251
|
Bridging biophysics and neurology: aberrant phase transitions in neurodegenerative disease. Nat Rev Neurol 2020; 15:272-286. [PMID: 30890779 DOI: 10.1038/s41582-019-0157-5] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biomolecular condensation arising through phase transitions has emerged as an essential organizational strategy that governs many aspects of cell biology. In particular, the role of phase transitions in the assembly of large, complex ribonucleoprotein (RNP) granules has become appreciated as an important regulator of RNA metabolism. In parallel, genetic, histopathological and cell and molecular studies have provided evidence that disturbance of phase transitions is an important driver of neurological diseases, notably amyotrophic lateral sclerosis (ALS), but most likely also other diseases. Indeed, our growing knowledge of the biophysics underlying biological phase transitions suggests that this process offers a unifying mechanism to explain the numerous and diverse disturbances in RNA metabolism that have been observed in ALS and some related diseases - specifically, that these diseases are driven by disturbances in the material properties of RNP granules. Here, we review the evidence for this hypothesis, emphasizing the reciprocal roles in which disease-related protein and disease-related RNA can lead to disturbances in the material properties of RNP granules and consequent pathogenesis. Additionally, we review evidence that implicates aberrant phase transitions as a contributing factor to a larger set of neurodegenerative diseases, including frontotemporal dementia, certain repeat expansion diseases and Alzheimer disease.
Collapse
|
252
|
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: 40] [Impact Index Per Article: 10.0] [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
|
253
|
Kasahara S, Ishihara T, Koike Y, Sugai A, Onodera O. [Molecular mechanism of amyotrophic lateral sclerosis (ALS) from the viewpoint of the formation and degeneration of transactive response DNA-binding protein 43 kDa (TDP-43) inclusions]. Rinsho Shinkeigaku 2020; 60:109-116. [PMID: 31956195 DOI: 10.5692/clinicalneurol.cn-001362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Sporadic amyotrophic lateral sclerosis (SALS) and many cases of familial ALS (FALS) demonstrate cytoplasmic transactive response DNA-binding protein 43 kDa (TDP-43)-positive inclusion bodies. Thus, TDP-43 plays a vital role in ALS pathogenesis. Functional analysis of the ALS causative genes advanced the elucidation of the mechanism associated with the formation and degradation of TDP-43 aggregates. Stress granules, which are non-membranous organelles, are attracting attention as sites of aggregate formation, with involvement of FUS and C9orf72. Concurrently, ALS causative genes related to the ubiquitin-proteasome and autophagy systems, which are aggregate degradation mechanisms, have also been reported. Therefore, therapeutic research based on the molecular pathology common to SALS and FALS has been advanced.
Collapse
Affiliation(s)
- Sou Kasahara
- Department of Neurology, Brain Research Institute, Niigata University
| | - Tomohiko Ishihara
- Department of Neurology, Brain Research Institute, Niigata University
| | - Yuka Koike
- Department of Neurology, Brain Research Institute, Niigata University
| | - Akihiro Sugai
- Department of Neurology, Brain Research Institute, Niigata University
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University
| |
Collapse
|
254
|
Lázaro DF, Bellucci A, Brundin P, Outeiro TF. Editorial: Protein Misfolding and Spreading Pathology in Neurodegenerative Diseases. Front Mol Neurosci 2020; 12:312. [PMID: 32009897 PMCID: PMC6978792 DOI: 10.3389/fnmol.2019.00312] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 12/04/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
- Diana F Lázaro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Gottingen, Göttingen, Germany
| | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Gottingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,The Medical School, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
255
|
Loganathan S, Lehmkuhl EM, Eck RJ, Zarnescu DC. To Be or Not To Be…Toxic-Is RNA Association With TDP-43 Complexes Deleterious or Protective in Neurodegeneration? Front Mol Biosci 2020; 6:154. [PMID: 31998750 PMCID: PMC6965497 DOI: 10.3389/fmolb.2019.00154] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022] Open
Abstract
TAR DNA binding protein (TDP-43) is a nucleic acid binding protein associated with insoluble cytoplasmic aggregates in several neurodegenerative disorders, including 97% of the ALS cases. In healthy individuals, TDP-43 is primarily localized to the nucleus; it can shuttle between the nucleus and the cytoplasm, and is involved in several aspects of RNA processing including transcription, splicing, RNA stability, transport, localization, stress granule (SG) formation, and translation. Upon stress, TDP-43 aggregates in the cytoplasm and associates with several types of RNA and protein assemblies, resulting in nuclear depletion of TDP-43. Under conditions of prolonged stress, cytoplasmic TDP-43 undergoes liquid-liquid phase separation (LLPS) and becomes less mobile. Evidence exists to support a scenario in which insoluble TDP-43 complexes sequester RNA and/or proteins causing disturbances in both ribostasis and proteostasis, which in turn contribute to neurodegeneration. However, the relationship between RNA binding and TDP-43 toxicity remains unclear. Recent studies provide conflicting views on the role of RNA in TDP-43 toxicity, with some finding RNA as a toxic factor whereby RNA binding contributes to TDP-43 toxicity, while others find RNA to be a protective factor that inhibits TDP-43 aggregation. Here we review and discuss these recent reports, which ultimately highlight the importance of understanding the heterogeneity of TDP-43 assemblies and collectively point to solubilizing TDP-43 as a potential therapeutic strategy.
