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research and
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research and Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research and Institute on Aging, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
226
|
King A, Troakes C, Smith B, Nolan M, Curran O, Vance C, Shaw CE, Al-Sarraj S. ALS-FUS pathology revisited: singleton FUS mutations and an unusual case with both a FUS and TARDBP mutation. Acta Neuropathol Commun 2015; 3:62. [PMID: 26452761 PMCID: PMC4600255 DOI: 10.1186/s40478-015-0235-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 08/25/2015] [Indexed: 12/12/2022] Open
Abstract
Introduction Mutations in the FUS gene have been shown to be a rare cause of amyotrophic lateral sclerosis (ALS-FUS) and whilst well documented clinically and genetically there have been relatively few neuropathological studies.Recent work suggested a possible correlation between pathological features such as frequency of basophilic inclusions in neurons and rate of clinical decline, other studies have revealed a discrepancy between the upper motor neuron features detected clinically and the associated pathology. The purpose of this study was to describe the pathological features associated with more recently discovered FUS mutations and reinvestigate those with well recognised mutations in an attempt to correlate the pathology with mutation and/or clinical phenotype. The brains and spinal cords of seven cases of ALS-FUS were examined neuropathologically, including cases with the newly described p.K510E mutation and a case with both a known p.P525L mutation in the FUS gene and a truncating p.Y374X mutation in the TARDBP gene. Results The neuropathology in all cases revealed basophilic and FUS inclusions in the cord. The density and type of inclusions varied markedly between cases, but did not allow a clear correlation with clinical progression. Only one case showed significant motor cortical pathology despite the upper motor neuron clinical features being evident in 4 patients. The case with both a FUS and TARDBP mutation revealed FUS positive inclusions but no TDP-43 pathology. Instead there were unusual p62 positive, FUS negative neuronal and glial inclusions as well as dot-like neurites. Conclusions The study confirms cases of ALS-FUS to be mainly a lower motor neuron disease and to have pathology that does not appear to neatly correlate with clinical features or genetics. Furthermore, the case with both a FUS and TARDBP mutation reveals an intriguing pathological profile which at least in part involves a very unusual staining pattern for the ubiquitin-binding protein p62.
Collapse
|
227
|
Collins MA, An J, Hood BL, Conrads TP, Bowser RP. Label-Free LC-MS/MS Proteomic Analysis of Cerebrospinal Fluid Identifies Protein/Pathway Alterations and Candidate Biomarkers for Amyotrophic Lateral Sclerosis. J Proteome Res 2015; 14:4486-501. [PMID: 26401960 DOI: 10.1021/acs.jproteome.5b00804] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Analysis of the cerebrospinal fluid (CSF) proteome has proven valuable to the study of neurodegenerative disorders. To identify new protein/pathway alterations and candidate biomarkers for amyotrophic lateral sclerosis (ALS), we performed comparative proteomic profiling of CSF from sporadic ALS (sALS), healthy control (HC), and other neurological disease (OND) subjects using label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS). A total of 1712 CSF proteins were detected and relatively quantified by spectral counting. Levels of several proteins with diverse biological functions were significantly altered in sALS samples. Enrichment analysis was used to link these alterations to biological pathways, which were predominantly related to inflammation, neuronal activity, and extracellular matrix regulation. We then used our CSF proteomic profiles to create a support vector machines classifier capable of discriminating training set ALS from non-ALS (HC and OND) samples. Four classifier proteins, WD repeat-containing protein 63, amyloid-like protein 1, SPARC-like protein 1, and cell adhesion molecule 3, were identified by feature selection and externally validated. The resultant classifier distinguished ALS from non-ALS samples with 83% sensitivity and 100% specificity in an independent test set. Collectively, our results illustrate the utility of CSF proteomic profiling for identifying ALS protein/pathway alterations and candidate disease biomarkers.
Collapse
Affiliation(s)
- Mahlon A Collins
- Department of Neurobiology, University of Pittsburgh , E1448 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, Pennsylvania 15261, United States.,Departments of Neurology and Neurobiology, Barrow Neurological Institute , NRC427, 350 West Thomas Road, Phoenix, Arizona 85013, United States
| | - Jiyan An
- Departments of Neurology and Neurobiology, Barrow Neurological Institute , NRC427, 350 West Thomas Road, Phoenix, Arizona 85013, United States
| | - Brian L Hood
- Women's Health Integrated Research Center , 3289 Woodburn Road, Annandale, Virginia 22003, United States
| | - Thomas P Conrads
- Women's Health Integrated Research Center , 3289 Woodburn Road, Annandale, Virginia 22003, United States
| | - Robert P Bowser
- Departments of Neurology and Neurobiology, Barrow Neurological Institute , NRC427, 350 West Thomas Road, Phoenix, Arizona 85013, United States
| |
Collapse
|
228
|
Gendron TF, van Blitterswijk M, Bieniek KF, Daughrity LM, Jiang J, Rush BK, Pedraza O, Lucas JA, Murray ME, Desaro P, Robertson A, Overstreet K, Thomas CS, Crook JE, Castanedes-Casey M, Rousseau L, Josephs KA, Parisi JE, Knopman DS, Petersen RC, Boeve BF, Graff-Radford NR, Rademakers R, Lagier-Tourenne C, Edbauer D, Cleveland DW, Dickson DW, Petrucelli L, Boylan KB. Cerebellar c9RAN proteins associate with clinical and neuropathological characteristics of C9ORF72 repeat expansion carriers. Acta Neuropathol 2015; 130:559-73. [PMID: 26350237 PMCID: PMC4575385 DOI: 10.1007/s00401-015-1474-4] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/29/2015] [Accepted: 08/30/2015] [Indexed: 12/12/2022]
Abstract
Clinical and neuropathological characteristics associated with G4C2 repeat expansions in chromosome 9 open reading frame 72 (C9ORF72), the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia, are highly variable. To gain insight on the molecular basis for the heterogeneity among C9ORF72 mutation carriers, we evaluated associations between features of disease and levels of two abundantly expressed “c9RAN proteins” produced by repeat-associated non-ATG (RAN) translation of the expanded repeat. For these studies, we took a departure from traditional immunohistochemical approaches and instead employed immunoassays to quantitatively measure poly(GP) and poly(GA) levels in cerebellum, frontal cortex, motor cortex, and/or hippocampus from 55 C9ORF72 mutation carriers [12 patients with ALS, 24 with frontotemporal lobar degeneration (FTLD) and 19 with FTLD with motor neuron disease (FTLD-MND)]. We additionally investigated associations between levels of poly(GP) or poly(GA) and cognitive impairment in 15 C9ORF72 ALS patients for whom neuropsychological data were available. Among the neuroanatomical regions investigated, poly(GP) levels were highest in the cerebellum. In this same region, associations between poly(GP) and both neuropathological and clinical features were detected. Specifically, cerebellar poly(GP) levels were significantly lower in patients with ALS compared to patients with FTLD or FTLD-MND. Furthermore, cerebellar poly(GP) associated with cognitive score in our cohort of 15 patients. In the cerebellum, poly(GA) levels similarly trended lower in the ALS subgroup compared to FTLD or FTLD-MND subgroups, but no association between cerebellar poly(GA) and cognitive score was detected. Both cerebellar poly(GP) and poly(GA) associated with C9ORF72 variant 3 mRNA expression, but not variant 1 expression, repeat size, disease onset, or survival after onset. Overall, these data indicate that cerebellar abnormalities, as evidenced by poly(GP) accumulation, associate with neuropathological and clinical phenotypes, in particular cognitive impairment, of C9ORF72 mutation carriers.
