101
|
Lin Z, Kim E, Ahmed M, Han G, Simmons C, Redhead Y, Bartlett J, Pena Altamira LE, Callaghan I, White MA, Singh N, Sawiak S, Spires-Jones T, Vernon AC, Coleman MP, Green J, Henstridge C, Davies JS, Cash D, Sreedharan J. MRI-guided histology of TDP-43 knock-in mice implicates parvalbumin interneuron loss, impaired neurogenesis and aberrant neurodevelopment in amyotrophic lateral sclerosis-frontotemporal dementia. Brain Commun 2021; 3:fcab114. [PMID: 34136812 PMCID: PMC8204366 DOI: 10.1093/braincomms/fcab114] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/13/2021] [Accepted: 04/17/2021] [Indexed: 01/01/2023] Open
Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia are overlapping diseases in which MRI reveals brain structural changes in advance of symptom onset. Recapitulating these changes in preclinical models would help to improve our understanding of the molecular causes underlying regionally selective brain atrophy in early disease. We therefore investigated the translational potential of the TDP-43Q331K knock-in mouse model of amyotrophic lateral sclerosis-frontotemporal dementia using MRI. We performed in vivo MRI of TDP-43Q331K knock-in mice. Regions of significant volume change were chosen for post-mortem brain tissue analyses. Ex vivo computed tomography was performed to investigate skull shape. Parvalbumin neuron density was quantified in post-mortem amyotrophic lateral sclerosis frontal cortex. Adult mutants demonstrated parenchymal volume reductions affecting the frontal lobe and entorhinal cortex in a manner reminiscent of amyotrophic lateral sclerosis-frontotemporal dementia. Subcortical, cerebellar and brain stem regions were also affected in line with observations in pre-symptomatic carriers of mutations in C9orf72, the commonest genetic cause of both amyotrophic lateral sclerosis and frontotemporal dementia. Volume loss was also observed in the dentate gyrus of the hippocampus, along with ventricular enlargement. Immunohistochemistry revealed reduced parvalbumin interneurons as a potential cellular correlate of MRI changes in mutant mice. By contrast, microglia was in a disease activated state even in the absence of brain volume loss. A reduction in immature neurons was found in the dentate gyrus, indicative of impaired adult neurogenesis, while a paucity of parvalbumin interneurons in P14 mutant mice suggests that TDP-43Q331K disrupts neurodevelopment. Computerized tomography imaging showed altered skull morphology in mutants, further suggesting a role for TDP-43Q331K in development. Finally, analysis of human post-mortem brains confirmed a paucity of parvalbumin interneurons in the prefrontal cortex in sporadic amyotrophic lateral sclerosis and amyotrophic lateral sclerosis linked to C9orf72 mutations. Regional brain MRI changes seen in human amyotrophic lateral sclerosis-frontotemporal dementia are recapitulated in TDP-43Q331K knock-in mice. By marrying in vivo imaging with targeted histology, we can unravel cellular and molecular processes underlying selective brain vulnerability in human disease. As well as helping to understand the earliest causes of disease, our MRI and histological markers will be valuable in assessing the efficacy of putative therapeutics in TDP-43Q331K knock-in mice.
Collapse
Affiliation(s)
- Ziqiang Lin
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London SE5 9RT, UK
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Eugene Kim
- BRAIN Centre (Biomarker Research And Imaging for Neuroscience), Department of Neuroimaging, IoPPN, King’s College London, London SE5 9NU, UK
| | - Mohi Ahmed
- Centre for Craniofacial and Regenerative Biology, Floor 27 Tower Wing, Guy’s Hospital, King’s College London, London SE1 9RT, UK
| | - Gang Han
- Molecular Neurobiology Group, Institute of Life Sciences, School of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Camilla Simmons
- BRAIN Centre (Biomarker Research And Imaging for Neuroscience), Department of Neuroimaging, IoPPN, King’s College London, London SE5 9NU, UK
| | - Yushi Redhead
- Centre for Craniofacial and Regenerative Biology, Floor 27 Tower Wing, Guy’s Hospital, King’s College London, London SE1 9RT, UK
| | - Jack Bartlett
- Molecular Neurobiology Group, Institute of Life Sciences, School of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Luis Emiliano Pena Altamira
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London SE5 9RT, UK
| | - Isobel Callaghan
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London SE5 9RT, UK
| | - Matthew A White
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London SE5 9RT, UK
| | - Nisha Singh
- BRAIN Centre (Biomarker Research And Imaging for Neuroscience), Department of Neuroimaging, IoPPN, King’s College London, London SE5 9NU, UK
- School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London, 4th floor Lambeth Wing, London SE1 7EH, UK
| | - Stephen Sawiak
- Department of Clinical Neurosciences, Cambridge University, Cambridge CB2 0QQ, UK
| | - Tara Spires-Jones
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London SE5 9RT, UK
| | | | - Jeremy Green
- Centre for Craniofacial and Regenerative Biology, Floor 27 Tower Wing, Guy’s Hospital, King’s College London, London SE1 9RT, UK
| | - Christopher Henstridge
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Jeffrey S Davies
- Molecular Neurobiology Group, Institute of Life Sciences, School of Medicine, Swansea University, Swansea SA2 8PP, UK
| | - Diana Cash
- BRAIN Centre (Biomarker Research And Imaging for Neuroscience), Department of Neuroimaging, IoPPN, King’s College London, London SE5 9NU, UK
| | - Jemeen Sreedharan
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London SE5 9RT, UK
| |
Collapse
|
102
|
Scekic-Zahirovic J, Sanjuan-Ruiz I, Kan V, Megat S, De Rossi P, Dieterlé S, Cassel R, Jamet M, Kessler P, Wiesner D, Tzeplaeff L, Demais V, Sahadevan S, Hembach KM, Muller HP, Picchiarelli G, Mishra N, Antonucci S, Dirrig-Grosch S, Kassubek J, Rasche V, Ludolph A, Boutillier AL, Roselli F, Polymenidou M, Lagier-Tourenne C, Liebscher S, Dupuis L. Cytoplasmic FUS triggers early behavioral alterations linked to cortical neuronal hyperactivity and inhibitory synaptic defects. Nat Commun 2021; 12:3028. [PMID: 34021132 PMCID: PMC8140148 DOI: 10.1038/s41467-021-23187-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Gene mutations causing cytoplasmic mislocalization of the RNA-binding protein FUS lead to severe forms of amyotrophic lateral sclerosis (ALS). Cytoplasmic accumulation of FUS is also observed in other diseases, with unknown consequences. Here, we show that cytoplasmic mislocalization of FUS drives behavioral abnormalities in knock-in mice, including locomotor hyperactivity and alterations in social interactions, in the absence of widespread neuronal loss. Mechanistically, we identified a progressive increase in neuronal activity in the frontal cortex of Fus knock-in mice in vivo, associated with altered synaptic gene expression. Synaptic ultrastructural and morphological defects were more pronounced in inhibitory than excitatory synapses and associated with increased synaptosomal levels of FUS and its RNA targets. Thus, cytoplasmic FUS triggers synaptic deficits, which is leading to increased neuronal activity in frontal cortex and causing related behavioral phenotypes. These results indicate that FUS mislocalization may trigger deleterious phenotypes beyond motor neuron impairment in ALS, likely relevant also for other neurodegenerative diseases characterized by FUS mislocalization.
Collapse
Affiliation(s)
- Jelena Scekic-Zahirovic
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Inmaculada Sanjuan-Ruiz
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Vanessa Kan
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany
- BioMedical Center, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Salim Megat
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Pierre De Rossi
- Department of Quantitative Biomedicine, University of Zurich, Zürich, Switzerland
| | - Stéphane Dieterlé
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Raphaelle Cassel
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
- Université de Strasbourg, UMR 7364 CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Strasbourg, France
| | - Marguerite Jamet
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Pascal Kessler
- Université de Strasbourg, Inserm, Unité mixte de service du CRBS, UMS 038, Strasbourg, France
| | - Diana Wiesner
- Department of Neurology, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | - Laura Tzeplaeff
- Université de Strasbourg, UMR 7364 CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Strasbourg, France
| | - Valérie Demais
- Plateforme Imagerie In Vitro, CNRS UPS-3156, NeuroPôle, Strasbourg, France
| | - Sonu Sahadevan
- Department of Quantitative Biomedicine, University of Zurich, Zürich, Switzerland
| | - Katharina M Hembach
- Department of Quantitative Biomedicine, University of Zurich, Zürich, Switzerland
| | | | - Gina Picchiarelli
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Nibha Mishra
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Stefano Antonucci
- Department of Neurology, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | - Sylvie Dirrig-Grosch
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France
| | - Jan Kassubek
- Department of Neurology, Ulm University, Ulm, Germany
| | - Volker Rasche
- Ulm University Medical Center, Department of Internal Medicine II, Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | - Anne-Laurence Boutillier
- Université de Strasbourg, UMR 7364 CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), Strasbourg, France
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Germany
| | | | - Clotilde Lagier-Tourenne
- Department of Neurology, The Sean M. Healey and AMG Center for ALS at Mass General, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard University and MIT, Cambridge, MA, USA
| | - Sabine Liebscher
- Institute of Clinical Neuroimmunology, Klinikum der Universität München, Ludwig-Maximilians-University Munich, Munich, Germany.
- BioMedical Center, Medical Faculty, Ludwig-Maximilians-University Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Luc Dupuis
- Université de Strasbourg, Inserm, Mécanismes centraux et périphériques de la neurodégénérescence, Strasbourg, France.
| |
Collapse
|
103
|
Chhangani D, Martín-Peña A, Rincon-Limas DE. Molecular, functional, and pathological aspects of TDP-43 fragmentation. iScience 2021; 24:102459. [PMID: 34013172 PMCID: PMC8113996 DOI: 10.1016/j.isci.2021.102459] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Transactive response DNA binding protein 43 (TDP-43) is a DNA/RNA binding protein involved in transcriptional regulation and RNA processing. It is linked to sporadic and familial amyotrophic lateral sclerosis and frontotemporal lobar degeneration. TDP-43 is predominantly nuclear, but it translocates to the cytoplasm under pathological conditions. Cytoplasmic accumulation, phosphorylation, ubiquitination and truncation of TDP-43 are the main hallmarks of TDP-43 proteinopathies. Among these processes, the pathways leading to TDP-43 fragmentation remain poorly understood. We review here the molecular and biochemical properties of several TDP-43 fragments, the mechanisms and factors mediating their production, and their potential role in disease progression. We also address the presence of TDP-43 C-terminal fragments in several neurological disorders, including Alzheimer's disease, and highlight their respective implications. Finally, we discuss features of animal models expressing TDP-43 fragments as well as recent therapeutic strategies to approach TDP-43 truncation.
Collapse
Affiliation(s)
- Deepak Chhangani
- Department of Neurology, McKnight Brain Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32611, USA
| | - Alfonso Martín-Peña
- Department of Neurology, McKnight Brain Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32611, USA
| | - Diego E Rincon-Limas
- Department of Neurology, McKnight Brain Institute, and Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL 32611, USA.,Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32611, USA.,Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
104
|
Baralle M, Romano M. Characterization of the human TARDBP gene promoter. Sci Rep 2021; 11:10438. [PMID: 34002018 PMCID: PMC8129075 DOI: 10.1038/s41598-021-89973-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/04/2021] [Indexed: 02/03/2023] Open
Abstract
The expression of TDP-43, the main component of neuronal intracellular inclusions across a broad spectrum of ALS and FTD disorders, is developmentally regulated and studies in vivo have shown that TDP-43 overexpression can be toxic, even before observation of pathological aggregates. Starting from these observations, the regulation of its expression at transcriptional level might represent a further key element for the pathogenesis of neurodegenerative diseases. Therefore, we have characterized the human TARDBP promoter, in order to study the transcriptional mechanisms of expression. Mapping of cis-acting elements by luciferase assays in different cell outlined that the activity of the promoter seems to be higher in SH-SY5Y, Neuro2A, and HeLa than in HEK293. In addition, we tested effects of two SNPs found in the promoter region of ALS patients and observed no significant effect on transcription levels in all tested cell lines. Lastly, while TDP-43 overexpression did not affect significantly the activity of its promoter (suggesting that TDP-43 does not influence its own transcription), the presence of the 5'UTR sequence and of intron-1 splicing seem to impact positively on TDP-43 expression without affecting transcript stability. In conclusion, we have identified the region spanning nucleotides 451-230 upstream from the transcription start site as the minimal region with a significant transcription activity. These results lay an important foundation for exploring the regulation of the TARDBP gene transcription by exogenous and endogenous stimuli and the implication of transcriptional mechanisms in the pathogenesis of TDP-43 proteinopathies.