Collapse
Affiliation(s)
| | - Erik M Lehmkuhl
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States
| | - Randall J Eck
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States.,Department of Neuroscience, University of Arizona, Tucson, AZ, United States
| | - Daniela C Zarnescu
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ, United States.,Department of Neuroscience, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
256
|
Perrone B, La Cognata V, Sprovieri T, Ungaro C, Conforti FL, Andò S, Cavallaro S. Alternative Splicing of ALS Genes: Misregulation and Potential Therapies. Cell Mol Neurobiol 2020; 40:1-14. [PMID: 31385134 DOI: 10.1007/s10571-019-00717-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022]
Abstract
Neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), Parkinson's, Alzheimer's, and Huntington's disease affect a rapidly increasing population worldwide. Although common pathogenic mechanisms have been identified (e.g., protein aggregation or dysfunction, immune response alteration and axonal degeneration), the molecular events underlying timing, dosage, expression, and location of RNA molecules are still not fully elucidated. In particular, the alternative splicing (AS) mechanism is a crucial player in RNA processing and represents a fundamental determinant for brain development, as well as for the physiological functions of neuronal circuits. Although in recent years our knowledge of AS events has increased substantially, deciphering the molecular interconnections between splicing and ALS remains a complex task and still requires considerable efforts. In the present review, we will summarize the current scientific evidence outlining the involvement of AS in the pathogenic processes of ALS. We will also focus on recent insights concerning the tuning of splicing mechanisms by epigenomic and epi-transcriptomic regulation, providing an overview of the available genomic technologies to investigate AS drivers on a genome-wide scale, even at a single-cell level resolution. In the future, gene therapy strategies and RNA-based technologies may be utilized to intercept or modulate the splicing mechanism and produce beneficial effects against ALS.
Collapse
Affiliation(s)
- Benedetta Perrone
- Institute for Biomedical Research and Innovation, National Research Council, Mangone, Cosenza, Italy
| | - Valentina La Cognata
- Institute for Biomedical Research and Innovation, National Research Council, Catania, Italy
| | - Teresa Sprovieri
- Institute for Biomedical Research and Innovation, National Research Council, Mangone, Cosenza, Italy
| | - Carmine Ungaro
- Institute for Biomedical Research and Innovation, National Research Council, Mangone, Cosenza, Italy
| | - Francesca Luisa Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Cosenza, Italy
- Centro Sanitario, University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, National Research Council, Catania, Italy.
| |
Collapse
|
257
|
Dutta R, Sarkar SR. Role of Dynein and Dynactin (DCTN-1) in Neurodegenerative Diseases. ACTA ACUST UNITED AC 2019. [DOI: 10.33805/2641-8991.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The pathophysiology and concept of degeneration in central nervous system is very complex and overwhelming at times. There is a complex mechanism which exists among different molecules in the cytoplasm of cell bodies of neurons, antegrade and retrograde axonal transport of cargoes and accumulation of certain substances and proteins which can influence the excitatory neurotransmitter like glutamate initiating the process of neurodegeneration. Neurons have extensive processes and communication between those processes and the cell body is crucial to neuronal function, viability and survival over time with progression of age. Researchers believe neurons are uniquely dependent on microtubule-based cargo transport. There is enough evidence to support that deficits in retrograde axonal transport contribute to pathogenesis in multiple neurodegenerative diseases. Cytoplasmic dynein and its regulation by Dynactin (DCTN1) is the major molecular motor cargo involved in autophagy, mitosis and neuronal cell survival. Mutation in dynactin gene located in 2p13.1,is indeed studied very extensively and is considered to be involved directly or indirectly to various conditions like Perry syndrome, familial and sporadic Amyotrophic lateral sclerosis, Hereditary spastic paraplegia, Spinocerebellar Ataxia (SCA-5), Huntingtons disease, Alzheimers disease, Charcot marie tooth disease, Hereditary motor neuropathy 7B, prion disease, parkinsons disease, malformation of cortical development, polymicrogyria to name a few with exception of Multiple Sclerosis (MS).