Collapse
Affiliation(s)
- Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Kevin F Bieniek
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Mayo Graduate School, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Jie Jiang
- Ludwig Institute, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Beth K Rush
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Otto Pedraza
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - John A Lucas
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Melissa E Murray
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Pamela Desaro
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Amelia Robertson
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Karen Overstreet
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Colleen S Thomas
- Section of Biostatistics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Julia E Crook
- Section of Biostatistics, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Linda Rousseau
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Keith A Josephs
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Joseph E Parisi
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Bradley F Boeve
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Rosa Rademakers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Clotilde Lagier-Tourenne
- Ludwig Institute, University of California at San Diego, La Jolla, CA, 92093, USA
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Metabolic Biochemistry, Ludwig-Maximilians University Munich, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Don W Cleveland
- Ludwig Institute, University of California at San Diego, La Jolla, CA, 92093, USA
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Kevin B Boylan
- Department of Neurology, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
229
|
Baldeiras I, Santana I, Leitão MJ, Ribeiro MH, Pascoal R, Duro D, Lemos R, Santiago B, Almeida MR, Oliveira CR. Cerebrospinal fluid Aβ40 is similarly reduced in patients with Frontotemporal Lobar Degeneration and Alzheimer's Disease. J Neurol Sci 2015; 358:308-16. [PMID: 26388316 DOI: 10.1016/j.jns.2015.09.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/11/2015] [Accepted: 09/08/2015] [Indexed: 12/12/2022]
Abstract
Cerebrospinal fluid (CSF) biomarkers have been increasingly studied for dementia diagnosis, however the accuracy to distinguish between different forms of dementia is still unsatisfactory. In this study, the added value of another CSF Aβ-peptide (Aβ40), along with the core CSF markers t-Tau, p-Tau, and Aβ42, in the discrimination between two large dementia groups of Frontotemporal Lobar Degeneration (FTLD; n=107), Alzheimer's Disease (AD; n=107) and non-demented subjects (n=33) was evaluated. In FTLD, t-Tau and p-Tau were significantly increased in relation to controls, but lower than in AD, while Aβ42 was similar in FTLD and controls, but higher than in AD. Equally reduced Aβ40 levels were seen in both dementia groups, and therefore the combination of Aβ40 with core CSF biomarkers optimally discriminated FTLD and AD patients from controls. Aβ42 and t-Tau were selected as the best biomarker subset to differentiate FTLD from AD, with no added value of Aβ40 to the model. Diagnostic accuracy between FTLD and AD was still sub-optimal, with a significant percentage (23%) of FTLD patients, in particularly women, carrying an ApoE-ε4 allele, showing a CSF-AD biomarkers profile. Although CSF Aβ40 does not appear to have an additional value in the distinction between FTLD and AD, it increases the discrimination between subjects with dementia from controls. A CSF-AD biomarker profile can be seen in patients with a clinical phenotype of FTLD, reinforcing the need for autopsy confirmation.
Collapse
Affiliation(s)
- Inês Baldeiras
- Laboratory of Neurochemistry, Coimbra University Hospital, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Portugal.
| | - Isabel Santana
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Portugal; Neurology Department, Coimbra University Hospital, Portugal
| | - Maria João Leitão
- Laboratory of Neurochemistry, Coimbra University Hospital, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Maria Helena Ribeiro
- Laboratory of Neurochemistry, Coimbra University Hospital, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Portugal
| | - Rui Pascoal
- Laboratory of Neurochemistry, Coimbra University Hospital, Portugal
| | - Diana Duro
- Neurology Department, Coimbra University Hospital, Portugal
| | - Raquel Lemos
- Neurology Department, Coimbra University Hospital, Portugal
| | | | | | - Catarina Resende Oliveira
- Laboratory of Neurochemistry, Coimbra University Hospital, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Portugal
| |
Collapse
|
230
|
Benussi A, Padovani A, Borroni B. Phenotypic Heterogeneity of Monogenic Frontotemporal Dementia. Front Aging Neurosci 2015; 7:171. [PMID: 26388768 PMCID: PMC4555036 DOI: 10.3389/fnagi.2015.00171] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/19/2015] [Indexed: 12/12/2022] Open
Abstract
Frontotemporal dementia (FTD) is a genetically and pathologically heterogeneous disorder characterized by personality changes, language impairment, and deficits of executive functions associated with frontal and temporal lobe degeneration. Different phenotypes have been defined on the basis of presenting clinical symptoms, i.e., the behavioral variant of FTD, the agrammatic variant of primary progressive aphasia, and the semantic variant of PPA. Some patients have an associated movement disorder, either parkinsonism, as in progressive supranuclear palsy and corticobasal syndrome, or motor neuron disease (FTD-MND). A family history of dementia is found in 40% of cases of FTD and about 10% have a clear autosomal-dominant inheritance. Genetic studies have identified several genes associated with monogenic FTD: microtubule-associated protein tau, progranulin, TAR DNA-binding protein 43, valosin-containing protein, charged multivesicular body protein 2B, fused in sarcoma, and the hexanucleotide repeat expansion in intron 1 of the chromosome 9 open reading frame 72. Patients often present with an extensive phenotypic variability, even among different members of the same kindred carrying an identical disease mutation. The objective of the present work is to review and evaluate available literature data in order to highlight recent advances in clinical, biological, and neuroimaging features of monogenic frontotemporal lobar degeneration and try to identify different mechanisms underlying the extreme phenotypic heterogeneity that characterizes this disease.