Collapse
Affiliation(s)
- Marco Baralle
- grid.425196.d0000 0004 1759 4810International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park, Padriciano, Trieste, Italy
| | - Maurizio Romano
- grid.5133.40000 0001 1941 4308Department of Life Sciences, University of Trieste, Via A. Valerio 28, 34127 Trieste, Italy
| |
Collapse
|
105
|
Solomon DA, Smikle R, Reid MJ, Mizielinska S. Altered Phase Separation and Cellular Impact in C9orf72-Linked ALS/FTD. Front Cell Neurosci 2021; 15:664151. [PMID: 33967699 PMCID: PMC8096919 DOI: 10.3389/fncel.2021.664151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/19/2021] [Indexed: 12/21/2022] Open
Abstract
Since the discovery of the C9orf72 repeat expansion mutation as causative for chromosome 9-linked amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) in 2011, a multitude of cellular pathways have been implicated. However, evidence has also been accumulating for a key mechanism of cellular compartmentalization—phase separation. Liquid-liquid phase separation (LLPS) is fundamental for the formation of membraneless organelles including stress granules, the nucleolus, Cajal bodies, nuclear speckles and the central channel of the nuclear pore. Evidence has now accumulated showing that the formation and function of these membraneless organelles is impaired by both the toxic arginine rich dipeptide repeat proteins (DPRs), translated from the C9orf72 repeat RNA transcript, and the repeat RNA itself. Both the arginine rich DPRs and repeat RNA themselves undergo phase separation and disrupt the physiological phase separation of proteins involved in the formation of these liquid-like organelles. Hence abnormal phase separation may explain a number of pathological cellular phenomena associated with C9orf72-ALS/FTD. In this review article, we will discuss the principles of phase separation, phase separation of the DPRs and repeat RNA themselves and how they perturb LLPS associated with membraneless organelles and the functional consequences of this. We will then discuss how phase separation may impact the major pathological feature of C9orf72-ALS/FTD, TDP-43 proteinopathy, and how LLPS may be targeted therapeutically in disease.
Collapse
Affiliation(s)
- Daniel A Solomon
- UK Dementia Research Institute at King's College London, London, United Kingdom.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Rebekah Smikle
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Matthew J Reid
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Sarah Mizielinska
- UK Dementia Research Institute at King's College London, London, United Kingdom.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| |
Collapse
|
106
|
Månberg A, Skene N, Sanders F, Trusohamn M, Remnestål J, Szczepińska A, Aksoylu IS, Lönnerberg P, Ebarasi L, Wouters S, Lehmann M, Olofsson J, von Gohren Antequera I, Domaniku A, De Schaepdryver M, De Vocht J, Poesen K, Uhlén M, Anink J, Mijnsbergen C, Vergunst-Bosch H, Hübers A, Kläppe U, Rodriguez-Vieitez E, Gilthorpe JD, Hedlund E, Harris RA, Aronica E, Van Damme P, Ludolph A, Veldink J, Ingre C, Nilsson P, Lewandowski SA. Altered perivascular fibroblast activity precedes ALS disease onset. Nat Med 2021; 27:640-646. [PMID: 33859435 DOI: 10.1038/s41591-021-01295-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022]
Abstract
Apart from well-defined factors in neuronal cells1, only a few reports consider that the variability of sporadic amyotrophic lateral sclerosis (ALS) progression can depend on less-defined contributions from glia2,3 and blood vessels4. In this study we use an expression-weighted cell-type enrichment method to infer cell activity in spinal cord samples from patients with sporadic ALS and mouse models of this disease. Here we report that patients with sporadic ALS present cell activity patterns consistent with two mouse models in which enrichments of vascular cell genes preceded microglial response. Notably, during the presymptomatic stage, perivascular fibroblast cells showed the strongest gene enrichments, and their marker proteins SPP1 and COL6A1 accumulated in enlarged perivascular spaces in patients with sporadic ALS. Moreover, in plasma of 574 patients with ALS from four independent cohorts, increased levels of SPP1 at disease diagnosis repeatedly predicted shorter survival with stronger effect than the established risk factors of bulbar onset or neurofilament levels in cerebrospinal fluid. We propose that the activity of the recently discovered perivascular fibroblast can predict survival of patients with ALS and provide a new conceptual framework to re-evaluate definitions of ALS etiology.
Collapse
Affiliation(s)
- Anna Månberg
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Nathan Skene
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.,Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK.,United Kingdom Dementia Research Institute, London, UK
| | - Folkert Sanders
- Department of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden
| | - Marta Trusohamn
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Julia Remnestål
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Anna Szczepińska
- Department of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden
| | - Inci Sevval Aksoylu
- Department of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden
| | - Peter Lönnerberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Lwaki Ebarasi
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Stefan Wouters
- Department of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden
| | - Manuela Lehmann
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Jennie Olofsson
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Inti von Gohren Antequera
- Department of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden
| | - Aylin Domaniku
- Department of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden
| | - Maxim De Schaepdryver
- Laboratory for Neurobiomarker Research, Department of Neurology, Leuven Brain Institute, KU Leuven (University of Leuven), Leuven, Belgium
| | - Joke De Vocht
- Neurology Department and Center for Brain & Disease Research, KU Leuven, VIB, Leuven, Belgium
| | - Koen Poesen
- Laboratory for Neurobiomarker Research, Department of Neurology, Leuven Brain Institute, KU Leuven (University of Leuven), Leuven, Belgium.,Laboratory Medicine, UZ Leuven (University Hospital Leuven), Leuven, Belgium
| | - Mathias Uhlén
- Division of Systems Biology, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jasper Anink
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Caroline Mijnsbergen
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Hermieneke Vergunst-Bosch
- UMC Utrecht Brain Center, University Medical Center Utrecht, Department of Neurology, Utrecht University, Utrecht, the Netherlands
| | - Annemarie Hübers
- University of Ulm, Neurology Clinic, Ulm, Germany.,Division of Neurology, Geneva University Hospital, Geneva, Switzerland
| | - Ulf Kläppe
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Elena Rodriguez-Vieitez
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | | | - Eva Hedlund
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Robert A Harris
- Department of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Philip Van Damme
- Neurology Department and Center for Brain & Disease Research, KU Leuven, VIB, Leuven, Belgium
| | - Albert Ludolph
- University of Ulm, Neurology Clinic, Ulm, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Ulm, Bonn, Germany
| | - Jan Veldink
- UMC Utrecht Brain Center, University Medical Center Utrecht, Department of Neurology, Utrecht University, Utrecht, the Netherlands
| | - Caroline Ingre
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Peter Nilsson
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden
| | - Sebastian A Lewandowski
- Division of Affinity Proteomics, Department of Protein Science, KTH Royal Institute of Technology, SciLifeLab, Stockholm, Sweden. .,Department of Clinical Neuroscience, Karolinska Institute, Centre for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden.
| |
Collapse
|
107
|
Pathway from TDP-43-Related Pathology to Neuronal Dysfunction in Amyotrophic Lateral Sclerosis and Frontotemporal Lobar Degeneration. Int J Mol Sci 2021; 22:ijms22083843. [PMID: 33917673 PMCID: PMC8068029 DOI: 10.3390/ijms22083843] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Transactivation response DNA binding protein 43 kDa (TDP-43) is known to be a pathologic protein in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). TDP-43 is normally a nuclear protein, but affected neurons of ALS or FTLD patients exhibit mislocalization of nuclear TDP-43 and cytoplasmic inclusions. Basic studies have suggested gain-of-neurotoxicity of aggregated TDP-43 or loss-of-function of intrinsic, nuclear TDP-43. It has also been hypothesized that the aggregated TDP-43 functions as a propagation seed of TDP-43 pathology. However, a mechanistic discrepancy between the TDP-43 pathology and neuronal dysfunctions remains. This article aims to review the observations of TDP-43 pathology in autopsied ALS and FTLD patients and address pathways of neuronal dysfunction related to the neuropathological findings, focusing on impaired clearance of TDP-43 and synaptic alterations in TDP-43-related ALS and FTLD. The former may be relevant to intraneuronal aggregation of TDP-43 and exocytosis of propagation seeds, whereas the latter may be related to neuronal dysfunction induced by TDP-43 pathology. Successful strategies of disease-modifying therapy might arise from further investigation of these subcellular alterations.
Collapse
|
108
|
Klim JR, Pintacuda G, Nash LA, Guerra San Juan I, Eggan K. Connecting TDP-43 Pathology with Neuropathy. Trends Neurosci 2021; 44:424-440. [PMID: 33832769 DOI: 10.1016/j.tins.2021.02.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/04/2021] [Accepted: 02/24/2021] [Indexed: 01/22/2023]
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43), a multifunctional nucleic acid-binding protein, is a primary component of insoluble aggregates associated with several devastating nervous system disorders; mutations in TARDBP, its encoding gene, are a cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here, we review established and emerging roles of TDP-43 and consider how its dysfunction impinges on RNA homeostasis in the nervous system, thereby contributing to neural degeneration. Notably, improper splicing of the axonal growth-associated factor STMN2 has recently been connected to TDP-43 dysfunction, providing a mechanistic link between TDP-43 proteinopathies and neuropathy. This review highlights how a deep understanding of the function of TDP-43 in the brain might be leveraged to develop new targeted therapies for several neurological disorders.
Collapse
Affiliation(s)
- Joseph R Klim
- Department of Stem Cell and Regenerative Biology, Department of Molecular and Cellular Biology, and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Greta Pintacuda
- Department of Stem Cell and Regenerative Biology, Department of Molecular and Cellular Biology, and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Leslie A Nash
- Department of Stem Cell and Regenerative Biology, Department of Molecular and Cellular Biology, and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Irune Guerra San Juan
- Department of Stem Cell and Regenerative Biology, Department of Molecular and Cellular Biology, and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Department of Molecular and Cellular Biology, and Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
109
|
Tziortzouda P, Van Den Bosch L, Hirth F. Triad of TDP43 control in neurodegeneration: autoregulation, localization and aggregation. Nat Rev Neurosci 2021; 22:197-208. [PMID: 33654312 DOI: 10.1038/s41583-021-00431-1] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 01/31/2023]
Abstract
Cytoplasmic aggregation of TAR DNA-binding protein 43 (TDP43; also known as TARDBP or TDP-43) is a key pathological feature of several neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). TDP43 typically resides in the nucleus but can shuttle between the nucleus and the cytoplasm to exert its multiple functions, which include regulation of the splicing, trafficking and stabilization of RNA. Cytoplasmic mislocalization and nuclear loss of TDP43 have both been associated with ALS and FTD, suggesting that calibrated levels and correct localization of TDP43 - achieved through an autoregulatory loop and tightly controlled nucleocytoplasmic transport - safeguard its normal function. Furthermore, TDP43 can undergo phase transitions, including its dispersion into liquid droplets and its accumulation into irreversible cytoplasmic aggregates. Thus, autoregulation, nucleocytoplasmic transport and phase transition are all part of an intrinsic control system regulating the physiological levels and localization of TDP43, and together are essential for the cellular homeostasis that is affected in neurodegenerative disease.
Collapse
Affiliation(s)
- Paraskevi Tziortzouda
- Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
- Laboratory of Neurobiology, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium.
- Laboratory of Neurobiology, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
| | - Frank Hirth
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
110
|
Fazal R, Boeynaems S, Swijsen A, De Decker M, Fumagalli L, Moisse M, Vanneste J, Guo W, Boon R, Vercruysse T, Eggermont K, Swinnen B, Beckers J, Pakravan D, Vandoorne T, Vanden Berghe P, Verfaillie C, Van Den Bosch L, Van Damme P. HDAC6 inhibition restores TDP-43 pathology and axonal transport defects in human motor neurons with TARDBP mutations. EMBO J 2021; 40:e106177. [PMID: 33694180 PMCID: PMC8013789 DOI: 10.15252/embj.2020106177] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 01/28/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
TDP-43 is the major component of pathological inclusions in most ALS patients and in up to 50% of patients with frontotemporal dementia (FTD). Heterozygous missense mutations in TARDBP, the gene encoding TDP-43, are one of the common causes of familial ALS. In this study, we investigate TDP-43 protein behavior in induced pluripotent stem cell (iPSC)-derived motor neurons from three ALS patients with different TARDBP mutations, three healthy controls and an isogenic control. TARDPB mutations induce several TDP-43 changes in spinal motor neurons, including cytoplasmic mislocalization and accumulation of insoluble TDP-43, C-terminal fragments, and phospho-TDP-43. By generating iPSC lines with allele-specific tagging of TDP-43, we find that mutant TDP-43 initiates the observed disease phenotypes and has an altered interactome as indicated by mass spectrometry. Our findings also indicate that TDP-43 proteinopathy results in a defect in mitochondrial transport. Lastly, we show that pharmacological inhibition of histone deacetylase 6 (HDAC6) restores the observed TDP-43 pathologies and the axonal mitochondrial motility, suggesting that HDAC6 inhibition may be an interesting therapeutic target for neurodegenerative disorders linked to TDP-43 pathology.