Collapse
|
258
|
Corrado L, Pensato V, Croce R, Di Pierro A, Mellone S, Dalla Bella E, Salsano E, Paraboschi EM, Giordano M, Saraceno M, Mazzini L, Gellera C, D'Alfonso S. The first case of the TARDBP p.G294V mutation in a homozygous state: is a single pathogenic allele sufficient to cause ALS? Amyotroph Lateral Scler Frontotemporal Degener 2019; 21:273-279. [PMID: 31852254 DOI: 10.1080/21678421.2019.1704011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Here, we described the first amyotrophic lateral sclerosis patient presenting the c.881 G > T p.G294V TARDBP mutation in homozygous status. The patient belongs to a large pedigree from Morocco. Except for one older affected brother his parents and remaining 8 sibs are referred to be healthy and do not show any neurological sign or symptom. The lack of evidence of TARDBP deletions of any sizes, together with the presence of several AOH segments, strongly suggests that the homozygosity status of p.G294V in the proband derived from parental consanguinity. A revision of the literature and our cohorts indicates that the p.G294V mutation has been detected in only 15 additional ALS patients in heterozygosity and, except for one additional Moroccan patient, all were of Italian origin. The analysis of microsatellite markers surrounding the TARDBP gene in 8 individuals carrying the p.G294V mutation showed that the haplotypic context of the Moroccan proband is shared with most patients of European origin indicating that they carry the p.G294V mutation inherited from a common ancestor. The analysis of the 15 ALS pedigrees (from literature data and present study), strongly suggests a reduced penetrance of the p.G294V mutation since for 13 of the 15 described p.G294V ALS cases the parents did not show any neurological symptoms. This result has potentially important implications in genetic counseling, since genetic testing of a reduced penetrance mutation on pre-symptomatic individuals proves very difficult to predict the outcome based on the genotype.
Collapse
Affiliation(s)
- Lucia Corrado
- Department of Health Sciences, Human Genetics Laboratory, UPO University, Novara, Italy
| | - Viviana Pensato
- Unit of Genetics of Neurodegenerative and Metabolic Diseases and Motor Neuron Diseases Centre, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Roberta Croce
- Department of Health Sciences, Human Genetics Laboratory, UPO University, Novara, Italy
| | - Alice Di Pierro
- Department of Health Sciences, Human Genetics Laboratory, UPO University, Novara, Italy
| | - Simona Mellone
- Department of Health Sciences, Human Genetics Laboratory, UPO University, Novara, Italy
| | - Eleonora Dalla Bella
- III Neurology Unit and Motor Neuron Diseases Centre, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ettore Salsano
- X Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Mara Giordano
- Department of Health Sciences, Human Genetics Laboratory, UPO University, Novara, Italy
| | - Massimo Saraceno
- Department of Neurology, UPO University and Maggiore della Carità Hospital, Corso Mazzini, Novara
| | - Letizia Mazzini
- Department of Neurology, UPO University and Maggiore della Carità Hospital, Corso Mazzini, Novara
| | - Cinzia Gellera
- Unit of Genetics of Neurodegenerative and Metabolic Diseases and Motor Neuron Diseases Centre, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, Human Genetics Laboratory, UPO University, Novara, Italy
| |
Collapse
|
259
|
Mejzini R, Flynn LL, Pitout IL, Fletcher S, Wilton SD, Akkari PA. ALS Genetics, Mechanisms, and Therapeutics: Where Are We Now? Front Neurosci 2019; 13:1310. [PMID: 31866818 PMCID: PMC6909825 DOI: 10.3389/fnins.2019.01310] [Citation(s) in RCA: 470] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022] Open
Abstract
The scientific landscape surrounding amyotrophic lateral sclerosis (ALS) continues to shift as the number of genes associated with the disease risk and pathogenesis, and the cellular processes involved, continues to grow. Despite decades of intense research and over 50 potentially causative or disease-modifying genes identified, etiology remains unexplained and treatment options remain limited for the majority of ALS patients. Various factors have contributed to the slow progress in understanding and developing therapeutics for this disease. Here, we review the genetic basis of ALS, highlighting factors that have contributed to the elusiveness of genetic heritability. The most commonly mutated ALS-linked genes are reviewed with an emphasis on disease-causing mechanisms. The cellular processes involved in ALS pathogenesis are discussed, with evidence implicating their involvement in ALS summarized. Past and present therapeutic strategies and the benefits and limitations of the model systems available to ALS researchers are discussed with future directions for research that may lead to effective treatment strategies outlined.