Collapse
Affiliation(s)
- Alberto Benussi
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Barbara Borroni
- Centre for Ageing Brain and Neurodegenerative Disorders, Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
231
|
Bury JJ, Highley JR, Cooper-Knock J, Goodall EF, Higginbottom A, McDermott CJ, Ince PG, Shaw PJ, Kirby J. Oligogenic inheritance of optineurin (OPTN) and C9ORF72 mutations in ALS highlights localisation of OPTN in the TDP-43-negative inclusions of C9ORF72-ALS. Neuropathology 2015; 36:125-34. [PMID: 26303227 DOI: 10.1111/neup.12240] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 07/24/2015] [Accepted: 07/25/2015] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by motor neurone loss resulting in muscle weakness, spasticity and ultimately death. 5-10% are caused by inherited mutations, most commonly C9ORF72, SOD1, TARDBP and FUS. Rarer genetic causes of ALS include mutation of optineurin (mt OPTN). Furthermore, optineurin protein has been localized to the ubiquitylated aggregates in several neurodegenerative diseases, including ALS. This study: (i) investigated the frequency of mt OPTN in ALS patients in England; (ii) characterized the clinical and neuropathological features of ALS associated with a mt OPTN; and (iii) investigated optineurin neuropathology in C9ORF72-related ALS (C9ORF72-ALS). We identified a heterozygous p.E322K missense mutation in exon 10 of OPTN in one familial ALS patient who additionally had a C9ORF72 mutation. This patient had bulbar, limb and respiratory disease without cognitive problems. Neuropathology revealed motor neurone loss, trans-activation response DNA protein 43 (TDP-43)-positive neuronal and glial cytoplasmic inclusions together with TDP-43-negative neuronal cytoplasmic inclusions in extra motor regions that are characteristic of C9ORF72-ALS. We have demonstrated that both TDP-43-positive and negative inclusion types had positive staining for optineurin by immunohistochemistry. We went on to show that optineurin was present in TDP-43-negative cytoplasmic extra motor inclusions in C9ORF72-ALS cases that do not carry mt OPTN. We conclude that: (i) OPTN mutations are associated with ALS; (ii) optineurin protein is present in a subset of the extramotor inclusions of C9ORF72-ALS; (iii) It is not uncommon for multiple ALS-causing mutations to occur in the same patient; and (iv) studies of optineurin are likely to provide useful dataregarding the pathophysiology of ALS and neurodegeneration.
Collapse
Affiliation(s)
- Joanna J Bury
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - J Robin Highley
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Emily F Goodall
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Adrian Higginbottom
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Christopher J McDermott
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, Sheffield, UK
| |
Collapse
|
232
|
Highley JR, Lorente Pons A, Cooper-Knock J, Wharton SB, Ince PG, Shaw PJ, Wood J, Kirby J. Motor neurone disease/amyotrophic lateral sclerosis associated with intermediate-length CAG repeat expansions inAtaxin-2does not have 1C2-positive polyglutamine inclusions. Neuropathol Appl Neurobiol 2015; 42:377-89. [DOI: 10.1111/nan.12254] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/14/2015] [Indexed: 12/13/2022]
Affiliation(s)
- John Robin Highley
- Sheffield Institute for Translational Neuroscience (SITraN); University of Sheffield; Sheffield UK
| | - Alejandro Lorente Pons
- Sheffield Institute for Translational Neuroscience (SITraN); University of Sheffield; Sheffield UK
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN); University of Sheffield; Sheffield UK
| | - Stephen B. Wharton
- Sheffield Institute for Translational Neuroscience (SITraN); University of Sheffield; Sheffield UK
| | - Paul G. Ince
- Sheffield Institute for Translational Neuroscience (SITraN); University of Sheffield; Sheffield UK
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN); University of Sheffield; Sheffield UK
| | - Jon Wood
- Sheffield Institute for Translational Neuroscience (SITraN); University of Sheffield; Sheffield UK
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience (SITraN); University of Sheffield; Sheffield UK
| |
Collapse
|
233
|
Lashley T, Rohrer JD, Mead S, Revesz T. Review: An update on clinical, genetic and pathological aspects of frontotemporal lobar degenerations. Neuropathol Appl Neurobiol 2015; 41:858-81. [DOI: 10.1111/nan.12250] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/27/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Tammaryn Lashley
- Queen Square Brain Bank for Neurological Disorders; Department of Molecular Neuroscience; UCL Institute of Neurology; London UK
| | | | - Simon Mead
- Department of Neurodegenerative Disease; UCL Institute of Neurology; London UK
| | - Tamas Revesz
- Queen Square Brain Bank for Neurological Disorders; Department of Molecular Neuroscience; UCL Institute of Neurology; London UK
| |
Collapse
|
234
|
Pottier C, Bieniek KF, Finch N, van de Vorst M, Baker M, Perkersen R, Brown P, Ravenscroft T, van Blitterswijk M, Nicholson AM, DeTure M, Knopman DS, Josephs KA, Parisi JE, Petersen RC, Boylan KB, Boeve BF, Graff-Radford NR, Veltman JA, Gilissen C, Murray ME, Dickson DW, Rademakers R. Whole-genome sequencing reveals important role for TBK1 and OPTN mutations in frontotemporal lobar degeneration without motor neuron disease. Acta Neuropathol 2015; 130:77-92. [PMID: 25943890 DOI: 10.1007/s00401-015-1436-x] [Citation(s) in RCA: 233] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 04/27/2015] [Accepted: 04/27/2015] [Indexed: 12/11/2022]