Collapse
Affiliation(s)
- Raheem Fazal
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Steven Boeynaems
- Department of GeneticsStanford University School of MedicineStanfordCAUSA
| | - Ann Swijsen
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Mathias De Decker
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Laura Fumagalli
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Matthieu Moisse
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Joni Vanneste
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Wenting Guo
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
- Stem Cell InstituteDepartment of Development and RegenerationStem Cell Biology and EmbryologyKU LeuvenLeuvenBelgium
| | - Ruben Boon
- Stem Cell InstituteDepartment of Development and RegenerationStem Cell Biology and EmbryologyKU LeuvenLeuvenBelgium
| | - Thomas Vercruysse
- Department of Microbiology, Immunology and TransplantationLaboratory of Virology and ChemotherapyRega Institute for Medical ResearchKU LeuvenLeuvenBelgium
| | - Kristel Eggermont
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Bart Swinnen
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
- Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| | - Jimmy Beckers
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Donya Pakravan
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Tijs Vandoorne
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Pieter Vanden Berghe
- Department of Chronic Diseases, Metabolism and AgeingTranslational Research in GastroIntestinal Disorders, KU LeuvenLeuvenBelgium
| | - Catherine Verfaillie
- Stem Cell InstituteDepartment of Development and RegenerationStem Cell Biology and EmbryologyKU LeuvenLeuvenBelgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental NeurologyLeuven Brain Institute (LBI)KU Leuven – University of LeuvenLeuvenBelgium
- Center for Brain & Disease ResearchLaboratory of NeurobiologyVIBLeuvenBelgium
- Department of NeurologyUniversity Hospitals LeuvenLeuvenBelgium
| |
Collapse
|
111
|
Zhang S, Shao Z, Liu X, Hou M, Cheng F, Lei D, Yuan H. The E50K optineurin mutation impacts autophagy-mediated degradation of TDP-43 and leads to RGC apoptosis in vivo and in vitro. Cell Death Dis 2021; 7:49. [PMID: 33723228 PMCID: PMC7960725 DOI: 10.1038/s41420-021-00432-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/23/2021] [Accepted: 02/13/2021] [Indexed: 01/31/2023]
Abstract
The glaucoma-associated E50K mutation in optineurin (OPTN) is known to affect autophagy and cause the apoptosis of retinal ganglion cells (RGCs), but the pathogenic mechanism remains unclear. In this study, we investigated whether the OPTN (E50K) mutation caused TDP-43 aggregation by disrupting autophagy in vivo and in vitro. OPTN (E50K) mutant mice were generated and analysed for genotype and phenotype. Adeno-associated virus type 2 vectors containing either GFP only, GFP-tagged wild-type OPTN or GFP-tagged E50K-mutated OPTN were used to transfect R28 cells. Loss of RGCs decreased retinal thickness and visual impairment were observed in OPTN (E50K) mice compared with WT mice. Moreover, overexpression of E50K OPTN induced R28 cell apoptosis. Increased p62/SQSTM1 and LC3-II levels indicated that autophagic flux was inhibited and contributed to TDP-43 aggregation in vivo and in vitro. We found that rapamycin effectively reduced the aggregation of TDP-43 in OPTN (E50K) mice and decreased the protein levels of p62/SQSTM1 and the autophagic marker LC3-II. Moreover, rapamycin increased the RGC number and visual function of E50K mice. In addition, we also observed increased cytoplasmic TDP-43 in the spinal cord and motor dysfunction in 24-month-old OPTN (E50K) mice, indicating that TDP-43 accumulation may be the common pathological mechanism of glaucoma and amyotrophic lateral sclerosis (ALS). In conclusion, the disruption of autophagy by OPTN (E50K) affected the degradation of TDP-43 and may play an important role in OPTN (E50K)-mediated glaucomatous retinal neurodegeneration.
Collapse
Affiliation(s)
- Shiqi Zhang
- grid.412463.60000 0004 1762 6325Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China ,grid.410736.70000 0001 2204 9268The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Heilongjiang Province, Harbin, China
| | - Zhengbo Shao
- grid.412463.60000 0004 1762 6325Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China ,grid.412463.60000 0004 1762 6325Research Institute, Second Affiliated Hospital of Harbin Medical University, Harbin, China ,grid.412463.60000 0004 1762 6325Future Medical Laboratory, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinna Liu
- grid.412463.60000 0004 1762 6325Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China ,grid.410736.70000 0001 2204 9268The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Heilongjiang Province, Harbin, China
| | - Mingying Hou
- grid.412463.60000 0004 1762 6325Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China ,grid.410736.70000 0001 2204 9268The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Heilongjiang Province, Harbin, China
| | - Fang Cheng
- grid.412463.60000 0004 1762 6325Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China ,grid.412463.60000 0004 1762 6325Research Institute, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dawei Lei
- grid.412463.60000 0004 1762 6325Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huiping Yuan
- grid.412463.60000 0004 1762 6325Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China ,grid.412463.60000 0004 1762 6325Research Institute, Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
112
|
Raffaele S, Boccazzi M, Fumagalli M. Oligodendrocyte Dysfunction in Amyotrophic Lateral Sclerosis: Mechanisms and Therapeutic Perspectives. Cells 2021; 10:cells10030565. [PMID: 33807572 PMCID: PMC8000560 DOI: 10.3390/cells10030565] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Myelin is the lipid-rich structure formed by oligodendrocytes (OLs) that wraps the axons in multilayered sheaths, assuring protection, efficient saltatory signal conduction and metabolic support to neurons. In the last few years, the impact of OL dysfunction and myelin damage has progressively received more attention and is now considered to be a major contributing factor to neurodegeneration in several neurological diseases, including amyotrophic lateral sclerosis (ALS). Upon OL injury, oligodendrocyte precursor cells (OPCs) of adult nervous tissue sustain the generation of new OLs for myelin reconstitution, but this spontaneous regeneration process fails to successfully counteract myelin damage. Of note, the functions of OPCs exceed the formation and repair of myelin, and also involve the trophic support to axons and the capability to exert an immunomodulatory role, which are particularly relevant in the context of neurodegeneration. In this review, we deeply analyze the impact of dysfunctional OLs in ALS pathogenesis. The possible mechanisms underlying OL degeneration, defective OPC maturation, and impairment in energy supply to motor neurons (MNs) have also been examined to provide insights on future therapeutic interventions. On this basis, we discuss the potential therapeutic utility in ALS of several molecules, based on their remyelinating potential or capability to enhance energy metabolism.
Collapse
|
113
|
Buratti E. Trends in Understanding the Pathological Roles of TDP-43 and FUS Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:243-267. [PMID: 33433879 DOI: 10.1007/978-3-030-51140-1_15] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Following the discovery of TDP-43 and FUS involvement in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar dementia (FTLD), the major challenge in the field has been to understand their physiological functions, both in normal and disease conditions. The hope is that this knowledge will improve our understanding of disease and lead to the development of effective therapeutic options. Initially, the focus has been directed at characterizing the role of these proteins in the control of RNA metabolism, because the main function of TDP-43 and FUS is to bind coding and noncoding RNAs to regulate their life cycle within cells. As a result, we now have an in-depth picture of the alterations that occur in RNA metabolism following their aggregation in various ALS/FTLD models and, to a somewhat lesser extent, in patients' brains. In parallel, progress has been made with regard to understanding how aggregation of these proteins occurs in neurons, how it can spread in different brain regions, and how these changes affect various metabolic cellular pathways to result in neuronal death. The aim of this chapter will be to provide a general overview of the trending topics in TDP-43 and FUS investigations and to highlight what might represent the most promising avenues of research in the years to come.
Collapse
Affiliation(s)
- Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy.
| |
Collapse
|
114
|
Zhang X, Wang F, Hu Y, Chen R, Meng D, Guo L, Lv H, Guan J, Jia Y. In vivo stress granule misprocessing evidenced in a FUS knock-in ALS mouse model. Brain 2020; 143:1350-1367. [PMID: 32358598 DOI: 10.1093/brain/awaa076] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 01/10/2020] [Accepted: 02/02/2020] [Indexed: 12/13/2022] Open
Abstract
Many RNA-binding proteins, including TDP-43, FUS, and TIA1, are stress granule components, dysfunction of which causes amyotrophic lateral sclerosis (ALS). However, whether a mutant RNA-binding protein disrupts stress granule processing in vivo in pathogenesis is unknown. Here we establish a FUS ALS mutation, p.R521C, knock-in mouse model that carries impaired motor ability and late-onset motor neuron loss. In disease-susceptible neurons, stress induces mislocalization of mutant FUS into stress granules and upregulation of ubiquitin, two hallmarks of disease pathology. Additionally, stress aggravates motor performance decline in the mutant mouse. By using two-photon imaging in TIA1-EGFP transduced animals, we document more intensely TIA1-EGFP-positive granules formed hours but cleared weeks after stress challenge in neurons in the mutant cortex. Moreover, neurons with severe granule misprocessing die days after stress challenge. Therefore, we argue that stress granule misprocessing is pathogenic in ALS, and the model we provide here is sound for further disease mechanistic study.
Collapse
Affiliation(s)
- Xue Zhang
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Fengchao Wang
- Animal core facility, National Institute of Biological Sciences, Beijing, China
| | - Yi Hu
- School of Life Sciences, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Runze Chen
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Dawei Meng
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Liang Guo
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Hailong Lv
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| | - Jisong Guan
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Yichang Jia
- Tsinghua-Peking Joint Center for Life Science, Beijing, China.,School of Medicine, Medical Science Building, Room D204, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research at Tsinghua Beijing, China
| |
Collapse
|
115
|
Shabir O, Moll TA, Matuszyk MM, Eyre B, Dake MD, Berwick J, Francis SE. Preclinical models of disease and multimorbidity with focus upon cardiovascular disease and dementia. Mech Ageing Dev 2020; 192:111361. [PMID: 32998028 DOI: 10.1016/j.mad.2020.111361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
|
116
|
de Boer EMJ, Orie VK, Williams T, Baker MR, De Oliveira HM, Polvikoski T, Silsby M, Menon P, van den Bos M, Halliday GM, van den Berg LH, Van Den Bosch L, van Damme P, Kiernan MC, van Es MA, Vucic S. TDP-43 proteinopathies: a new wave of neurodegenerative diseases. J Neurol Neurosurg Psychiatry 2020; 92:jnnp-2020-322983. [PMID: 33177049 PMCID: PMC7803890 DOI: 10.1136/jnnp-2020-322983] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/28/2020] [Accepted: 09/13/2020] [Indexed: 12/31/2022]
Abstract
Inclusions of pathogenic deposits containing TAR DNA-binding protein 43 (TDP-43) are evident in the brain and spinal cord of patients that present across a spectrum of neurodegenerative diseases. For instance, the majority of patients with sporadic amyotrophic lateral sclerosis (up to 97%) and a substantial proportion of patients with frontotemporal lobar degeneration (~45%) exhibit TDP-43 positive neuronal inclusions, suggesting a role for this protein in disease pathogenesis. In addition, TDP-43 inclusions are evident in familial ALS phenotypes linked to multiple gene mutations including the TDP-43 gene coding (TARDBP) and unrelated genes (eg, C9orf72). While TDP-43 is an essential RNA/DNA binding protein critical for RNA-related metabolism, determining the pathophysiological mechanisms through which TDP-43 mediates neurodegeneration appears complex, and unravelling these molecular processes seems critical for the development of effective therapies. This review highlights the key physiological functions of the TDP-43 protein, while considering an expanding spectrum of neurodegenerative diseases associated with pathogenic TDP-43 deposition, and dissecting key molecular pathways through which TDP-43 may mediate neurodegeneration.
Collapse
Affiliation(s)
- Eva Maria Johanna de Boer
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Viyanti K Orie
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Timothy Williams
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mark R Baker
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Clinical Neurophysiology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Hugo M De Oliveira
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Tuomo Polvikoski
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Neuropathology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Matthew Silsby
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Parvathi Menon
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Mehdi van den Bos
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Glenda M Halliday
- Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Leonard H van den Berg
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
| | - Philip van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
- Center for Brain & Disease Research, Laboratory of Neurobiology, VIB, Leuven, Belgium
- Department of Neurology, University Hospital Leuven, Leuven, Belgium
| | - Matthew C Kiernan
- Brain and Mind Center, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Michael A van Es
- Department of Neurology, Brain Centre Rudolf Magnus, Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Steve Vucic
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
117
|
Watkins J, Ghosh A, Keerie AFA, Alix JJP, Mead RJ, Sreedharan J. Female sex mitigates motor and behavioural phenotypes in TDP-43 Q331K knock-in mice. Sci Rep 2020; 10:19220. [PMID: 33154447 PMCID: PMC7645778 DOI: 10.1038/s41598-020-76070-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are overlapping neurodegenerative disorders. ALS is more commonly seen in men than women and the same may be the case for FTD. Preclinical models demonstrating sex-specific vulnerability may help to understand female resistance to ALS-FTD and thereby identify routes to therapy. We previously characterised a TDP-43Q331K knock-in mouse, which demonstrated behavioural phenotypes reminiscent of ALS-FTD in males. Here we present our behavioural observations of female TDP-43Q331K mutants. Female TDP-43Q331K knock-in mice displayed increased weight relative to wild-type and increased food intake at 20 months of age, much later than previously observed in male mutants. Spontaneous digging behaviour was initially normal and only declined in mutants in the second year of life. Gait analysis using Catwalk (https://www.noldus.com/catwalk-xt) found significant deficits in the second year of life, while nocturnal running behaviour was attenuated from ~ 250 days of life. These results indicate that while female TDP-43Q331K knock-in mice do display progressive behavioural phenotypes, these are less severe than we previously noted in male mutants. Further studies of male and female TDP-43Q331K knock-in mice may help to unravel the mechanisms underlying sex-specific vulnerability in ALS-FTD.