Collapse
Affiliation(s)
- Rita Mejzini
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- The Perron Institute for Neurological and Translational Science, Perth, WA, Australia
| | - Loren L. Flynn
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- The Perron Institute for Neurological and Translational Science, Perth, WA, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Ianthe L. Pitout
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- The Perron Institute for Neurological and Translational Science, Perth, WA, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- The Perron Institute for Neurological and Translational Science, Perth, WA, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- The Perron Institute for Neurological and Translational Science, Perth, WA, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| | - P. Anthony Akkari
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, Australia
- The Perron Institute for Neurological and Translational Science, Perth, WA, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
260
|
Bogaert E, Boeynaems S, Kato M, Guo L, Caulfield TR, Steyaert J, Scheveneels W, Wilmans N, Haeck W, Hersmus N, Schymkowitz J, Rousseau F, Shorter J, Callaerts P, Robberecht W, Van Damme P, Van Den Bosch L. Molecular Dissection of FUS Points at Synergistic Effect of Low-Complexity Domains in Toxicity. Cell Rep 2019; 24:529-537.e4. [PMID: 30021151 PMCID: PMC6077250 DOI: 10.1016/j.celrep.2018.06.070] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 03/11/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022] Open
Abstract
RNA-binding protein aggregation is a pathological hallmark of several neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). To gain better insight into the molecular interactions underlying this process, we investigated FUS, which is mutated and aggregated in both ALS and FTLD. We generated a Drosophila model of FUS toxicity and identified a previously unrecognized synergistic effect between the N-terminal prion-like domain and the C-terminal arginine-rich domain to mediate toxicity. Although the prion-like domain is generally considered to mediate aggregation of FUS, we find that arginine residues in the C-terminal low-complexity domain are also required for maturation of FUS in cellular stress granules. These data highlight an important role for arginine-rich domains in the pathology of RNA-binding proteins. Both QGSY and RGG2 domains are necessary for FUS-induced neurodegeneration in flies Arginine-rich domains interact with QGSY hydrogels and liquid droplets RGG2 arginines promote phase separation of FUS in vitro and in cells FUS phase separation behavior in vitro correlates with neurodegeneration in vivo
Collapse
Affiliation(s)
- Elke Bogaert
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium
| | - Steven Boeynaems
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Masato Kato
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lin Guo
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas R Caulfield
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Jolien Steyaert
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium
| | - Wendy Scheveneels
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium
| | - Nathalie Wilmans
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium
| | - Wanda Haeck
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium
| | - Nicole Hersmus
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium; Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Frederic Rousseau
- Switch Laboratory, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium; Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - James Shorter
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patrick Callaerts
- Laboratory of Behavioral and Developmental Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Wim Robberecht
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium; Department of Neurology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Philip Van Damme
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium; Department of Neurology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ludo Van Den Bosch
- Experimental Neurology, Department of Neurosciences, and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, 3000 Leuven, Belgium; Laboratory of Neurobiology, Center for Brain & Disease Research, VIB, 3000 Leuven, Belgium.
| |
Collapse
|
261
|
Chen HJ, Topp SD, Hui HS, Zacco E, Katarya M, McLoughlin C, King A, Smith BN, Troakes C, Pastore A, Shaw CE. RRM adjacent TARDBP mutations disrupt RNA binding and enhance TDP-43 proteinopathy. Brain 2019; 142:3753-3770. [PMID: 31605140 PMCID: PMC6885686 DOI: 10.1093/brain/awz313] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/24/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) presents with focal muscle weakness due to motor neuron degeneration that becomes generalized, leading to death from respiratory failure within 3-5 years from symptom onset. Despite the heterogeneity of aetiology, TDP-43 proteinopathy is a common pathological feature that is observed in >95% of ALS and tau-negative frontotemporal dementia (FTD) cases. TDP-43 is a DNA/RNA-binding protein that in ALS and FTD translocates from being predominantly nuclear to form detergent-resistant, hyperphosphorylated aggregates in the cytoplasm of affected neurons and glia. Mutations in TARDBP account for 1-4% of all ALS cases and almost all arise in the low complexity C-terminal domain that does not affect RNA binding and processing. Here we report an ALS/FTD kindred with a novel K181E TDP-43 mutation that is located in close proximity to the RRM1 domain. To offer predictive gene testing to at-risk family members, we undertook a series of functional studies to characterize the properties of the mutation. Spectroscopy studies of the K181E protein revealed no evidence of significant misfolding. Although it is unable to bind to or splice RNA, it forms abundant aggregates in transfected cells. We extended our study to include other ALS-linked mutations adjacent to the RRM domains that also disrupt RNA binding and greatly enhance TDP-43 aggregation, forming detergent-resistant and hyperphosphorylated inclusions. Lastly, we demonstrate that K181E binds to, and sequesters, wild-type TDP-43 within nuclear and cytoplasmic inclusions. Thus, we demonstrate that TDP-43 mutations that disrupt RNA binding greatly enhance aggregation and are likely to be pathogenic as they promote wild-type TDP-43 to mislocalize and aggregate acting in a dominant-negative manner. This study highlights the importance of RNA binding to maintain TDP-43 solubility and the role of TDP-43 aggregation in disease pathogenesis.