Abstract
Frontotemporal lobar degeneration with TAR DNA-binding protein 43 inclusions (FTLD-TDP) is the most common pathology associated with frontotemporal dementia (FTD). Repeat expansions in chromosome 9 open reading frame 72 (C9ORF72) and mutations in progranulin (GRN) are the major known genetic causes of FTLD-TDP; however, the genetic etiology in the majority of FTLD-TDP remains unexplained. In this study, we performed whole-genome sequencing in 104 pathologically confirmed FTLD-TDP patients from the Mayo Clinic brain bank negative for C9ORF72 and GRN mutations and report on the contribution of rare single nucleotide and copy number variants in 21 known neurodegenerative disease genes. Interestingly, we identified 5 patients (4.8 %) with variants in optineurin (OPTN) and TANK-binding kinase 1 (TBK1) that are predicted to be highly pathogenic, including two double mutants. Case A was a compound heterozygote for mutations in OPTN, carrying the p.Q235* nonsense and p.A481V missense mutation in trans, while case B carried a deletion of OPTN exons 13-15 (p.Gly538Glufs*27) and a loss-of-function mutation (p.Arg117*) in TBK1. Cases C-E carried heterozygous missense mutations in TBK1, including the p.Glu696Lys mutation which was previously reported in two amyotrophic lateral sclerosis (ALS) patients and is located in the OPTN binding domain. Quantitative mRNA expression and protein analysis in cerebellar tissue showed a striking reduction of OPTN and/or TBK1 expression in 4 out of 5 patients supporting pathogenicity in these specific patients and suggesting a loss-of-function disease mechanism. Importantly, neuropathologic examination showed FTLD-TDP type A in the absence of motor neuron disease in 3 pathogenic mutation carriers. In conclusion, we highlight TBK1 as an important cause of pure FTLD-TDP, identify the first OPTN mutations in FTLD-TDP, and suggest a potential oligogenic basis for at least a subset of FTLD-TDP patients. Our data further add to the growing body of evidence linking ALS and FTD and suggest a key role for the OPTN/TBK1 pathway in these diseases.
Collapse
Affiliation(s)
- Cyril Pottier
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Abstract
Amyotrophic lateral sclerosis (ALS) is now recognised to be a heterogeneous neurodegenerative syndrome of the motor system and its frontotemporal cortical connections. The development and application of structural and functional imaging over the last three decades, in particular magnetic resonance imaging (MRI), has allowed traditional post mortem histopathological and emerging molecular findings in ALS to be placed in a clinical context. Cerebral grey and white matter structural MRI changes are increasingly being understood in terms of brain connectivity, providing insights into the advancing degenerative process and producing candidate biomarkers. Such markers may refine the prognostic stratification of patients and the diagnostic pathway, as well as providing an objective assessment of changes in disease activity in response to future therapeutic agents. Studies are being extended to the spinal cord, and the application of neuroimaging to unaffected carriers of highly penetrant genetic mutations linked to the development of ALS offers a unique window to the pre-symptomatic landscape.
Collapse
Affiliation(s)
- Martin R. Turner
- />Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- />John Radcliffe Hospital, West Wing Level 3, Oxford, OX3 9DU UK
| | - Esther Verstraete
- />University of Utrecht, Utrecht, Netherlands
- />University Medical Center, Heidelberglaan 100, Utrecht, Netherlands
| |
Collapse
|
236
|
Kao PF, Chen YR, Liu XB, DeCarli C, Seeley WW, Jin LW. Detection of TDP-43 oligomers in frontotemporal lobar degeneration-TDP. Ann Neurol 2015; 78:211-21. [PMID: 25921485 DOI: 10.1002/ana.24431] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 04/22/2015] [Accepted: 04/22/2015] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The proteinaceous inclusions in TDP-43 proteinopathies such as frontotemporal lobar degeneration (FTLD)-TDP are made of high-molecular-weight aggregates of TDP-43. These aggregates have not been classified as amyloids, as prior amyloid staining results were not conclusive. Here we used a specific TDP-43 amyloid oligomer antibody called TDP-O to determine the presence and abundance of TDP-43 oligomers among different subtypes of FTLD-TDP as well as in hippocampal sclerosis (HS), which represents a non-FTLD pathology with TDP-43 inclusions. METHODS Postmortem tissue from the hippocampus and anterior orbital gyrus from 54 prospectively assessed and diagnosed subjects was used for immunostaining with TDP-O. Electron microscopy was used to assess the subcellular locations of TDP-O-decorated structures. RESULTS TDP-43 inclusions staining with TDP-O were present in FTLD-TDP and were most conspicuous for FTLD-TDP type C, the subtype seen in most patients with semantic variant primary progressive aphasia. TDP-O immunoreactivity was absent in the hippocampus of HS patients despite abundant TDP-43 inclusions. Ultrastructurally, TDP-43 oligomers resided in granular or tubular structures, frequently in close proximity to, but not within, neuronal lysosomes. INTERPRETATION TDP-43 forms amyloid oligomers in the human brain, which may cause neurotoxicity in a manner similar to other amyloid oligomers. Oligomer formation may contribute to the conformational heterogeneity of TDP-43 aggregates and mark the different properties of TDP-43 inclusions between FTLD-TDP and HS.