Collapse
Affiliation(s)
- Jodie Watkins
- Department of Neuroscience, School of Medicine, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Anshua Ghosh
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK
| | - Amy F A Keerie
- Department of Neuroscience, School of Medicine, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
| | - James J P Alix
- Department of Neuroscience, School of Medicine, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
| | - Richard J Mead
- Department of Neuroscience, School of Medicine, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK.
| | - Jemeen Sreedharan
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London, SE5 9RX, UK.
| |
Collapse
|
118
|
McAlary L, Chew YL, Lum JS, Geraghty NJ, Yerbury JJ, Cashman NR. Amyotrophic Lateral Sclerosis: Proteins, Proteostasis, Prions, and Promises. Front Cell Neurosci 2020; 14:581907. [PMID: 33328890 PMCID: PMC7671971 DOI: 10.3389/fncel.2020.581907] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is characterized by the progressive degeneration of the motor neurons that innervate muscle, resulting in gradual paralysis and culminating in the inability to breathe or swallow. This neuronal degeneration occurs in a spatiotemporal manner from a point of onset in the central nervous system (CNS), suggesting that there is a molecule that spreads from cell-to-cell. There is strong evidence that the onset and progression of ALS pathology is a consequence of protein misfolding and aggregation. In line with this, a hallmark pathology of ALS is protein deposition and inclusion formation within motor neurons and surrounding glia of the proteins TAR DNA-binding protein 43, superoxide dismutase-1, or fused in sarcoma. Collectively, the observed protein aggregation, in conjunction with the spatiotemporal spread of symptoms, strongly suggests a prion-like propagation of protein aggregation occurs in ALS. In this review, we discuss the role of protein aggregation in ALS concerning protein homeostasis (proteostasis) mechanisms and prion-like propagation. Furthermore, we examine the experimental models used to investigate these processes, including in vitro assays, cultured cells, invertebrate models, and murine models. Finally, we evaluate the therapeutics that may best prevent the onset or spread of pathology in ALS and discuss what lies on the horizon for treating this currently incurable disease.
Collapse
Affiliation(s)
- Luke McAlary
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Yee Lian Chew
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Jeremy Stephen Lum
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Nicholas John Geraghty
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Justin John Yerbury
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia
- Molecular Horizons and School of Chemistry and Molecular Bioscience, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| | - Neil R. Cashman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
119
|
Kim E, White MA, Phillips BU, Lopez-Cruz L, Kim H, Heath CJ, Lee JE, Saksida LM, Sreedharan J, Bussey TJ. Coexistence of perseveration and apathy in the TDP-43 Q331K knock-in mouse model of ALS-FTD. Transl Psychiatry 2020; 10:377. [PMID: 33149110 PMCID: PMC7643138 DOI: 10.1038/s41398-020-01078-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 05/21/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
Perseveration and apathy are two of the most common behavioural and psychological symptoms of dementia (BPSDs) in amyotrophic lateral sclerosis-frontotemporal dementia (ALS-FTD). Availability of a validated and behaviourally characterised animal model is crucial for translational research into BPSD in the FTD context. We behaviourally evaluated the male TDP-43Q331K mouse, an ALS-FTD model with a human-equivalent mutation (TDP-43Q331K) knocked into the endogenous Tardbp gene. We utilised a panel of behavioural tasks delivered using the rodent touchscreen apparatus, including progressive ratio (PR), extinction and visual discrimination/reversal learning (VDR) assays to examine motivation, response inhibition and cognitive flexibility, respectively. Relative to WT littermates, TDP-43Q331K mice exhibited increased responding under a PR schedule. While elevated PR responding is typically an indication of increased motivation for reward, a trial-by-trial response rate analysis revealed that TDP-43Q331K mice exhibited decreased maximal response rate and slower response decay rate, suggestive of reduced motivation and a perseverative behavioural phenotype, respectively. In the extinction assay, TDP-43Q331K mice displayed increased omissions during the early phase of each session, consistent with a deficit in activational motivation. Finally, the VDR task revealed cognitive inflexibility, manifesting as stimulus-bound perseveration. Together, our data indicate that male TDP-43Q331K mice exhibit a perseverative phenotype with some evidence of apathy-like behaviour, similar to BPSDs observed in human ALS-FTD patients. The TDP-43Q331K knock-in mouse therefore has features that recommend it as a useful platform to facilitate translational research into behavioural symptoms in the context of ALS-FTD.
Collapse
Affiliation(s)
- Eosu Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Matthew A White
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Benjamin U Phillips
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Laura Lopez-Cruz
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- School of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, UK
| | - Hyunjeong Kim
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Christopher J Heath
- School of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, UK
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Lisa M Saksida
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
- Molecular Medicine Research Laboratories, Robarts Research Institute & Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
- The Brain and Mind Institute, Western University, London, ON, Canada
| | - Jemeen Sreedharan
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Timothy J Bussey
- Department of Psychology and MRC/Wellcome Trust Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK.
- Molecular Medicine Research Laboratories, Robarts Research Institute & Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada.
- The Brain and Mind Institute, Western University, London, ON, Canada.
| |
Collapse
|
120
|
The role of hnRNPs in frontotemporal dementia and amyotrophic lateral sclerosis. Acta Neuropathol 2020; 140:599-623. [PMID: 32748079 PMCID: PMC7547044 DOI: 10.1007/s00401-020-02203-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022]
Abstract
Dysregulated RNA metabolism is emerging as a crucially important mechanism underpinning the pathogenesis of frontotemporal dementia (FTD) and the clinically, genetically and pathologically overlapping disorder of amyotrophic lateral sclerosis (ALS). Heterogeneous nuclear ribonucleoproteins (hnRNPs) comprise a family of RNA-binding proteins with diverse, multi-functional roles across all aspects of mRNA processing. The role of these proteins in neurodegeneration is far from understood. Here, we review some of the unifying mechanisms by which hnRNPs have been directly or indirectly linked with FTD/ALS pathogenesis, including their incorporation into pathological inclusions and their best-known roles in pre-mRNA splicing regulation. We also discuss the broader functionalities of hnRNPs including their roles in cryptic exon repression, stress granule assembly and in co-ordinating the DNA damage response, which are all emerging pathogenic themes in both diseases. We then present an integrated model that depicts how a broad-ranging network of pathogenic events can arise from declining levels of functional hnRNPs that are inadequately compensated for by autoregulatory means. Finally, we provide a comprehensive overview of the most functionally relevant cellular roles, in the context of FTD/ALS pathogenesis, for hnRNPs A1-U.
Collapse
|
121
|
Ruiz-Ruiz C, García-Magro N, Negredo P, Avendaño C, Bhattacharya A, Ceusters M, García AG. Chronic administration of P2X7 receptor antagonist JNJ-47965567 delays disease onset and progression, and improves motor performance in ALS SOD1 G93A female mice. Dis Model Mech 2020; 13:13/10/dmm045732. [PMID: 33174532 PMCID: PMC7648608 DOI: 10.1242/dmm.045732] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is one of the main physiopathological mechanisms of amyotrophic lateral sclerosis (ALS), produced by the chronic activation of microglia in the CNS. This process is triggered by the persistent activation of the ATP-gated P2X7 receptor (P2RX7, hereafter referred to as P2X7R). The present study aimed to evaluate the effects of the chronic treatment with the P2X7R antagonist JNJ-47965567 in the development and progression of ALS in the SOD1G93A murine model. SOD1G93A mice were intraperitoneally (i.p.) injected with either 30 mg/kg of JNJ-47965567 or vehicle 4 times per week, from pre-onset age (here, postnatal day 60; P60) until study endpoint. Body weight, motor coordination, phenotypic score, disease onset and survival were measured throughout the study, and compared between vehicle- and drug-injected groups. Treatment with the P2X7R antagonist JNJ-47965567 delayed disease onset, reduced body weight loss and improved motor coordination and phenotypic score in female SOD1G93A mice, although it did not increase lifespan. Interestingly, neither beneficial nor detrimental effects were observed in males in any of the analyzed parameters. Treatment did not affect motor neuron survival or ChAT, Iba-1 and P2X7R protein expression in endpoint individuals of mixed sexes. Overall, chronic administration of JNJ-47965567 for 4 times per week to SOD1G93A mice from pre-onset stage altered disease progression in female individuals while it did not have any effect in males. Our results suggest a partial, yet important, effect of P2X7R in the development and progression of ALS.
Collapse
Affiliation(s)
- Cristina Ruiz-Ruiz
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid 28029, Spain.,Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid 28029, Spain
| | - Nuria García-Magro
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid 28029, Spain
| | - Pilar Negredo
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid 28029, Spain
| | - Carlos Avendaño
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid 28029, Spain
| | - Anindya Bhattacharya
- Neuroscience Therapeutic Area, Janssen Research and Development LLC., 3210 Merryfield Row, San Diego, CA 92121, USA
| | - Marc Ceusters
- Neuroscience Therapeutic Area, Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Beerse B-2340, Belgium
| | - Antonio G García
- Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid 28029, Spain .,Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid 28029, Spain.,Instituto de Investigación Sanitaria, Hospital Universitario de La Princesa, Madrid 28006, Spain
| |
Collapse
|
122
|
Selective neuronal degeneration in MATR3 S85C knock-in mouse model of early-stage ALS. Nat Commun 2020; 11:5304. [PMID: 33082323 PMCID: PMC7576598 DOI: 10.1038/s41467-020-18949-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
A missense mutation, S85C, in the MATR3 gene is a genetic cause for amyotrophic lateral sclerosis (ALS). It is unclear how the S85C mutation affects MATR3 function and contributes to disease. Here, we develop a mouse model that harbors the S85C mutation in the endogenous Matr3 locus using the CRISPR/Cas9 system. MATR3 S85C knock-in mice recapitulate behavioral and neuropathological features of early-stage ALS including motor impairment, muscle atrophy, neuromuscular junction defects, Purkinje cell degeneration and neuroinflammation in the cerebellum and spinal cord. Our neuropathology data reveals a loss of MATR3 S85C protein in the cell bodies of Purkinje cells and motor neurons, suggesting that a decrease in functional MATR3 levels or loss of MATR3 function contributes to neuronal defects. Our findings demonstrate that the MATR3 S85C mouse model mimics aspects of early-stage ALS and would be a promising tool for future basic and preclinical research.
Collapse
|
123
|
Yu CH, Davidson S, Harapas CR, Hilton JB, Mlodzianoski MJ, Laohamonthonkul P, Louis C, Low RRJ, Moecking J, De Nardo D, Balka KR, Calleja DJ, Moghaddas F, Ni E, McLean CA, Samson AL, Tyebji S, Tonkin CJ, Bye CR, Turner BJ, Pepin G, Gantier MP, Rogers KL, McArthur K, Crouch PJ, Masters SL. TDP-43 Triggers Mitochondrial DNA Release via mPTP to Activate cGAS/STING in ALS. Cell 2020; 183:636-649.e18. [PMID: 33031745 PMCID: PMC7599077 DOI: 10.1016/j.cell.2020.09.020] [Citation(s) in RCA: 602] [Impact Index Per Article: 120.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 07/21/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023]
Abstract
Cytoplasmic accumulation of TDP-43 is a disease hallmark for many cases of amyotrophic lateral sclerosis (ALS), associated with a neuroinflammatory cytokine profile related to upregulation of nuclear factor κB (NF-κB) and type I interferon (IFN) pathways. Here we show that this inflammation is driven by the cytoplasmic DNA sensor cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) when TDP-43 invades mitochondria and releases DNA via the permeability transition pore. Pharmacologic inhibition or genetic deletion of cGAS and its downstream signaling partner STING prevents upregulation of NF-κB and type I IFN induced by TDP-43 in induced pluripotent stem cell (iPSC)-derived motor neurons and in TDP-43 mutant mice. Finally, we document elevated levels of the specific cGAS signaling metabolite cGAMP in spinal cord samples from patients, which may be a biomarker of mtDNA release and cGAS/STING activation in ALS. Our results identify mtDNA release and cGAS/STING activation as critical determinants of TDP-43-associated pathology and demonstrate the potential for targeting this pathway in ALS. TDP-43 enters mitochondria, triggers mtDNA release via the mPTP TDP-43-induced cytosolic mtDNA accumulation activates the cGAS/STING pathway Evidence of cytoplasmic mtDNA was found in ALS patient cells and disease models Blocking STING prevents inflammation and neurodegeneration in vitro and in vivo
Collapse
Affiliation(s)
- Chien-Hsiung Yu
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sophia Davidson
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Cassandra R Harapas
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - James B Hilton
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC 3010, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael J Mlodzianoski
- Centre for Dynamic Imaging, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Pawat Laohamonthonkul
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Cynthia Louis
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ronnie Ren Jie Low
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jonas Moecking
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Institute of Structural Biology, University of Bonn, 53127 Bonn, Germany
| | - Dominic De Nardo
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia
| | - Katherine R Balka
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia
| | - Dale J Calleja
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Fiona Moghaddas
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Immunology and Allergy, The Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| | - Erya Ni
- Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Catriona A McLean
- Anatomical Pathology, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Andre L Samson
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Shiraz Tyebji
- Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Christopher J Tonkin
- Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Christopher R Bye
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Bradley J Turner
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Genevieve Pepin
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Michael P Gantier
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Kelly L Rogers
- Centre for Dynamic Imaging, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Kate McArthur
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia
| | - Peter J Crouch
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC 3010, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Seth L Masters
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Immunology Laboratory, Guangzhou Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou, Guangdong 510623, China.
| |
Collapse
|
124
|
Schieweck R, Ninkovic J, Kiebler MA. RNA-binding proteins balance brain function in health and disease. Physiol Rev 2020; 101:1309-1370. [PMID: 33000986 DOI: 10.1152/physrev.00047.2019] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Posttranscriptional gene expression including splicing, RNA transport, translation, and RNA decay provides an important regulatory layer in many if not all molecular pathways. Research in the last decades has positioned RNA-binding proteins (RBPs) right in the center of posttranscriptional gene regulation. Here, we propose interdependent networks of RBPs to regulate complex pathways within the central nervous system (CNS). These are involved in multiple aspects of neuronal development and functioning, including higher cognition. Therefore, it is not sufficient to unravel the individual contribution of a single RBP and its consequences but rather to study and understand the tight interplay between different RBPs. In this review, we summarize recent findings in the field of RBP biology and discuss the complex interplay between different RBPs. Second, we emphasize the underlying dynamics within an RBP network and how this might regulate key processes such as neurogenesis, synaptic transmission, and synaptic plasticity. Importantly, we envision that dysfunction of specific RBPs could lead to perturbation within the RBP network. This would have direct and indirect (compensatory) effects in mRNA binding and translational control leading to global changes in cellular expression programs in general and in synaptic plasticity in particular. Therefore, we focus on RBP dysfunction and how this might cause neuropsychiatric and neurodegenerative disorders. Based on recent findings, we propose that alterations in the entire regulatory RBP network might account for phenotypic dysfunctions observed in complex diseases including neurodegeneration, epilepsy, and autism spectrum disorders.