Collapse
Affiliation(s)
- Han-Jou Chen
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 125 Coldharbour Lane, Camberwell, SE5 9NU, London, UK
- York Biomedical Research Institute, Department of Biology, University of York, Wentworth Way, YO10 5DD, York, UK
| | - Simon D Topp
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 125 Coldharbour Lane, Camberwell, SE5 9NU, London, UK
| | - Ho Sang Hui
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 125 Coldharbour Lane, Camberwell, SE5 9NU, London, UK
| | - Elsa Zacco
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 125 Coldharbour Lane, Camberwell, SE5 9NU, London, UK
| | - Malvika Katarya
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 125 Coldharbour Lane, Camberwell, SE5 9NU, London, UK
| | - Conor McLoughlin
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 125 Coldharbour Lane, Camberwell, SE5 9NU, London, UK
| | - Andrew King
- MRC London Neurodegenerative Diseases Brain Bank, De Crespigny Park, SE5 8AF, London, UK
| | - Bradley N Smith
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 125 Coldharbour Lane, Camberwell, SE5 9NU, London, UK
| | - Claire Troakes
- MRC London Neurodegenerative Diseases Brain Bank, De Crespigny Park, SE5 8AF, London, UK
| | - Annalisa Pastore
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 125 Coldharbour Lane, Camberwell, SE5 9NU, London, UK
| | - Christopher E Shaw
- United Kingdom Dementia Research Institute Centre, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, 125 Coldharbour Lane, Camberwell, SE5 9NU, London, UK
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
262
|
Salapa HE, Libner CD, Levin MC. Dysfunctional RNA-binding protein biology and neurodegeneration in experimental autoimmune encephalomyelitis in female mice. J Neurosci Res 2019; 98:704-717. [PMID: 31755578 DOI: 10.1002/jnr.24554] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/04/2019] [Accepted: 10/24/2019] [Indexed: 12/11/2022]
Abstract
Altered stress granule (SG) and RNA-binding protein (RBP) biology have been shown to contribute to the pathogenesis of several neurodegenerative diseases, yet little is known about their role in multiple sclerosis (MS). Pathological features associated with dysfunctional RBPs include RBP mislocalization from its normal nuclear location to the cytoplasm and the formation of chronic SGs. We tested the hypothesis that altered SG and RBP biology might contribute to the neurodegeneration in experimental autoimmune encephalomyelitis (EAE). C57BL/6 female mice were actively immunized with MOG35-55 to induce EAE. Spinal cords were examined for mislocalization of the RBPs, heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) and TAR-DNA binding protein-43 (TDP-43), SGs, neurodegeneration (SMI-32), T cells (CD3), and macrophages (CD68). In contrast to naive mice, mice with EAE showed SG formation (p < 0.0001) and mislocalization of hnRNP A1 (p < 0.05) in neurons of the ventral spinal cord gray matter, which correlated with clinical score (R = 0.8104, p = 0.0253). In these same areas, there was a neuronal loss (p < 0.0001) and increased SMI-32 immunoreactivity (both markers of neurodegeneration) and increased staining for CD3+ T cells and IFN-gamma. These findings recapitulate the SG and RBP biology and markers of neurodegeneration in MS tissues and suggest that altered SG and RBP biology contribute to the neurodegeneration in EAE, which might also apply to the pathogenesis of MS.
Collapse
Affiliation(s)
- Hannah E Salapa
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Cole D Libner
- Department of Health Sciences, University of Saskatchewan, Saskatoon, SK, Canada
| | - Michael C Levin
- Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada.,Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK, Canada.,College of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
263
|
Fernandopulle M, Wang G, Nixon-Abell J, Qamar S, Balaji V, Morihara R, St George-Hyslop PH. Inherited and Sporadic Amyotrophic Lateral Sclerosis and Fronto-Temporal Lobar Degenerations arising from Pathological Condensates of Phase Separating Proteins. Hum Mol Genet 2019; 28:R187-R196. [PMID: 31595953 PMCID: PMC6872449 DOI: 10.1093/hmg/ddz162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022] Open
Abstract
Recent work on the biophysics of proteins with low complexity, intrinsically disordered domains that have the capacity to form biological condensates has profoundly altered the concepts about the pathogenesis of inherited and sporadic neurodegenerative disorders associated with pathological accumulation of these proteins. In the present review, we use the FUS, TDP-43 and A11 proteins as examples to illustrate how missense mutations and aberrant post-translational modifications of these proteins cause amyotrophic lateral sclerosis (ALS) and fronto-temporal lobar degeneration (FTLD).