Collapse
Affiliation(s)
- Patricia F Kao
- Department of Pathology and Laboratory Medicine, University of California, Davis, School of Medicine, Sacramento, CA.,Alzheimer's Disease Center, University of California, Davis, School of Medicine, Sacramento, CA
| | - Yun-Ru Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Xiao-Bo Liu
- Department of Pathology and Laboratory Medicine, University of California, Davis, School of Medicine, Sacramento, CA
| | - Charles DeCarli
- Alzheimer's Disease Center, University of California, Davis, School of Medicine, Sacramento, CA.,Department of Neurology, University of California, Davis, School of Medicine, Sacramento, CA
| | - William W Seeley
- Departments of Neurology and Pathology, University of California, San Francisco, San Francisco, CA
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, University of California, Davis, School of Medicine, Sacramento, CA.,Alzheimer's Disease Center, University of California, Davis, School of Medicine, Sacramento, CA
| |
Collapse
|
237
|
Gomez-Deza J, Lee YB, Troakes C, Nolan M, Al-Sarraj S, Gallo JM, Shaw CE. Dipeptide repeat protein inclusions are rare in the spinal cord and almost absent from motor neurons in C9ORF72 mutant amyotrophic lateral sclerosis and are unlikely to cause their degeneration. Acta Neuropathol Commun 2015; 3:38. [PMID: 26108573 PMCID: PMC4479315 DOI: 10.1186/s40478-015-0218-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/12/2015] [Indexed: 12/14/2022] Open
Abstract
Introduction Cytoplasmic TDP-43 inclusions are the pathological hallmark of amyotrophic lateral sclerosis (ALS) and tau-negative frontotemporal lobar dementia (FTLD). The G4C2 repeat mutation in C9ORF72 is the most common cause of ALS and FTLD in which, in addition to TDP-43 inclusions, five different di-peptide repeat (DPR) proteins have been identified. Di-peptide repeat proteins are translated in a non-canonical fashion from sense and antisense transcripts of the G4C2 repeat (GP, GA, GR, PA, PR). DPR inclusions are abundant in the cerebellum, as well as in the frontal and temporal lobes of ALS and FTLD patients and some are neurotoxic in a range of cellular and animal models, implying that DPR aggregation directly contributes to disease pathogenesis. Here we sought to quantify inclusions for each DPR and TDP-43 in ALS cases with and without the C9ORF72 mutation. We characterised the abundance of DPRs and their cellular location and compared this to cytoplasmic TDP-43 inclusions in order to explore the role of each inclusion in lower motor neuron degeneration. Results Spinal cord sections from ten cases positive for the C9ORF72 repeat expansion (ALS-C9+ve) and five cases that were not were probed by double immunofluorescence staining for individual DPRs and TDP-43. Inclusions immunoreactive for each of the DPRs were present in the spinal cord but they were rare or very rare in abundance (in descending order of frequency: GA, GP, GR, PA and PR). TDP-43 cytoplasmic inclusions were 45- to 750-fold more frequent than any DPR, and fewer than 4 % of DPR inclusions colocalized with TDP-43 inclusions. In motor neurons, a single cytoplasmic DPR inclusion was detected (0.1 %) in contrast to the 34 % of motor neurons that contained cytoplasmic TDP-43 inclusions. Furthermore, the number of TDP-43 inclusions in ALS cases with and without the C9ORF72 mutation was nearly identical. Conclusions For all other neurodegenerative diseases, the neurotoxic protein aggregates are detected in the affected population of neurons. TDP-43 cytoplasmic aggregation is the dominant feature of ALS spinal cord pathology irrespective of C9ORF72 mutation status. The near absence of DPR inclusions in spinal cord motor neurons challenges their contribution to lower motor neuron degeneration in ALS-C9+ve cases. Electronic supplementary material The online version of this article (doi:10.1186/s40478-015-0218-y) contains supplementary material, which is available to authorized users.
Collapse
|
238
|
Drosha inclusions are new components of dipeptide-repeat protein aggregates in FTLD-TDP and ALS C9orf72 expansion cases. J Neuropathol Exp Neurol 2015; 74:380-7. [PMID: 25756586 PMCID: PMC4362478 DOI: 10.1097/nen.0000000000000182] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Supplemental digital content is available in the text. Frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS) are 2 neurodegenerative disorders that share clinical, genetic, and neuropathologic features. The presence of abnormal expansions of GGGGCC repeats (G4C2 repeats) in a noncoding region of the Chromosome 9 open reading frame 72 (C9orf72) gene is the major genetic cause of both FTLD and ALS. Transcribed G4C2 repeats can form nuclear RNA foci and recruit RNA-binding proteins, thereby inhibiting their normal function. Moreover, through a repeat-associated non-ATG translation mechanism, G4C2 repeats translation leads to dipeptide-repeat protein aggregation in the cytoplasm of neurons. Here, we identify Drosha protein as a new component of these dipeptide-repeat aggregates. In C9orf72 mutation cases of FTLD-TDP (c9FTLD-TDP) and ALS (c9ALS), but not in FTLD or ALS cases without C9orf72 mutation, Drosha is mislocalized to form neuronal cytoplasmic inclusions in the hippocampus, frontal cortex, and cerebellum. Further characterization of Drosha-positive neuronal cytoplasmic inclusions in the hippocampus, frontal cortex, and cerebellum revealed colocalization with p62 and ubiquilin-2, 2 pathognomonic signatures of c9FTLD-TDP and c9ALS cases; however, Drosha inclusions rarely colocalized with TDP-43 pathology. We conclude that Drosha may play a unique pathogenic role in the onset or progression of FTLD-TDP/ALS in patients with the C9orf72 mutation.
Collapse
|
239
|
Kitano S, Kino Y, Yamamoto Y, Takitani M, Miyoshi J, Ishida T, Saito Y, Arima K, Satoh JI. Bioinformatics Data Mining Approach Suggests Coexpression of AGTPBP1 with an ALS-linked Gene C9orf72. J Cent Nerv Syst Dis 2015; 7:15-26. [PMID: 26106267 PMCID: PMC4467204 DOI: 10.4137/jcnsd.s24317] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/10/2015] [Accepted: 05/12/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Expanded GGGGCC hexanucleotide repeats located in the noncoding region of the chromosome 9 open reading frame 72 (C9orf72) gene represent the most common genetic abnormality for familial and sporadic amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Formation of nuclear RNA foci, accumulation of repeat-associated non-ATG-translated dipeptide-repeat proteins, and haploinsufficiency of C9orf72 are proposed for pathological mechanisms of C9ALS/FTD. However, at present, the physiological function of C9orf72 remains largely unknown. METHODS By searching on a bioinformatics database named COXPRESdb composed of the comprehensive gene coexpression data, we studied potential C9orf72 interactors. RESULTS We identified the ATP/GTP binding protein 1 (AGTPBP1) gene alternatively named NNA1 encoding a cytosolic carboxypeptidase whose mutation is causative of the degeneration of Purkinje cells and motor neurons as the most significant gene coexpressed with C9orf72. We verified coexpression and interaction of AGTPBP1 and C9orf72 in transfected cells by immunoprecipitation and in neurons of the human brain by double-labeling immunohistochemistry. Furthermore, we found a positive correlation between AGTPBP1 and C9orf72 mRNA expression levels in the set of 21 human brains examined. CONCLUSIONS These results suggest that AGTPBP1 serves as a C9orf72 interacting partner that plays a role in the regulation of neuronal function in a coordinated manner within the central nervous system.