Collapse
Affiliation(s)
- Rico Schieweck
- Biomedical Center (BMC), Department for Cell Biology and Anatomy, Medical Faculty, Ludwig-Maximilians-University, Planegg-Martinsried, Germany
| | - Jovica Ninkovic
- Biomedical Center (BMC), Department for Cell Biology and Anatomy, Medical Faculty, Ludwig-Maximilians-University, Planegg-Martinsried, Germany
| | - Michael A Kiebler
- Biomedical Center (BMC), Department for Cell Biology and Anatomy, Medical Faculty, Ludwig-Maximilians-University, Planegg-Martinsried, Germany
| |
Collapse
|
125
|
Solomon DA, Mitchell JC, Salcher-Konrad MT, Vance CA, Mizielinska S. Review: Modelling the pathology and behaviour of frontotemporal dementia. Neuropathol Appl Neurobiol 2020; 45:58-80. [PMID: 30582188 DOI: 10.1111/nan.12536] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/16/2018] [Indexed: 12/11/2022]
Abstract
Frontotemporal dementia (FTD) encompasses a collection of clinically and pathologically diverse neurological disorders. Clinical features of behavioural and language dysfunction are associated with neurodegeneration, predominantly of frontal and temporal cortices. Over the past decade, there have been significant advances in the understanding of the genetic aetiology and neuropathology of FTD which have led to the creation of various disease models to investigate the molecular pathways that contribute to disease pathogenesis. The generation of in vivo models of FTD involves either targeting genes with known disease-causative mutations such as GRN and C9orf72 or genes encoding proteins that form the inclusions that characterize the disease pathologically, such as TDP-43 and FUS. This review provides a comprehensive summary of the different in vivo model systems used to understand pathomechanisms in FTD, with a focus on disease models which reproduce aspects of the wide-ranging behavioural phenotypes seen in people with FTD. We discuss the emerging disease pathways that have emerged from these in vivo models and how this has shaped our understanding of disease mechanisms underpinning FTD. We also discuss the challenges of modelling the complex clinical symptoms shown by people with FTD, the confounding overlap with features of motor neuron disease, and the drive to make models more disease-relevant. In summary, in vivo models can replicate many pathological and behavioural aspects of clinical FTD, but robust and thorough investigations utilizing shared features and variability between disease models will improve the disease-relevance of findings and thus better inform therapeutic development.
Collapse
Affiliation(s)
- D A Solomon
- UK Dementia Research Institute, King's College London, London, Camberwell, UK.,Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, Camberwell, UK
| | - J C Mitchell
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, Camberwell, UK
| | - M-T Salcher-Konrad
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, Camberwell, UK
| | - C A Vance
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, Camberwell, UK
| | - S Mizielinska
- UK Dementia Research Institute, King's College London, London, Camberwell, UK.,Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, Camberwell, UK
| |
Collapse
|
126
|
Neumann M, Mackenzie IRA. Review: Neuropathology of non-tau frontotemporal lobar degeneration. Neuropathol Appl Neurobiol 2020; 45:19-40. [PMID: 30357887 DOI: 10.1111/nan.12526] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/29/2018] [Indexed: 12/12/2022]
Abstract
Frontotemporal dementia (FTD) is a heterogeneous clinical syndrome associated with frontotemporal lobar degeneration (FTLD) as a relatively consistent neuropathological hallmark feature. However, the discoveries in the past decade of many of the relevant pathological proteins aggregating in human FTD brains in addition to several new FTD causing gene mutations underlined that FTD is a diverse condition on the neuropathological and genetic basis. This resulted in a novel molecular classification of these conditions based on the predominant protein abnormality and allows most cases of FTD to be placed now into one of three broad molecular subgroups; FTLD with tau, TAR DNA-binding protein 43 or FET protein accumulation (FTLD-tau, FTLD-TDP and FTLD-FET respectively). This review will provide an overview of the molecular neuropathology of non-tau FTLD, insights into disease mechanisms gained from the study of human post mortem tissue as well as discussion of current controversies in the field.
Collapse
Affiliation(s)
- M Neumann
- Department of Neuropathology, University Hospital of Tübingen, Tübingen, Germany.,Molecular Neuropathology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - I R A Mackenzie
- Department of Pathology, University of British Columbia and Vancouver General Hospital, Vancouver, BC, Canada
| |
Collapse
|
127
|
Mueller C, Berry JD, McKenna-Yasek DM, Gernoux G, Owegi MA, Pothier LM, Douthwright CL, Gelevski D, Luppino SD, Blackwood M, Wightman NS, Oakley DH, Frosch MP, Flotte TR, Cudkowicz ME, Brown RH. SOD1 Suppression with Adeno-Associated Virus and MicroRNA in Familial ALS. N Engl J Med 2020; 383:151-158. [PMID: 32640133 PMCID: PMC11836664 DOI: 10.1056/nejmoa2005056] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Two patients with familial amyotrophic lateral sclerosis (ALS) and mutations in the gene encoding superoxide dismutase 1 (SOD1) were treated with a single intrathecal infusion of adeno-associated virus encoding a microRNA targeting SOD1. In Patient 1, SOD1 levels in spinal cord tissue as analyzed on autopsy were lower than corresponding levels in untreated patients with SOD1-mediated ALS and in healthy controls. Levels of SOD1 in cerebrospinal fluid were transiently and only slightly lower in Patient 1 but were not affected in Patient 2. In Patient 1, meningoradiculitis developed after the infusion; Patient 2 was pretreated with immunosuppressive drugs and did not have this complication. Patient 1 had transient improvement in the strength of his right leg, a measure that had been relatively stable throughout his disease course, but there was no change in his vital capacity. Patient 2 had stable scores on a composite measure of ALS function and a stable vital capacity during a 12-month period. This study showed that intrathecal microRNA can be used as a potential treatment for SOD1-mediated ALS.
Collapse
Affiliation(s)
- Christian Mueller
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - James D Berry
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Diane M McKenna-Yasek
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Gwladys Gernoux
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Margaret A Owegi
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Lindsay M Pothier
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Catherine L Douthwright
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Dario Gelevski
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Sarah D Luppino
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Meghan Blackwood
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Nicholas S Wightman
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Derek H Oakley
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Matthew P Frosch
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Terrence R Flotte
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Merit E Cudkowicz
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| | - Robert H Brown
- From the Horae Gene Therapy Center, Department of Pediatrics, University of Massachusetts Medical School (UMMS) (C.M., G.G., M.B., T.R.F.), and the Department of Neurology, UMMS and UMass Memorial Medical Center (D.M.M.-Y., M.A.O., C.L.D., N.S.W., R.H.B.), Worcester, and the Healey Center for ALS, Department of Neurology (J.D.B., L.M.P., D.G., S.D.L., M.P.F., M.E.C.), and the C.S. Kubik Laboratory for Neuropathology (D.H.O., M.P.F.), Massachusetts General Hospital and Harvard Medical School, Boston
| |
Collapse
|
128
|
Humphrey J, Birsa N, Milioto C, McLaughlin M, Ule AM, Robaldo D, Eberle AB, Kräuchi R, Bentham M, Brown AL, Jarvis S, Bodo C, Garone M, Devoy A, Soraru G, Rosa A, Bozzoni I, Fisher EMC, Mühlemann O, Schiavo G, Ruepp MD, Isaacs AM, Plagnol V, Fratta P. FUS ALS-causative mutations impair FUS autoregulation and splicing factor networks through intron retention. Nucleic Acids Res 2020; 48:6889-6905. [PMID: 32479602 PMCID: PMC7337901 DOI: 10.1093/nar/gkaa410] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 04/21/2020] [Accepted: 05/06/2020] [Indexed: 12/12/2022] Open
Abstract
Mutations in the RNA-binding protein FUS cause amyotrophic lateral sclerosis (ALS), a devastating neurodegenerative disease. FUS plays a role in numerous aspects of RNA metabolism, including mRNA splicing. However, the impact of ALS-causative mutations on splicing has not been fully characterized, as most disease models have been based on overexpressing mutant FUS, which will alter RNA processing due to FUS autoregulation. We and others have recently created knockin models that overcome the overexpression problem, and have generated high depth RNA-sequencing on FUS mutants in parallel to FUS knockout, allowing us to compare mutation-induced changes to genuine loss of function. We find that FUS-ALS mutations induce a widespread loss of function on expression and splicing. Specifically, we find that mutant FUS directly alters intron retention levels in RNA-binding proteins. Moreover, we identify an intron retention event in FUS itself that is associated with its autoregulation. Altered FUS levels have been linked to disease, and we show here that this novel autoregulation mechanism is altered by FUS mutations. Crucially, we also observe this phenomenon in other genetic forms of ALS, including those caused by TDP-43, VCP and SOD1 mutations, supporting the concept that multiple ALS genes interact in a regulatory network.
Collapse
Affiliation(s)
- Jack Humphrey
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
| | - Nicol Birsa
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute
| | - Carmelo Milioto
- UK Dementia Research Institute
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Martha McLaughlin
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Agnieszka M Ule
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - David Robaldo
- UK Dementia Research Institute
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Andrea B Eberle
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, Bern 3012, Switzerland
| | - Rahel Kräuchi
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, Bern 3012, Switzerland
| | - Matthew Bentham
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Anna-Leigh Brown
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
| | - Seth Jarvis
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
| | - Cristian Bodo
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | | | - Anny Devoy
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London SE5 9RT, UK
| | - Gianni Soraru
- Department of Neurosciences, Università degli Studi di Padova, Padova 35121, Italy
| | - Alessandro Rosa
- Sapienza University of Rome, Rome 00185, Italy
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome 00161, Italy
| | - Irene Bozzoni
- Sapienza University of Rome, Rome 00185, Italy
- Center for Life Nano Science, Istituto Italiano di Tecnologia, Rome 00161, Italy
| | - Elizabeth M C Fisher
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Oliver Mühlemann
- Department of Chemistry and Biochemistry, University of Bern, Freiestrasse 3, Bern 3012, Switzerland
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute
- Discoveries Centre for Regenerative and Precision Medicine, University College London Campus, London WC1N 3BG, UK
| | - Marc-David Ruepp
- UK Dementia Research Institute
- Maurice Wohl Clinical Neuroscience Institute, King’s College London, London SE5 9RT, UK
| | - Adrian M Isaacs
- UK Dementia Research Institute
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Vincent Plagnol
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
| | - Pietro Fratta
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
129
|
Fisher EMC, Bannerman DM. Mouse models of neurodegeneration: Know your question, know your mouse. Sci Transl Med 2020; 11:11/493/eaaq1818. [PMID: 31118292 DOI: 10.1126/scitranslmed.aaq1818] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 02/26/2018] [Accepted: 06/19/2018] [Indexed: 12/19/2022]
Abstract
Many mutant mouse strains have been developed as models to investigate neurodegenerative disease in humans. However, variability in results among studies using these mouse strains has led to questions about the value of these models. Here, we appraise various mouse models for dissecting neurodegenerative disease mechanisms and emphasize the importance of asking appropriate research questions. In therapeutic studies, we suggest that understanding variability among and within mouse models is crucial for preventing translational failures in human patients.
Collapse
Affiliation(s)
- Elizabeth M C Fisher
- Department of Neuromuscular Diseases, University College London, London WC1N 3BG, UK.
| | - David M Bannerman
- Department of Experimental Psychology, University of Oxford, Oxford OX1 3TA, UK.
| |
Collapse
|
130
|
Bendotti C, Bonetto V, Pupillo E, Logroscino G, Al-Chalabi A, Lunetta C, Riva N, Mora G, Lauria G, Weishaupt JH, Agosta F, Malaspina A, Basso M, Greensmith L, Van Den Bosch L, Ratti A, Corbo M, Hardiman O, Chiò A, Silani V, Beghi E. Focus on the heterogeneity of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:485-495. [PMID: 32583689 DOI: 10.1080/21678421.2020.1779298] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The clinical manifestations of amyotrophic lateral sclerosis (ALS) are variable in terms of age at disease onset, site of onset, progression of symptoms, motor neuron involvement, and the occurrence of cognitive and behavioral changes. Genetic background is a key determinant of the ALS phenotype. The mortality of the disease also varies with the ancestral origin of the affected population and environmental factors are likely to be associated with ALS at least within some cohorts. Disease heterogeneity is likely underpinned by the presence of different pathogenic mechanisms. A variety of ALS animal models can be informative about the heterogeneity of the neuropathological or genetic aspects of the disease and can support the development of new therapeutic intervention. Evolving biomarkers can contribute to the identification of differing genotypes and phenotypes, and can be used to explore whether genotypic and phenotypic differences in animal models might help to provide a better definition of the heterogeneity of ALS in humans. These include neurofilaments, peripheral blood mononuclear cells, extracellular vesicles, microRNA and imaging findings. These biomarkers might predict not only the development of the disease, but also the variability in progression, although robust validation is required. A promising area of progress in modeling the heterogeneity of human ALS is represented by the use of human induced pluripotent stem cell (iPSCs)-derived motor neurons. Although the translational value of iPSCs remains unclear, this model is attractive in the perspective of replicating the heterogeneity of sporadic ALS as a first step toward a personalized medicine strategy.