Collapse
Affiliation(s)
- Michael Fernandopulle
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - GuoZhen Wang
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Jonathon Nixon-Abell
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Seema Qamar
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Varun Balaji
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| | - Ryuta Morihara
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| | - Peter H St George-Hyslop
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| |
Collapse
|
264
|
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: 51] [Impact Index Per Article: 10.2] [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
|
265
|
Wei L, Tian Y, Chen Y, Wei Q, Chen F, Cao B, Wu Y, Zhao B, Chen X, Xie C, Xi C, Yu X, Wang J, Lv X, Du J, Wang Y, Shen L, Wang X, Shen B, Guo Q, Guo L, Xia K, Xie P, Zhang X, Zuo X, Shang H, Wang K. Identification of TYW3/CRYZ and FGD4 as susceptibility genes for amyotrophic lateral sclerosis. NEUROLOGY-GENETICS 2019; 5:e375. [PMID: 31872054 PMCID: PMC6878836 DOI: 10.1212/nxg.0000000000000375] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/10/2019] [Indexed: 02/05/2023]
Abstract
Objective A 2-stage genome-wide association was conducted to explore the genetic etiology of amyotrophic lateral sclerosis (ALS) in the Chinese Han population. Methods Totally, 700 cases and 4,027 controls were genotyped in the discovery stage using Illumina Human660W-Quad BeadChips. Top associated single nucleotide polymorphisms from the discovery stage were then genotyped in an independent cohort with 884 cases and 5,329 controls. Combined analysis was conducted by combining all samples from the 2 stages. Results Two novel loci, 1p31 and 12p11, showed strong associations with ALS. These novel loci explained 2.2% of overall variance in disease risk. Expression quantitative trait loci searches identified TYW/CRYZ and FGD4 as risk genes at 1p13 and 12p11, respectively. Conclusions This study identifies novel susceptibility genes for ALS. Identification of TYW3/CRYZ in the current study supports the notion that insulin resistance may be involved in ALS pathogenesis, whereas FGD4 suggests an association with Charcot-Marie-Tooth disease.
Collapse
Affiliation(s)
- Ling Wei
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Yanghua Tian
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Yongping Chen
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Qianqian Wei
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Fangfang Chen
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Bei Cao
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Ying Wu
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Bi Zhao
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Xueping Chen
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Chengjuan Xie
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Chunhua Xi
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Xu'en Yu
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Juan Wang
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Xinyi Lv
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Jing Du
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Yu Wang
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Lu Shen
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Xin Wang
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Bin Shen
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Qihao Guo
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Li Guo
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Kun Xia
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Peng Xie
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Xuejun Zhang
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Xianbo Zuo
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Huifang Shang
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| | - Kai Wang
- Department of Neurology (L.W., Y.T., C. Xie, Y. Wang, K.W.), the First Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (Y.C., Q.W., B.C., Y. Wu, B.Z., X.C., H.S.), West China Hospital of Sichuan University, Chengdu; Department of Medical Psychology (F.C., K.W.), Anhui Medical University; Department of Neurology (C. Xi), the Third Affiliated Hospital of Anhui Medical University; Institution of Neurology (X.Y.), Anhui College of Traditional Medicine; Department of Neurology (J.W.), the Second People's Hospital of Hefei; Department of Neurology (X.L.), Anhui Provincial Hospital; Department of Neurology (J.D.), the Second Affiliated Hospital of Anhui Medical University, Hefei; Department of Neurology (L.S.), Xiangya Hospital of Central South University, Changsha; Department of Neurology (X.W.), Zhongshan Hospital of Fudan University, Shanghai; Department of Physiology (B.S.), School of Basic Medicine, Anhui Medical University, Hefei; Department of Neurology (Q.G.), Huashan Hospital of Fudan University, Shanghai; Department of Neurology (L.G.), the Second Hospital of Hebei Medical University, Shijiazhuang; School of Life Science (K.X.), Central South University, Changsha; Department of Neurology (P.X.), the First Affiliated Hospital of Chongqing Medical University, Chongqing; Department of Dermatology (X. Zhang, X. Zuo), the First Affiliated Hospital of Anhui Medical University; and State Key Laboratory Incubation Base of Dermatology (X. Zhang, X. Zuo), Ministry of National Science and Technology, Hefei, China
| |
Collapse
|
266
|
Quadri Z, Johnson N, Zamudio F, Miller A, Peters M, Smeltzer S, Hunt JB, Housley SB, Brown B, Kraner S, Norris CM, Nash K, Weeber E, Lee DC, Selenica MLB. Overexpression of human wtTDP-43 causes impairment in hippocampal plasticity and behavioral deficits in CAMKII-tTa transgenic mouse model. Mol Cell Neurosci 2019; 102:103418. [PMID: 31705957 DOI: 10.1016/j.mcn.2019.103418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 10/23/2019] [Accepted: 10/31/2019] [Indexed: 12/13/2022] Open
Abstract
AIMS The current study utilizes the adeno-associated viral gene transfer system in the CAMKIIα-tTA mouse model to overexpress human wild type TDP-43 (wtTDP-43) and α-synuclein (α-Syn) proteins. The co-existence of these proteins is evident in the pathology of neurodegenerative disorders such as frontotemporal lobar degeneration (FTLD), Parkinson disease (PD), and dementia with Lewy bodies (DLB). METHODS The novel bicistronic recombinant adeno-associated virus (rAAV) serotype 9 drives wtTDP-43 and α-Syn expression in the hippocampus via "TetO" CMV promoter. Behavior, electrophysiology, and biochemical and histological assays were used to validate neuropathology. RESULTS We report that overexpression of wtTDP-43 but not α-Syn contributes to hippocampal CA2-specific pyramidal neuronal loss and overall hippocampal atrophy. Further, we report a reduction of hippocampal long-term potentiation and decline in learning and memory performance of wtTDP-43 expressing mice. Elevated wtTDP-43 levels induced selective degeneration of Purkinje cell protein 4 (PCP-4) positive neurons while both wtTDP-43 and α-Syn expression reduced subsets of the glutamate receptor expression in the hippocampus. CONCLUSIONS Overall, our findings suggest the significant vulnerability of hippocampal neurons toward elevated wtTDP-43 levels possibly via PCP-4 and GluR-dependent calcium signaling pathways. Further, we report that wtTDP-43 expression induced selective CA2 subfield degeneration, contributing to the deterioration of the hippocampal-dependent cognitive phenotype.