Collapse
Affiliation(s)
- Shouta Kitano
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Yoshihiro Kino
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Yoji Yamamoto
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Mika Takitani
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Junko Miyoshi
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Tsuyoshi Ishida
- Department of Pathology and Laboratory Medicine, Kohnodai Hospital, NCGM, Ichikawa, Chiba, Japan
| | - Yuko Saito
- Department of Laboratory Medicine, National Center Hospital, NCNP, Kodaira, Tokyo, Japan
| | - Kunimasa Arima
- Department of Psychiatry, Komoro Kogen Hospital, Komoro, Nagano, Japan
| | - Jun-Ichi Satoh
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| |
Collapse
|
240
|
Le Ber I. Frontotemporal lobar dementia and amyotrophic lateral sclerosis associated with c9orf72 expansion. Rev Neurol (Paris) 2015; 171:475-81. [PMID: 26032484 DOI: 10.1016/j.neurol.2015.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 04/24/2015] [Accepted: 04/24/2015] [Indexed: 12/12/2022]
Abstract
An intronic GGGGCC repeat expansion in c9orf72 gene has been identified as the most common genetic cause of frontotemporal lobar dementia (FTLD), amyotrophic lateral sclerosis (ALS) and FTLD-ALS. The discovery of c9orf72 gene has led to important scientific progresses and has considerably changed our clinical practice over the last few years. This paper summarizes the common and less typical phenotypes associated with c9orf72 expansion, the complex pathological pattern characterized by p62/dipeptide repeat aggregates, as well as the pathological mechanisms by which the expansion might produce neurodegeneration implicating loss-of-function, RNA toxicity, RNA-binding protein sequestration and accumulation of dipeptide repeats. We also discuss the recommendations and limits for genetic testing and counseling in clinical practice.
Collapse
Affiliation(s)
- I Le Ber
- Institut du cerveau et de la moelle épinière (ICM), Inserm U1127, CNRS UMR 7225, Sorbonne Universités, Université Pierre-et-Marie-Curie, université Paris 06, UPMC-P6 UMR S 1127, hôpital Pitié-Salpêtrière, 75013 Paris, France; Centre de référence des rémences rares, hôpital de la Pitié-Salpêtrière, AP-HP, 75013 Paris, France; Département des maladies du système nerveux, hôpital de la Pitié-Salpêtrière, AP-HP, 75013 Paris, France.
| |
Collapse
|
241
|
Lant SB, Robinson AC, Thompson JC, Rollinson S, Pickering-Brown S, Snowden JS, Davidson YS, Gerhard A, Mann DMA. Patterns of microglial cell activation in frontotemporal lobar degeneration. Neuropathol Appl Neurobiol 2015; 40:686-96. [PMID: 24117616 DOI: 10.1111/nan.12092] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/02/2013] [Indexed: 12/12/2022]
Abstract
AIMS Pathological heterogeneity within patients with frontotemporal lobar degeneration (FTLD) in general precludes the accurate assignment of diagnostic subtype in life. The aim of this study was to assess the extent of microglial cell activation in FTLD in order to determine whether it might be possible to employ this as a diagnostic marker in vivo using PET ligand [11C](R)-PK11195 in order to differentiate cases of FTLD according to histological subtype. METHODS The distribution and extent of microglial cell activation was assessed semi-quantitatively in cortical grey and subcortical white matter of CD68 immunostained sections of frontal and temporal cortex from 78 pathologically confirmed cases of FTLD, 13 of Alzheimer's disease (AD) and 13 controls. RESULTS Significantly higher levels of microglial cell activation than controls occurred in all four regions in FTLD, and in three of the four regions in AD. Microglial activation was greater in frontal subcortical white matter in FTLD than AD, whereas it was higher in temporal cortical grey matter in AD than FTLD. Microglial cell activation was significantly higher in temporal subcortical white matter in FTLD-MAPT than in other genetic (GRN, C9ORF72) or non-genetic forms of FTLD. CONCLUSIONS The present study suggests that high levels of microglial cell involvement in temporal lobe (subcortical white matter) might serve as a marker of inherited FTLD associated with intronic mutations in MAPT, with a relatively intense signal in this region in PET studies using [11C](R)-PK11195 as microglial cell marker could indicate the presence of MAPT mutation in vivo.
Collapse
Affiliation(s)
- Suzannah B Lant
- Clinical and Cognitive Sciences Research Group, Institute of Brain, Behaviour and Mental Health, Faculty of Medical and Human Sciences, University of Manchester, Salford Royal Hospital, Salford, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Konno T, Tada M, Shiga A, Tsujino A, Eguchi H, Masuda-Suzukake M, Hasegawa M, Nishizawa M, Onodera O, Kakita A, Takahashi H. C9ORF72 repeat-associated non-ATG-translated polypeptides are distributed independently of TDP-43 in a Japanese patient with c9ALS. Neuropathol Appl Neurobiol 2015; 40:783-8. [PMID: 24861677 DOI: 10.1111/nan.12157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 05/21/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Takuya Konno
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia: gain or loss of function? Curr Opin Neurol 2015; 27:515-23. [PMID: 25188012 PMCID: PMC4165481 DOI: 10.1097/wco.0000000000000130] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Purpose of review The molecular mechanisms that underlie chromosome 9 open reading frame 72 (C9orf72)-associated amyotrophic lateral sclerosis and frontotemporal dementia are rapidly emerging. Two potential disease mechanisms have been postulated – gain or loss of function. We provide an overview of recent advances that support or oppose gain-of-function and loss-of-function mechanisms. Recent findings Since the discovery that a noncoding repeat expansion in C9orf72 was responsible for chromosome 9-linked amyotrophic lateral sclerosis and frontotemporal dementia in 2011, a plethora of studies have investigated clinical, pathological and mechanistic aspects of the disease. Loss of function is supported by reduced levels of C9orf72 in patient brain and functional work, revealing a role of the C9orf72 protein in endocytic and autophagic pathways and motor function. Gain of function is supported by the presence in patient brain of both repeat RNA and protein aggregates. Repeat RNA aggregates termed RNA foci, a hallmark of noncoding repeat expansion diseases, have been shown to sequester proteins involved in RNA splicing, editing, nuclear export and nucleolar function. Repeat-associated non-ATG dependent translation gives rise to toxic dipeptide repeat proteins that form inclusions in patient tissue. Antisense oligonucleotides targeting C9orf72 have shown promise for combating gain-of-function toxicity. Summary Rapid progress is being made towards understanding this common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Overall, the weight of data currently sits in favour of gain of function as the most important disease mechanism, which has important implications for the development of effective and targeted therapies.