Collapse
Affiliation(s)
- Caterina Bendotti
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Valentina Bonetto
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Elisabetta Pupillo
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Giancarlo Logroscino
- Department of Neurosciences and Sense Organs, Center for Neurodegenerative Diseases and the Aging Brain Università degli Studi di Bari, Bari; Fondazione Giovanni Panico Tricase, Lecce, Italy
| | - Ammar Al-Chalabi
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Christian Lunetta
- NEuroMuscular Omnicentre (NEMO), Serena Onlus Foundation, Milano, Italy
| | - Nilo Riva
- Neuroimaging Research Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milano, Italy
| | - Gabriela Mora
- Department of Neurorehabilitation, ICS Maugeri IRCCS, Milano, Italy
| | - Giuseppe Lauria
- Unit of Neurology, Motor Neuron Disease Center, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy.,Department of Biomedical and Clinical Sciences "Lduigi Sacco", University of Milan, Milan, Italy
| | | | - Federica Agosta
- Neuroimaging Research Unit, Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milano, Italy
| | | | - Manuela Basso
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.,Department of Cellular, Computational and Integrative Biology (CIBIO), Università degli Studi di Trento, Trento, Italy
| | - Linda Greensmith
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Ludo Van Den Bosch
- Center for Brain & Disease Research (VIB) and Laboratory of Neurobiology (KU Leuven), Leuven, Belgium
| | - Antonia Ratti
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, Milano, Italy.,Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico (CCP), Milano, Italy
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Adriano Chiò
- "Rita Levi Montalcini" Department of Neuroscience, Università degli Studi di Torino, Torino, Italy
| | - Vincenzo Silani
- Department of Neurology - Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, Milano, Italy.,Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milano, Italy
| | - Ettore Beghi
- Mario Negri-ALS Study Group, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| |
Collapse
|
131
|
McGurk L, Rifai OM, Bonini NM. TDP-43, a protein central to amyotrophic lateral sclerosis, is destabilized by tankyrase-1 and -2. J Cell Sci 2020; 133:jcs245811. [PMID: 32409565 PMCID: PMC7328137 DOI: 10.1242/jcs.245811] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022] Open
Abstract
In >95% of cases of amyotrophic lateral sclerosis (ALS) and ∼45% of frontotemporal degeneration (FTD), the RNA/DNA-binding protein TDP-43 is cleared from the nucleus and abnormally accumulates in the cytoplasm of affected brain cells. Although the cellular triggers of disease pathology remain enigmatic, mounting evidence implicates the poly(ADP-ribose) polymerases (PARPs) in TDP-43 neurotoxicity. Here we show that inhibition of the PARP enzymes tankyrase 1 and tankyrase 2 (referred to as Tnks-1/2) protect primary rodent neurons from TDP-43-associated neurotoxicity. We demonstrate that Tnks-1/2 interacts with TDP-43 via a newly defined tankyrase-binding domain. Upon investigating the functional effect, we find that interaction with Tnks-1/2 inhibits the ubiquitination and proteasomal turnover of TDP-43, leading to its stabilization. We further show that proteasomal turnover of TDP-43 occurs preferentially in the nucleus; our data indicate that Tnks-1/2 stabilizes TDP-43 by promoting cytoplasmic accumulation, which sequesters the protein from nuclear proteasome degradation. Thus, Tnks-1/2 activity modulates TDP-43 and is a potential therapeutic target in diseases associated with TDP-43, such as ALS and FTD.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Leeanne McGurk
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Olivia M Rifai
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy M Bonini
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
132
|
Flores BN, Li X, Malik AM, Martinez J, Beg AA, Barmada SJ. An Intramolecular Salt Bridge Linking TDP43 RNA Binding, Protein Stability, and TDP43-Dependent Neurodegeneration. Cell Rep 2020; 27:1133-1150.e8. [PMID: 31018129 PMCID: PMC6499398 DOI: 10.1016/j.celrep.2019.03.093] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/28/2019] [Accepted: 03/25/2019] [Indexed: 12/13/2022] Open
Abstract
The majority of individuals with amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) exhibit neuronal cytoplasmic inclusions rich in the RNA binding protein TDP43. Even so, the relation between the RNA binding properties of TDP43 and neurodegeneration remains obscure. Here, we show that engineered mutations disrupting a salt bridge between the RNA recognition motifs of TDP43 interfere with RNA binding and eliminate the recognition of native TDP43 substrates. The same mutations dramatically destabilize TDP43, alter its subcellular localization, and abrogate TDP43-dependent neuro-degeneration. Worms harboring homologous TDP-1 mutations phenocopy knockout strains, confirming the necessity of salt bridge residues for TDP43 function. Moreover, the accumulation of functional TDP43, but not RNA binding-deficient variants, disproportionately affects transcripts encoding ribo-some and oxidative phosphorylation components. These studies demonstrate the significance of the salt bridge in sustaining TDP43 stability and RNA binding properties, factors that are crucial for neurodegeneration arising from TDP43 deposition in ALS and FTD. Flores et al. uncover essential roles for an intramolecular salt bridge in the function of TDP43, an RNA binding protein implicated in neurodegenerative diseases. Salt bridge interruption attenuates TDP43 RNA binding affinity and specificity, destabilizes the protein, and prevents TDP43-mediated neurotoxicity arising from misprocessing of ribosomal and mitochondrial transcripts.
Collapse
Affiliation(s)
- Brittany N Flores
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48104, USA; Department of Neurology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Xingli Li
- Department of Neurology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Ahmed M Malik
- Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48104, USA; Neuroscience Graduate Program, Department of Pharmacology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Jose Martinez
- Neuroscience Graduate Program, Department of Pharmacology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Asim A Beg
- Neuroscience Graduate Program, Department of Pharmacology, University of Michigan, Ann Arbor, MI 48104, USA
| | - Sami J Barmada
- Cellular and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI 48104, USA; Department of Neurology, University of Michigan, Ann Arbor, MI 48104, USA; Neuroscience Graduate Program, Department of Pharmacology, University of Michigan, Ann Arbor, MI 48104, USA.
| |
Collapse
|
133
|
Liscic RM, Alberici A, Cairns NJ, Romano M, Buratti E. From basic research to the clinic: innovative therapies for ALS and FTD in the pipeline. Mol Neurodegener 2020; 15:31. [PMID: 32487123 PMCID: PMC7268618 DOI: 10.1186/s13024-020-00373-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and Frontotemporal Degeneration (FTD) are neurodegenerative disorders, related by deterioration of motor and cognitive functions and short survival. Aside from cases with an inherited pathogenic mutation, the causes of the disorders are still largely unknown and no effective treatment currently exists. It has been shown that FTD may coexist with ALS and this overlap occurs at clinical, genetic, and molecular levels. In this work, we review the main pathological aspects of these complex diseases and discuss how the integration of the novel pathogenic molecular insights and the analysis of molecular interaction networks among all the genetic players represents a critical step to shed light on discovering novel therapeutic strategies and possibly tailoring personalized medicine approaches to specific ALS and FTD patients.
Collapse
Affiliation(s)
- Rajka Maria Liscic
- Department of Neurology, Johannes Kepler University, Linz, Austria
- School of Medicine, University of Osijek, Osijek, Croatia
| | - Antonella Alberici
- Neurology Unit, Department of Neurological Sciences and Vision, ASST-Spedali Civili-University of Brescia, Brescia, Italy
| | - Nigel John Cairns
- College of Medicine and Health and Living Systems Institute, University of Exeter, Exeter, UK
| | - Maurizio Romano
- Department of Life Sciences, Via Valerio 28, University of Trieste, 34127, Trieste, Italy
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149, Trieste, Italy.
| |
Collapse
|
134
|
Lee JH, Cho SY, Kim E. Translational cognitive neuroscience of dementia with touchscreen operant chambers. GENES BRAIN AND BEHAVIOR 2020; 20:e12664. [PMID: 32374080 DOI: 10.1111/gbb.12664] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 04/29/2020] [Indexed: 12/20/2022]
Abstract
Translational cognitive neuroscience of dementia involves mainly two areas: the validation of newly developed dementia animal models and the preclinical assessment of novel drug candidates in such model animals. To validate new animal models, a multidomain panel (battery) approach is essential in that dementia is, by definition, not merely a memory disorder but rather a multidomain cognitive/behavior disorder: animal modeling with a certain type of dementia would develop cognitive impairments in multiple (two at minimum) domains in a specific order according to unique spreading patterns of its neuropathology. In new drug development, the availability of highly sensitive tools assessing animal cognition is crucial to the detection of cognitive decline at the earliest stage of the disease, which may be an optimal time point to test a drug candidate. Using interspecies translatable (analogous) cognitive tasks would also be necessary to successfully predict the efficacy of drug candidates in subsequent clinical trials. Currently, this translational prediction is seriously limited given discrepancies in behavioral assessment methods between animals and humans in the preclinical and clinical trials, respectively. Since neurodegenerative diseases are often accompanied by not only cognitive but also affective and movement disorders, simultaneous assessment of task-relevant locomotor behavior and motivation is also important to rule out the effects of potential confounders. The touchscreen operant platform may satisfy these needs by offering several advantages over conventional methodology. In this review, we discuss the touchscreen operant chamber system and highlight some of its qualities as a promising and desirable tool for translational research of dementia.
Collapse
Affiliation(s)
- Ji Han Lee
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Yeon Cho
- Department of Psychiatry, Institute of Behavioral Science in Medicine, Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eosu Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Psychiatry, Institute of Behavioral Science in Medicine, Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
135
|
Wong P, Ho WY, Yen YC, Sanford E, Ling SC. The vulnerability of motor and frontal cortex-dependent behaviors in mice expressing ALS-linked mutation in TDP-43. Neurobiol Aging 2020; 92:43-60. [PMID: 32422502 DOI: 10.1016/j.neurobiolaging.2020.03.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/18/2020] [Accepted: 03/29/2020] [Indexed: 02/01/2023]
Abstract
TDP-43 aggregates are the defining pathological hallmark for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Strikingly, these TDP-43 proteinopathies are also found in other neurodegenerative diseases, including Alzheimer's disease and are prevalent in the brains of old-aged humans. Furthermore, disease-causal mutations in TDP-43 have been identified for ALS and FTD. Collectively, the evidence indicates that TDP-43 dysfunctions lead to motor and cognitive deficits. To determine whether the mouse line expressing an ALS-linked mutation in TDP-43 (Q331K) can be used to study ALS-FTD spectrum disorders, we performed a systematic and longitudinal behavioral assessment that covered motor and cognitive functions. Deficits in motor and cognitive abilities were observed as early as 3 months of age and persisted through to 12 months of age. Within the cognitive modalities, the hippocampus-mediated spatial learning and memory, and contextual fear conditioning, were normal; whereas the frontal cortex-mediated working memory and cognitive flexibility were impaired. Biochemically, the human TDP-43 transgene downregulates endogenous mouse TDP-43 mRNA and protein, resulting in human TDP-43 protein that is comparable with the physiological level in cerebral cortex and hippocampus. Furthermore, Q331K TDP-43 is largely retained at the nucleus without apparent aggregates. Taken together, our data suggest that motor and frontal cortex may be more vulnerable to disease-linked mutation in TDP-43 and, this mouse model may be used to assess ALS-FTD-related spectrum diseases and the molecular underpinnings associated with the phenotypes.
Collapse
Affiliation(s)
- Peiyan Wong
- Department of Pharmacology, National University of Singapore, Singapore
| | - Wan Yun Ho
- Department of Physiology, National University of Singapore, Singapore
| | - Yi-Chun Yen
- Department of Physiology, National University of Singapore, Singapore
| | - Emma Sanford
- Department of Physiology, National University of Singapore, Singapore
| | - Shuo-Chien Ling
- Department of Physiology, National University of Singapore, Singapore; Department of Neurobiology/Ageing Programme, National University of Singapore, Singapore; Program in Neuroscience and Behavior Disorders, Duke-NUS Medical School, Singapore.
| |
Collapse
|
136
|
Sleigh JN, Tosolini AP, Gordon D, Devoy A, Fratta P, Fisher EMC, Talbot K, Schiavo G. Mice Carrying ALS Mutant TDP-43, but Not Mutant FUS, Display In Vivo Defects in Axonal Transport of Signaling Endosomes. Cell Rep 2020; 30:3655-3662.e2. [PMID: 32187538 PMCID: PMC7090381 DOI: 10.1016/j.celrep.2020.02.078] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 10/11/2019] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, progressive neurodegenerative disease resulting from a complex interplay between genetics and environment. Impairments in axonal transport have been identified in several ALS models, but in vivo evidence remains limited, thus their pathogenetic importance remains to be fully resolved. We therefore analyzed the in vivo dynamics of retrogradely transported, neurotrophin-containing signaling endosomes in nerve axons of two ALS mouse models with mutations in the RNA processing genes TARDBP and FUS. TDP-43M337V mice, which show neuromuscular pathology without motor neuron loss, display axonal transport perturbations manifesting between 1.5 and 3 months and preceding symptom onset. Contrastingly, despite 20% motor neuron loss, transport remained largely unaffected in FusΔ14/+ mice. Deficiencies in retrograde axonal transport of signaling endosomes are therefore not shared by all ALS-linked genes, indicating that there are mechanistic distinctions in the pathogenesis of ALS caused by mutations in different RNA processing genes.
Collapse
Affiliation(s)
- James N Sleigh
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK; UK Dementia Research Institute, University College London, London WC1E 6BT, UK.
| | - Andrew P Tosolini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - David Gordon
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Anny Devoy
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Pietro Fratta
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Elizabeth M C Fisher
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK; UK Dementia Research Institute, University College London, London WC1E 6BT, UK; Discoveries Centre for Regenerative and Precision Medicine, University College London Campus, London WC1N 3BG, UK.
| |
Collapse
|
137
|
Motor neuron preservation and decrease of in vivo TDP-43 phosphorylation by protein CK-1δ kinase inhibitor treatment. Sci Rep 2020; 10:4449. [PMID: 32157143 PMCID: PMC7064575 DOI: 10.1038/s41598-020-61265-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/25/2020] [Indexed: 01/06/2023] Open
Abstract
Pathogenesis of amyotrophic lateral sclerosis (ALS), a devastating disease where no treatment exists, involves the compartmentalization of the nuclear protein TDP-43 (TAR DNA-binding protein 43) in the cytoplasm which is promoted by its aberrant phosphorylation and others posttranslational modifications. Recently, it was reported that CK-1δ (protein casein kinase-1δ) is able to phosphorylate TDP-43. Here, the preclinical efficacy of a benzothiazole-based CK-1δ inhibitor IGS-2.7, both in a TDP-43 (A315T) transgenic mouse and in a human cell-based model of ALS, is shown. Treatment with IGS-2.7 produces a significant preservation of motor neurons in the anterior horn at lumbar level, a decrease in both astroglial and microglial reactivity in this area, and in TDP-43 phosphorylation in spinal cord samples. Furthermore, the recovery of TDP-43 homeostasis (phosphorylation and localization) in a human-based cell model from ALS patients after treatment with IGS-2.7 is also reported. Moreover, we have shown a trend to increase in CK-1δ mRNA in spinal cord and significantly in frontal cortex of sALS cases. All these data show for the first time the in vivo modulation of TDP-43 toxicity by CK-1δ inhibition with IGS-2.7, which may explain the benefits in the preservation of spinal motor neurons and point to the relevance of CK-1δ inhibitors in a future disease-modifying treatment for ALS.
Collapse
|
138
|
Picchiarelli G, Dupuis L. Role of RNA Binding Proteins with prion-like domains in muscle and neuromuscular diseases. Cell Stress 2020; 4:76-91. [PMID: 32292882 PMCID: PMC7146060 DOI: 10.15698/cst2020.04.217] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A number of neuromuscular and muscular diseases, including amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA) and several myopathies, are associated to mutations in related RNA-binding proteins (RBPs), including TDP-43, FUS, MATR3 or hnRNPA1/B2. These proteins harbor similar modular primary sequence with RNA binding motifs and low complexity domains, that enables them to phase separate and create liquid microdomains. These RBPs have been shown to critically regulate multiple events of RNA lifecycle, including transcriptional events, splicing and RNA trafficking and sequestration. Here, we review the roles of these disease-related RBPs in muscle and motor neurons, and how their dysfunction in these cell types might contribute to disease.
Collapse
Affiliation(s)
- Gina Picchiarelli
- Université de Strasbourg, INSERM, Mécanismes Centraux et Périphériques de la Neurodégénérescence, UMR_S 1118, Strasbourg, France
| | - Luc Dupuis
- Université de Strasbourg, INSERM, Mécanismes Centraux et Périphériques de la Neurodégénérescence, UMR_S 1118, Strasbourg, France
| |
Collapse
|
139
|
Weskamp K, Tank EM, Miguez R, McBride JP, Gómez NB, White M, Lin Z, Gonzalez CM, Serio A, Sreedharan J, Barmada SJ. Shortened TDP43 isoforms upregulated by neuronal hyperactivity drive TDP43 pathology in ALS. J Clin Invest 2020; 130:1139-1155. [PMID: 31714900 PMCID: PMC7269575 DOI: 10.1172/jci130988] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
Cortical hyperexcitability and mislocalization of the RNA-binding protein TDP43 are highly conserved features in amyotrophic lateral sclerosis (ALS). Nevertheless, the relationship between these phenomena remains poorly defined. Here, we showed that hyperexcitability recapitulates TDP43 pathology by upregulating shortened TDP43 (sTDP43) splice isoforms. These truncated isoforms accumulated in the cytoplasm and formed insoluble inclusions that sequestered full-length TDP43 via preserved N-terminal interactions. Consistent with these findings, sTDP43 overexpression was toxic to mammalian neurons, suggesting neurodegeneration arising from complementary gain- and loss-of-function mechanisms. In humans and mice, sTDP43 transcripts were enriched in vulnerable motor neurons, and we observed a striking accumulation of sTDP43 within neurons and glia of ALS patients. Collectively, these studies uncover a pathogenic role for alternative TDP43 isoforms in ALS, and implicate sTDP43 as a key contributor to the susceptibility of motor neurons in this disorder.
Collapse
Affiliation(s)
| | | | | | - Jonathon P. McBride
- Department of Neurology
- Cellular and Molecular Biology Program, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicolás B. Gómez
- Department of Neurology
- Cellular and Molecular Biology Program, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Ziqiang Lin
- Department of Basic and Clinical Neuroscience and
| | - Carmen Moreno Gonzalez
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
| | - Andrea Serio
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, United Kingdom
| | | | - Sami J. Barmada
- Department of Neurology
- Neuroscience Graduate Program, and
- Cellular and Molecular Biology Program, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
140
|
Asakawa K, Handa H, Kawakami K. Optogenetic modulation of TDP-43 oligomerization accelerates ALS-related pathologies in the spinal motor neurons. Nat Commun 2020; 11:1004. [PMID: 32081878 PMCID: PMC7035286 DOI: 10.1038/s41467-020-14815-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 02/05/2020] [Indexed: 11/19/2022] Open
Abstract
Cytoplasmic aggregation of TDP-43 characterizes degenerating neurons in most cases of amyotrophic lateral sclerosis (ALS). Here, we develop an optogenetic TDP-43 variant (opTDP-43), whose multimerization status can be modulated in vivo through external light illumination. Using the translucent zebrafish neuromuscular system, we demonstrate that short-term light stimulation reversibly induces cytoplasmic opTDP-43 mislocalization, but not aggregation, in the spinal motor neuron, leading to an axon outgrowth defect associated with myofiber denervation. In contrast, opTDP-43 forms pathological aggregates in the cytoplasm after longer-term illumination and seeds non-optogenetic TDP-43 aggregation. Furthermore, we find that an ALS-linked mutation in the intrinsically disordered region (IDR) exacerbates the light-dependent opTDP-43 toxicity on locomotor behavior. Together, our results propose that IDR-mediated TDP-43 oligomerization triggers both acute and long-term pathologies of motor neurons, which may be relevant to the pathogenesis and progression of ALS. Optogenetic approaches for inducing TDP-43 aggregation have been described previously in cellular models. Here the authors develop an approach to optogenetically induce TDP-43 aggregation in vivo using zebrafish to model ALS pathologies.
Collapse
Affiliation(s)
- Kazuhide Asakawa
- Department of Chemical Biology, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan. .,Division of Molecular and Developmental Biology, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan. .,Department of Genetics, Graduate University for Advanced Studies (SOKENDAI), 1111 Yata, Mishima, Shizuoka, 411-8540, Japan.
| | - Hiroshi Handa
- Department of Chemical Biology, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, 1111 Yata, Mishima, Shizuoka, 411-8540, Japan. .,Department of Genetics, Graduate University for Advanced Studies (SOKENDAI), 1111 Yata, Mishima, Shizuoka, 411-8540, Japan.
| |
Collapse
|
141
|
Abramzon YA, Fratta P, Traynor BJ, Chia R. The Overlapping Genetics of Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Front Neurosci 2020; 14:42. [PMID: 32116499 PMCID: PMC7012787 DOI: 10.3389/fnins.2020.00042] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two diseases that form a broad neurodegenerative continuum. Considerable effort has been made to unravel the genetics of these disorders, and, based on this work, it is now clear that ALS and FTD have a significant genetic overlap. TARDBP, SQSTM1, VCP, FUS, TBK1, CHCHD10, and most importantly C9orf72, are the critical genetic players in these neurological disorders. Discoveries of these genes have implicated autophagy, RNA regulation, and vesicle and inclusion formation as the central pathways involved in neurodegeneration. Here we provide a summary of the significant genes identified in these two intrinsically linked neurodegenerative diseases and highlight the genetic and pathological overlaps.
Collapse
Affiliation(s)
- Yevgeniya A. Abramzon
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD, United States
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, United Kingdom
| | - Pietro Fratta
- Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, United Kingdom
| | - Bryan J. Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD, United States
- Department of Neurology, Brain Science Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Ruth Chia
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD, United States
| |
Collapse
|
142
|
Huang SL, Wu LS, Lee M, Chang CW, Cheng WC, Fang YS, Chen YR, Cheng PL, Shen CKJ. A robust TDP-43 knock-in mouse model of ALS. Acta Neuropathol Commun 2020; 8:3. [PMID: 31964415 PMCID: PMC6975031 DOI: 10.1186/s40478-020-0881-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, adult-onset degenerative disorder of motor neurons. The diseased spinal cord motor neurons of more than 95% of amyotrophic lateral sclerosis (ALS) patients are characterized by the mis-metabolism of the RNA/DNA-binding protein TDP-43 (ALS-TDP), in particular, the presence of cytosolic aggregates of the protein. Most available mouse models for the basic or translational studies of ALS-TDP are based on transgenic overexpression of the TDP-43 protein. Here, we report the generation and characterization of mouse lines bearing homologous knock-in of fALS-associated mutation A315T and sALS-associated mutation N390D, respectively. Remarkably, the heterozygous TDP-43 (N390D/+) mice but not those heterozygous for the TDP-43 (A315T/+) mice develop a full spectrum of ALS-TDP-like pathologies at the molecular, cellular and behavioral levels. Comparative analysis of the mutant mice and spinal cord motor neurons (MN) derived from their embryonic stem (ES) cells demonstrates that different ALS-associated TDP-43 mutations possess critical ALS-causing capabilities and pathogenic pathways, likely modified by their genetic background and the environmental factors. Mechanistically, we identify aberrant RNA splicing of spinal cord Bcl-2 pre-mRNA and consequent increase of a negative regulator of autophagy, Bcl-2, which correlate with and are caused by a progressive increase of TDP-43, one of the early events associated with ALS-TDP pathogenesis, in the spinal cord of TDP-43 (N390D/+) mice and spinal cord MN derived from their ES cells. The TDP-43 (N390D/+) knock-in mice appear to be an ideal rodent model for basic as well as translational studies of ALS- TDP.
Collapse
|
143
|
Watanabe S, Oiwa K, Murata Y, Komine O, Sobue A, Endo F, Takahashi E, Yamanaka K. ALS-linked TDP-43 M337V knock-in mice exhibit splicing deregulation without neurodegeneration. Mol Brain 2020; 13:8. [PMID: 31959210 PMCID: PMC6971932 DOI: 10.1186/s13041-020-0550-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/12/2020] [Indexed: 11/10/2022] Open
Abstract
Abnormal accumulation of TAR DNA-binding protein 43 (TDP-43), a DNA/RNA binding protein, is a pathological signature of amyotrophic lateral sclerosis (ALS). Missense mutations in the TARDBP gene are also found in inherited and sporadic ALS, indicating that dysfunction in TDP-43 is causative for ALS. To model TDP-43-linked ALS in rodents, we generated TDP-43 knock-in mice with inherited ALS patient-derived TDP-43M337V mutation. Homozygous TDP-43M337V mice developed normally without exhibiting detectable motor dysfunction and neurodegeneration. However, splicing of mRNAs regulated by TDP-43 was deregulated in the spinal cords of TDP-43M337V mice. Together with the recently reported TDP-43 knock-in mice with ALS-linked mutations, our finding indicates that ALS patient-derived mutations in the TARDBP gene at a carboxyl-terminal domain of TDP-43 may cause a gain of splicing function by TDP-43, however, were insufficient to induce robust neurodegeneration in mice.
Collapse
Affiliation(s)
- Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan.,Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Kotaro Oiwa
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan.,Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Aichi, 466-8550, Japan
| | - Yuri Murata
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Okiru Komine
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan.,Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Akira Sobue
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan.,Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Fumito Endo
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan.,Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan
| | - Eiki Takahashi
- Support Unit for Animal Resources Development, Research Resource Division, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, 464-8601, Japan. .,Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
144
|
Hawrot J, Imhof S, Wainger BJ. Modeling cell-autonomous motor neuron phenotypes in ALS using iPSCs. Neurobiol Dis 2019; 134:104680. [PMID: 31759135 DOI: 10.1016/j.nbd.2019.104680] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 10/29/2019] [Accepted: 11/15/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an aggressive and uniformly fatal degenerative disease of the motor nervous system. In order to understand underlying disease mechanisms, researchers leverage a host of in vivo and in vitro models, including yeast, worms, flies, zebrafish, mice, and more recently, human induced pluripotent stem cells (iPSCs) derived from ALS patients. While mouse models have been the main workhorse of preclinical ALS research, the development of iPSCs provides a new opportunity to explore molecular phenotypes of ALS within human cells. Importantly, this technology enables modeling of both familial and sporadic ALS in the relevant human genetic backgrounds, as well as a personalized or targeted approach to therapy development. Harnessing these powerful tools requires addressing numerous challenges, including different variance components associated with iPSCs and motor neurons as well as concomitant limits of reductionist approaches. In order to overcome these obstacles, optimization of protocols and assays, confirmation of phenotype robustness at scale, and validation of findings in human tissue and genetics will cement the role for iPSC models as a valuable complement to animal models in ALS and more broadly among neurodegenerative diseases.