Collapse
|
244
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating degenerative disease characterized by progressive loss of motor neurons in the motor cortex, brainstem, and spinal cord. Although defined as a motor disorder, ALS can arise concurrently with frontotemporal lobal dementia (FTLD). ALS begins focally but disseminates to cause paralysis and death. About 10% of ALS cases are caused by gene mutations, and more than 40 ALS-associated genes have been identified. While important questions about the biology of this disease remain unanswered, investigations of ALS genes have delineated pathogenic roles for (a) perturbations in protein stability and degradation, (b) altered homeostasis of critical RNA- and DNA-binding proteins, (c) impaired cytoskeleton function, and (d) non-neuronal cells as modifiers of the ALS phenotype. The rapidity of progress in ALS genetics and the subsequent acquisition of insights into the molecular biology of these genes provide grounds for optimism that meaningful therapies for ALS are attainable.
Collapse
|
245
|
Baborie A, Griffiths TD, Jaros E, Perry R, McKeith IG, Burn DJ, Masuda-Suzukake M, Hasegawa M, Rollinson S, Pickering-Brown S, Robinson AC, Davidson YS, Mann DMA. Accumulation of dipeptide repeat proteins predates that of TDP-43 in frontotemporal lobar degeneration associated with hexanucleotide repeat expansions in C9ORF72 gene. Neuropathol Appl Neurobiol 2015; 41:601-12. [PMID: 25185840 PMCID: PMC4934135 DOI: 10.1111/nan.12178] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 08/14/2014] [Indexed: 12/13/2022]
Abstract
AIMS Frontotemporal lobar degeneration (FTLD) and motor neurone disease are linked by the possession of a hexanucleotide repeat expansion in C9ORF72, and both show neuronal cytoplasmic inclusions within cerebellar and hippocampal neurones which are TDP-43 negative but immunoreactive for p62 and dipeptide repeat proteins (DPR), these being generated by a non-ATG RAN translation of the expanded region of the gene. METHODS Twenty-two cases of FTLD from Newcastle were analysed for an expansion in C9ORF72 by repeat primed PCR and Southern blot. Detailed case note analysis was performed, and blinded retrospective clinical impressions were achieved by review of clinical histories. Sections from all major brain regions were immunostained for TDP-43, p62 and DPR. The extent of TDP-43 and DPR pathology in expansion bearers was compared with that in 13 other previously identified cases from the Manchester Brain Bank with established disease. RESULTS Three Newcastle patients bearing an expansion in C9ORF72 were identified. These three patients died prematurely, two from bronchopneumonia within 10 months and 3 years of onset, and one from myocardial infarction 3 years after onset. In all three, DPR were plentiful throughout all cerebral cortical regions, hippocampus and cerebellum, but TDP-43 pathological changes were sparse. The severity of DPR pathological changes in these three patients was similar to that in the Manchester series, although the extent of TDP-43 pathology was significantly less. CONCLUSION Widespread accumulation of DPR within nerve cells may occur much earlier than that of TDP-43 in patients with FTLD bearing expansion in C9ORF72.
Collapse
Affiliation(s)
- Atik Baborie
- Department of Neuropathology, Walton Centre for Neurology and Neurosurgery, Liverpool, UK
| | - Timothy D Griffiths
- Institute of Neuroscience, Newcastle University Medical School, Newcastle upon Tyne, UK
| | - Evelyn Jaros
- Neuropathology/Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne, UK.,Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Robert Perry
- Neuropathology/Cellular Pathology, Royal Victoria Infirmary, Newcastle upon Tyne, UK.,Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Ian G McKeith
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - David J Burn
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Masami Masuda-Suzukake
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masato Hasegawa
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Sara Rollinson
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Stuart Pickering-Brown
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Manchester, UK
| | - Andrew C Robinson
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Salford Royal Hospital, Salford, UK
| | - Yvonne S Davidson
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Salford Royal Hospital, Salford, UK
| | - David M A Mann
- Institute of Brain, Behaviour and Mental Health, University of Manchester, Salford Royal Hospital, Salford, UK
| |
Collapse
|
246
|
Clinical Neuropathology teaching case 3-2015: female or male brain? Anti-ubiquitin visualizes Barr bodies in hippocampal granule cells which allows the determination of gender in human brains. Clin Neuropathol 2015; 34:115-6. [PMID: 25909954 PMCID: PMC4542183 DOI: 10.5414/np300877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
No Abstract available.