Collapse
Affiliation(s)
- James Hawrot
- Departments of Neurology and Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sophie Imhof
- Departments of Neurology and Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; University of Amsterdam, Amsterdam, The Netherlands
| | - Brian J Wainger
- Departments of Neurology and Anesthesia, Critical Care & Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Broad Institute of Harvard University and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
145
|
St Martin JL, Wang L, Kaprielian Z. Toxicity in ALS: TDP-43 modifiers and C9orf72. Neurosci Lett 2019; 716:134621. [PMID: 31726180 DOI: 10.1016/j.neulet.2019.134621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 10/24/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating and fatal neurodegenerative disease affecting approximately 30,000 individuals in the United States. The average age of onset is 55 years and progression of the disease is rapid with most patients dying of respiratory failure within 3-5 years. Currently available therapeutics have modest effects on patient survival, underscoring the immediate need for more effective medicines. Recent technological advances in next generation sequencing have led to a substantial uptick in the discovery of genes linked to ALS. Since 90 % of ALS cases are sporadic, risk genes identified in familial cases provide invaluable insights into the molecular pathogenesis of the disease. Most notably, TDP-43-expressing neuronal inclusions and C9orf72 mutations have emerged as the key pathological and genetic hallmarks, respectively, of ALS. In this review, we will discuss recent advances in modifiers of TDP-43 toxicity, with an emphasis on Ataxin-2, one of the most well-characterized TDP-43 modifiers. An understanding of Ataxin-2 function and related biological pathways could provide a framework for the discovery of other novel modifiers of TDP-43. We will also describe the pathogenic mechanisms underlying C9orf72 toxicity and how these impact the disease process. Finally, we will explore emerging therapeutic strategies for dampening TDP-43 and C9orf72 toxicity and, ultimately, slowing or halting the progression of ALS.
Collapse
Affiliation(s)
| | - Lina Wang
- Amgen, Neuroscience Discovery, Cambridge, MA, United States
| | - Zaven Kaprielian
- Dementia Discovery Foundation US Discovery, Boston, United States.
| |
Collapse
|
146
|
Splicing repression is a major function of TDP-43 in motor neurons. Acta Neuropathol 2019; 138:813-826. [PMID: 31332509 DOI: 10.1007/s00401-019-02042-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/07/2019] [Accepted: 07/07/2019] [Indexed: 02/08/2023]
Abstract
Nuclear depletion of TDP-43, an essential RNA binding protein, may underlie neurodegeneration in amyotrophic lateral sclerosis (ALS). As several functions have been ascribed to this protein, the critical role(s) of TDP-43 in motor neurons that may be compromised in ALS remains unknown. We show here that TDP-43 mediated splicing repression, which serves to protect the transcriptome by preventing aberrant splicing, is central to the physiology of motor neurons. Expression in Drosophila TDP-43 knockout models of a chimeric repressor, comprised of the RNA recognition domain of TDP-43 fused to an unrelated splicing repressor, RAVER1, attenuated motor deficits and extended lifespan. Likewise, AAV9-mediated delivery of this chimeric rescue repressor to mice lacking TDP-43 in motor neurons delayed the onset, slowed the progression of motor symptoms, and markedly extended their lifespan. In treated mice lacking TDP-43 in motor neurons, aberrant splicing was significantly decreased and accompanied by amelioration of axon degeneration and motor neuron loss. This AAV9 strategy allowed long-term expression of the chimeric repressor without any adverse effects. Our findings establish that splicing repression is a major function of TDP-43 in motor neurons and strongly support the idea that loss of TDP-43-mediated splicing fidelity represents a key pathogenic mechanism underlying motor neuron loss in ALS.
Collapse
|
147
|
Kawakami I, Arai T, Hasegawa M. The basis of clinicopathological heterogeneity in TDP-43 proteinopathy. Acta Neuropathol 2019; 138:751-770. [PMID: 31555895 PMCID: PMC6800885 DOI: 10.1007/s00401-019-02077-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/27/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022]
Abstract
Transactive response DNA-binding protein 43 kDa (TDP-43) was identified as a major disease-associated component in the brain of patients with amyotrophic lateral sclerosis (ALS), as well as the largest subset of patients with frontotemporal lobar degeneration with ubiquitinated inclusions (FTLD-U), which characteristically exhibits cytoplasmic inclusions that are positive for ubiquitin but negative for tau and α-synuclein. TDP-43 pathology occurs in distinct brain regions, involves disparate brain networks, and features accumulation of misfolded proteins in various cell types and in different neuroanatomical regions. The clinical phenotypes of ALS and FTLD-TDP (FTLD with abnormal intracellular accumulations of TDP-43) correlate with characteristic distribution patterns of the underlying pathology across specific brain regions with disease progression. Recent studies support the idea that pathological protein spreads from neuron to neuron via axonal transport in a hierarchical manner. However, little is known to date about the basis of the selective cellular and regional vulnerability, although the information would have important implications for the development of targeted and personalized therapies. Here, we aim to summarize recent advances in the neuropathology, genetics and animal models of TDP-43 proteinopathy, and their relationship to clinical phenotypes for the underlying selective neuronal and regional susceptibilities. Finally, we attempt to integrate these findings into the emerging picture of TDP-43 proteinopathy, and to highlight key issues for future therapy and research.
Collapse
Affiliation(s)
- Ito Kawakami
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
- Department of Neuropathology, Tokyo Metropolitan Geriatric Hospital and Institute, Tokyo, Japan
| | - Tetsuaki Arai
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
- Department of Psychiatry, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.
| | - Masato Hasegawa
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6, Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
| |
Collapse
|
148
|
White MA, Lin Z, Kim E, Henstridge CM, Pena Altamira E, Hunt CK, Burchill E, Callaghan I, Loreto A, Brown-Wright H, Mead R, Simmons C, Cash D, Coleman MP, Sreedharan J. Sarm1 deletion suppresses TDP-43-linked motor neuron degeneration and cortical spine loss. Acta Neuropathol Commun 2019; 7:166. [PMID: 31661035 PMCID: PMC6819591 DOI: 10.1186/s40478-019-0800-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/30/2019] [Indexed: 02/05/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative condition that primarily affects the motor system and shares many features with frontotemporal dementia (FTD). Evidence suggests that ALS is a 'dying-back' disease, with peripheral denervation and axonal degeneration occurring before loss of motor neuron cell bodies. Distal to a nerve injury, a similar pattern of axonal degeneration can be seen, which is mediated by an active axon destruction mechanism called Wallerian degeneration. Sterile alpha and TIR motif-containing 1 (Sarm1) is a key gene in the Wallerian pathway and its deletion provides long-term protection against both Wallerian degeneration and Wallerian-like, non-injury induced axonopathy, a retrograde degenerative process that occurs in many neurodegenerative diseases where axonal transport is impaired. Here, we explored whether Sarm1 signalling could be a therapeutic target for ALS by deleting Sarm1 from a mouse model of ALS-FTD, a TDP-43Q331K, YFP-H double transgenic mouse. Sarm1 deletion attenuated motor axon degeneration and neuromuscular junction denervation. Motor neuron cell bodies were also significantly protected. Deletion of Sarm1 also attenuated loss of layer V pyramidal neuronal dendritic spines in the primary motor cortex. Structural MRI identified the entorhinal cortex as the most significantly atrophic region, and histological studies confirmed a greater loss of neurons in the entorhinal cortex than in the motor cortex, suggesting a prominent FTD-like pattern of neurodegeneration in this transgenic mouse model. Despite the reduction in neuronal degeneration, Sarm1 deletion did not attenuate age-related behavioural deficits caused by TDP-43Q331K. However, Sarm1 deletion was associated with a significant increase in the viability of male TDP-43Q331K mice, suggesting a detrimental role of Wallerian-like pathways in the earliest stages of TDP-43Q331K-mediated neurodegeneration. Collectively, these results indicate that anti-SARM1 strategies have therapeutic potential in ALS-FTD.
Collapse
Affiliation(s)
- Matthew A White
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE5 9RT, UK
| | - Ziqiang Lin
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE5 9RT, UK
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Eugene Kim
- BRAIN Centre (Biomarker Research And Imaging for Neuroscience), Department of Neuroimaging, IoPPN, King's College London, London, UK
| | | | - Emiliano Pena Altamira
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE5 9RT, UK
| | - Camille K Hunt
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE5 9RT, UK
| | - Ella Burchill
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE5 9RT, UK
| | - Isobel Callaghan
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE5 9RT, UK
| | - Andrea Loreto
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Heledd Brown-Wright
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Richard Mead
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Camilla Simmons
- BRAIN Centre (Biomarker Research And Imaging for Neuroscience), Department of Neuroimaging, IoPPN, King's College London, London, UK
| | - Diana Cash
- BRAIN Centre (Biomarker Research And Imaging for Neuroscience), Department of Neuroimaging, IoPPN, King's College London, London, UK
| | - Michael P Coleman
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Jemeen Sreedharan
- Department of Basic and Clinical Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE5 9RT, UK.
| |
Collapse
|
149
|
Chang YH, Dubnau J. The Gypsy Endogenous Retrovirus Drives Non-Cell-Autonomous Propagation in a Drosophila TDP-43 Model of Neurodegeneration. Curr Biol 2019; 29:3135-3152.e4. [PMID: 31495585 PMCID: PMC6783360 DOI: 10.1016/j.cub.2019.07.071] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/11/2019] [Accepted: 07/23/2019] [Indexed: 12/16/2022]
Abstract
A hallmark of neurodegenerative disease is focal onset of pathological protein aggregation, followed by progressive spread of pathology to connected brain regions. In amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), pathology is often associated with aggregation of TAR DNA-binding protein 43 (TDP-43). Although aggregated TDP-43 protein moves between cells, it is not clear whether and how this movement propagates the degeneration. Here, we have established a Drosophila model of human TDP-43 in which we initiated toxic expression of human TDP-43 focally within small groups of glial cells. We found that this focal onset kills adjacent neurons. Surprisingly, we show that this spreading death is caused by an endogenous retrovirus within the glia, which leads to DNA damage and death in adjacent neurons. These findings suggest a possible mechanism by which human retroviruses such as HERV-K might contribute to TDP-43-mediated propagation of neurodegeneration.
Collapse
Affiliation(s)
- Yung-Heng Chang
- Department of Anesthesiology, Stony Brook School of Medicine, NY 11794, USA
| | - Josh Dubnau
- Department of Anesthesiology, Stony Brook School of Medicine, NY 11794, USA; Department of Neurobiology and Behavior, Stony Brook University, NY 11794, USA.
| |
Collapse
|
150
|
Müller HP, Brenner D, Roselli F, Wiesner D, Abaei A, Gorges M, Danzer KM, Ludolph AC, Tsao W, Wong PC, Rasche V, Weishaupt JH, Kassubek J. Longitudinal diffusion tensor magnetic resonance imaging analysis at the cohort level reveals disturbed cortical and callosal microstructure with spared corticospinal tract in the TDP-43 G298S ALS mouse model. Transl Neurodegener 2019; 8:27. [PMID: 31485326 PMCID: PMC6716821 DOI: 10.1186/s40035-019-0163-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
Background In vivo diffusion tensor imaging (DTI) of the mouse brain was used to identify TDP-43 associated alterations in a mouse model for amyotrophic lateral sclerosis (ALS). Methods Ten mice with TDP-43 G298S overexpression under control of the Thy1.2 promoter and 10 wild type (wt) underwent longitudinal DTI scans at 11.7 T, including one baseline and one follow-up scan with an interval of about 5 months. Whole brain-based spatial statistics (WBSS) of DTI-based parameter maps was used to identify longitudinal alterations of TDP-43 G298S mice compared to wt at the cohort level. Results were supplemented by tractwise fractional anisotropy statistics (TFAS) and histological evaluation of motor cortex for signs of neuronal loss. Results Alterations at the cohort level in TDP-43 G298S mice were observed cross-sectionally and longitudinally in motor areas M1/M2 and in transcallosal fibers but not in the corticospinal tract. Neuronal loss in layer V of motor cortex was detected in TDP-43 G298S at the later (but not at the earlier) timepoint compared to wt. Conclusion DTI mapping of TDP-43 G298S mice demonstrated progression in motor areas M1/M2. WBSS and TFAS are useful techniques to localize TDP-43 G298S associated alterations over time in this ALS mouse model, as a biological marker.
Collapse
Affiliation(s)
- Hans-Peter Müller
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - David Brenner
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Francesco Roselli
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany.,2German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Diana Wiesner
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Alireza Abaei
- 3Core Facility Small Animal MRI, University of Ulm, Ulm, Germany
| | - Martin Gorges
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Karin M Danzer
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Albert C Ludolph
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - William Tsao
- 4Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Philip C Wong
- 4Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Volker Rasche
- 3Core Facility Small Animal MRI, University of Ulm, Ulm, Germany
| | - Jochen H Weishaupt
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| | - Jan Kassubek
- 1Department of Neurology, University of Ulm, Oberer Eselsberg 45, RKU, D-89081 Ulm, Germany
| |
Collapse
|