Collapse
|
247
|
Irwin DJ, Cairns NJ, Grossman M, McMillan CT, Lee EB, Van Deerlin VM, Lee VMY, Trojanowski JQ. Frontotemporal lobar degeneration: defining phenotypic diversity through personalized medicine. Acta Neuropathol 2015; 129:469-91. [PMID: 25549971 PMCID: PMC4369168 DOI: 10.1007/s00401-014-1380-1] [Citation(s) in RCA: 199] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 12/18/2014] [Accepted: 12/20/2014] [Indexed: 12/11/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) comprises two main classes of neurodegenerative diseases characterized by neuronal/glial proteinaceous inclusions (i.e., proteinopathies) including tauopathies (i.e., FTLD-Tau) and TDP-43 proteinopathies (i.e., FTLD-TDP) while other very rare forms of FTLD are known such as FTLD with FUS pathology (FTLD-FUS). This review focuses mainly on FTLD-Tau and FLTD-TDP, which may present as several clinical syndromes: a behavioral/dysexecutive syndrome (behavioral variant frontotemporal dementia); language disorders (primary progressive aphasia variants); and motor disorders (amyotrophic lateral sclerosis, corticobasal syndrome, progressive supranuclear palsy syndrome). There is considerable heterogeneity in clinical presentations of underlying neuropathology and current clinical criteria do not reliably predict underlying proteinopathies ante-mortem. In contrast, molecular etiologies of hereditary FTLD are consistently associated with specific proteinopathies. These include MAPT mutations with FTLD-Tau and GRN, C9orf72, VCP and TARDBP with FTLD-TDP. The last decade has seen a rapid expansion in our knowledge of the molecular pathologies associated with this clinically and neuropathologically heterogeneous group of FTLD diseases. Moreover, in view of current limitations to reliably diagnose specific FTLD neuropathologies prior to autopsy, we summarize the current state of the science in FTLD biomarker research including neuroimaging, biofluid and genetic analyses. We propose that combining several of these biomarker modalities will improve diagnostic specificity in FTLD through a personalized medicine approach. The goals of these efforts are to enhance power for clinical trials focused on slowing or preventing progression of spread of tau, TDP-43 and other FTLD-associated pathologies and work toward the goal of defining clinical endophenotypes of FTD.
Collapse
Affiliation(s)
- David J Irwin
- Center for Neurodegenerative Disease Research Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Frontotemporal Degeneration Center, Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nigel J. Cairns
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Murray Grossman
- Penn Frontotemporal Degeneration Center, Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Corey T. McMillan
- Penn Frontotemporal Degeneration Center, Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward B. Lee
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vivianna M. Van Deerlin
- Center for Neurodegenerative Disease Research Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Virginia M.-Y. Lee
- Center for Neurodegenerative Disease Research Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Q. Trojanowski
- Center for Neurodegenerative Disease Research Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
248
|
Sporadic and hereditary amyotrophic lateral sclerosis (ALS). Biochim Biophys Acta Mol Basis Dis 2015; 1852:679-84. [DOI: 10.1016/j.bbadis.2014.08.010] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/16/2014] [Accepted: 08/18/2014] [Indexed: 12/31/2022]
|
249
|
Scotter EL, Chen HJ, Shaw CE. TDP-43 Proteinopathy and ALS: Insights into Disease Mechanisms and Therapeutic Targets. Neurotherapeutics 2015; 12:352-63. [PMID: 25652699 PMCID: PMC4404432 DOI: 10.1007/s13311-015-0338-x] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Therapeutic options for patients with amyotrophic lateral sclerosis (ALS) are currently limited. However, recent studies show that almost all cases of ALS, as well as tau-negative frontotemporal dementia (FTD), share a common neuropathology characterized by the deposition of TAR-DNA binding protein (TDP)-43-positive protein inclusions, offering an attractive target for the design and testing of novel therapeutics. Here we demonstrate how diverse environmental stressors linked to stress granule formation, as well as mutations in genes encoding RNA processing proteins and protein degradation adaptors, initiate ALS pathogenesis via TDP-43. We review the progressive development of TDP-43 proteinopathy from cytoplasmic mislocalization and misfolding through to macroaggregation and the addition of phosphate and ubiquitin moieties. Drawing from cellular and animal studies, we explore the feasibility of therapeutics that act at each point in pathogenesis, from mitigating genetic risk using antisense oligonucleotides to modulating TDP-43 proteinopathy itself using small molecule activators of autophagy, the ubiquitin-proteasome system, or the chaperone network. We present the case that preventing the misfolding of TDP-43 and/or enhancing its clearance represents the most important target for effectively treating ALS and frontotemporal dementia.
Collapse
Affiliation(s)
- Emma L. Scotter
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, de Crespigny Park, London, SE5 8AF UK
- Department of Pharmacology, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Han-Jou Chen
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, de Crespigny Park, London, SE5 8AF UK
| | - Christopher E. Shaw
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, de Crespigny Park, London, SE5 8AF UK
| |
Collapse
|
250
|
Cooper-Knock J, Kirby J, Highley R, Shaw PJ. The Spectrum of C9orf72-mediated Neurodegeneration and Amyotrophic Lateral Sclerosis. Neurotherapeutics 2015; 12:326-39. [PMID: 25731823 PMCID: PMC4404438 DOI: 10.1007/s13311-015-0342-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The discovery that a hexanucleotide repeat expansion in C9orf72 is the most numerous genetic variant of both amyotrophic lateral sclerosis and frontotemporal dementia has opened a rapidly growing field, which may provide long hoped for advances in the understanding and treatment of these devastating diseases. In this review we describe the various phenotypes, clinical and pathological, associated with expansion of C9orf72, which go beyond amyotrophic lateral sclerosis and frontotemporal dementia to include neurodegeneration more broadly. Next we take a step back and summarize the current understanding of the C9orf72 expansion and its protein products at a molecular level. Three mechanisms are prominent: toxicity mediated directly by RNA transcribed from the repeat; toxicity mediated by dipeptide repeat proteins translated from the repeat sequence; and haploinsufficiency resulting from reduced transcription of the C9orf72 exonic sequence. A series of exciting advances have recently described how dipeptide repeat proteins might interfere with the normal role of the nucleolus in maturation of RNA binding proteins and in production of ribosomes. Importantly, these mechanisms are unlikely to be mutually exclusive. We draw attention to the fact that clinical and pathological similarities to other genetic variants without a repeat expansion must not be overlooked in ascribing a pathogenic mechanism to C9orf72-disease. Finally, with a view to impact on patient care, we discuss current practice with respect to genetic screening in patients with and without a family history of disease, and the most promising developments towards therapy that have been reported to date.
Collapse
Affiliation(s)
- Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ UK
| | - Janine Kirby
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ UK
| | - Robin Highley
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ UK
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ UK
| |
Collapse
|