Collapse
Affiliation(s)
- Zainuddin Quadri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA; Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Nicholas Johnson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA; Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Frank Zamudio
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA; Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Abraian Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA; Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Melinda Peters
- Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Shayna Smeltzer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA; Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Jerry B Hunt
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA; Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Steven B Housley
- Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Breanna Brown
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA; Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Susan Kraner
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Kevin Nash
- Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Edwin Weeber
- Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA; Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Daniel C Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA; Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA
| | - Maj-Linda B Selenica
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, USA; Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
267
|
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: 56] [Impact Index Per Article: 11.2] [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
|
268
|
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: 64] [Impact Index Per Article: 12.8] [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
|
269
|
Gao J, Wang L, Gao C, Arakawa H, Perry G, Wang X. TDP-43 inhibitory peptide alleviates neurodegeneration and memory loss in an APP transgenic mouse model for Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165580. [PMID: 31678156 DOI: 10.1016/j.bbadis.2019.165580] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly, characterized clinically by progressive decline in cognitive function and neuropathologically by the presence of senile plaques and neuronal loss in the brain. While current drugs for AD are always employed as symptomatic therapies with variable benefits, there is no treatment to delay its progression or halt neurodegeneration. TAR DNA-binding protein 43 (TDP-43) proteinopathy has increasingly been implicated as a prominent histopathological feature of AD and related dementias. Our recent studies have implicated mitochondria as critical targets of TDP-43 neurotoxicity. Here, we demonstrate that the suppression of mitochondrial-associated TDP-43 protects against neuronal loss and behavioral deficits in 5XFAD transgenic mice recapitulating AD-related phenotypes. In AD patients and 5XFAD mice, the level of TDP-43 is increased in mitochondria, and TDP-43 highly co-localizes with mitochondria in brain neurons exhibiting TDP-43 proteinopathy. Chronic administration of a TDP-43 mitochondrial localization inhibitory peptide, PM1, significantly alleviates TDP-43 proteinopathy, mitochondrial abnormalities, microgliosis and even neuronal loss without effect on amyloid plaque load in 12-month-old 5XFAD mice well after the onset of symptoms. Additionally, PM1 also improves the cognitive and motor function in 12-month-old 5XFAD mice and completely prevents the onset of mild cognitive impairment in 6-month-old 5XFAD mice. These data indicate that mitochondria-associated TDP-43 is likely involved in AD pathogenesis and that the inhibitor of mitochondria-associated TDP-43 may be a valuable drug to treat underlying AD.
Collapse
Affiliation(s)
- Ju Gao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Luwen Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Chao Gao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Hiroyuki Arakawa
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Xinglong Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA; Center for Mitochondrial Diseases, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
270
|
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: 53] [Impact Index Per Article: 10.6] [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
|
271
|
Schmidt HB, Barreau A, Rohatgi R. Phase separation-deficient TDP43 remains functional in splicing. Nat Commun 2019; 10:4890. [PMID: 31653829 PMCID: PMC6814767 DOI: 10.1038/s41467-019-12740-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 09/27/2019] [Indexed: 12/12/2022] Open
Abstract
Intrinsically disordered regions (IDRs) are often fast-evolving protein domains of low sequence complexity that can drive phase transitions and are commonly found in many proteins associated with neurodegenerative diseases, including the RNA processing factor TDP43. Yet, how phase separation contributes to the physiological functions of TDP43 in cells remains enigmatic. Here, we combine systematic mutagenesis guided by evolutionary sequence analysis with a live-cell reporter assay of TDP43 phase dynamics to identify regularly-spaced hydrophobic motifs separated by flexible, hydrophilic segments in the IDR as a key determinant of TDP43 phase properties. This heuristic framework allows customization of the material properties of TDP43 condensates to determine effects on splicing function. Remarkably, even a mutant that fails to phase-separate at physiological concentrations can still efficiently mediate the splicing of a quantitative, single-cell splicing reporter and endogenous targets. This suggests that the ability of TDP43 to phase-separate is not essential for its splicing function.
Collapse
Affiliation(s)
| | - Ariana Barreau
- Department of Biochemistry, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Rajat Rohatgi
- Department of Biochemistry, Stanford School of Medicine, Stanford, CA, 94305, USA.
- Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
272
|
Abstract
Motor neuron disorders are highly debilitating and mostly fatal conditions for which only limited therapeutic options are available. To overcome this limitation and develop more effective therapeutic strategies, it is critical to discover the pathogenic mechanisms that trigger and sustain motor neuron degeneration with the greatest accuracy and detail. In the case of Amyotrophic Lateral Sclerosis (ALS), several genes have been associated with familial forms of the disease, whilst the vast majority of cases develop sporadically and no defined cause can be held responsible. On the contrary, the huge majority of Spinal Muscular Atrophy (SMA) occurrences are caused by loss-of-function mutations in a single gene, SMN1. Although the typical hallmark of both diseases is the loss of motor neurons, there is increasing awareness that pathological lesions are also present in the neighbouring glia, whose dysfunction clearly contributes to generating a toxic environment in the central nervous system. Here, ALS and SMA are sequentially presented, each disease section having a brief introduction, followed by a focussed discussion on the role of the astrocytes in the disease pathogenesis. Such a dissertation is substantiated by the findings that built awareness on the glial involvement and how the glial-neuronal interplay is perturbed, along with the appraisal of this new cellular site for possible therapeutic intervention.
Collapse
|
273
|
Jinn S, Blauwendraat C, Toolan D, Gretzula CA, Drolet RE, Smith S, Nalls MA, Marcus J, Singleton AB, Stone DJ. Functionalization of the TMEM175 p.M393T variant as a risk factor for Parkinson disease. Hum Mol Genet 2019; 28:3244-3254. [PMID: 31261387 PMCID: PMC6859430 DOI: 10.1093/hmg/ddz136] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 06/07/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple genome-wide association studies (GWAS) in Parkinson disease (PD) have identified a signal at chromosome 4p16.3; however, the causal variant has not been established for this locus. Deep investigation of the region resulted in one identified variant, the rs34311866 missense SNP (p.M393T) in TMEM175, which is 20 orders of magnitude more significant than any other SNP in the region. Because TMEM175 is a lysosomal gene that has been shown to influence α-synuclein phosphorylation and autophagy, the p.M393T variant is an attractive candidate, and we have examined its effect on TMEM175 protein and PD-related biology. After knocking down each of the genes located under the GWAS peak via multiple shRNAs, only TMEM175 was found to consistently influence accumulation of phosphorylated α-synuclein (p-α-syn). Examination of the p.M393T variant showed effects on TMEM175 function that were intermediate between the wild-type (WT) and knockout phenotypes, with reduced regulation of lysosomal pH in response to starvation and minor changes in clearance of autophagy substrates, reduced lysosomal localization, and increased accumulation of p-α-syn. Finally, overexpression of WT TMEM175 protein reduced p-α-syn, while overexpression of the p.M393T variant resulted in no change in α-synuclein phosphorylation. These results suggest that the main signal in the chromosome 4p16.3 PD risk locus is driven by the TMEM175 p.M393T variant. Modulation of TMEM175 may impact α-synuclein biology and therefore may be a rational therapeutic strategy for PD.
Collapse
Affiliation(s)
- Sarah Jinn
- Merck & Co., Inc., West Point, PA 19486, USA
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Dawn Toolan
- Merck & Co., Inc., West Point, PA 19486, USA
| | | | | | - Sean Smith
- Merck & Co., Inc., West Point, PA 19486, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | | | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
274
|
Roczniak-Ferguson A, Ferguson SM. Pleiotropic requirements for human TDP-43 in the regulation of cell and organelle homeostasis. Life Sci Alliance 2019; 2:2/5/e201900358. [PMID: 31527135 PMCID: PMC6749094 DOI: 10.26508/lsa.201900358] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 08/26/2019] [Accepted: 09/02/2019] [Indexed: 12/12/2022] Open
Abstract
TDP-43 is an RNA-binding protein that forms cytoplasmic aggregates in multiple neurodegenerative diseases. Although the loss of normal TDP-43 functions likely contributes to disease pathogenesis, the cell biological consequences of human TDP-43 depletion are not well understood. We, therefore, generated human TDP-43 knockout (KO) cells and subjected them to parallel cell biological and transcriptomic analyses. These efforts yielded three important discoveries. First, complete loss of TDP-43 resulted in widespread morphological defects related to multiple organelles, including Golgi, endosomes, lysosomes, mitochondria, and the nuclear envelope. Second, we identified a new role for TDP-43 in controlling mRNA splicing of Nup188 (nuclear pore protein). Third, analysis of multiple amyotrophic lateral sclerosis causing TDP-43 mutations revealed a broad ability to support splicing of TDP-43 target genes. However, as some TDP-43 disease-causing mutants failed to fully support the regulation of specific target transcripts, our results raise the possibility of mutation-specific loss-of-function contributions to disease pathology.
Collapse
Affiliation(s)
- Agnes Roczniak-Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Shawn M Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA .,Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
275
|
Imaging Hypoxic Stress and the Treatment of Amyotrophic Lateral Sclerosis with Dimethyloxalylglycine in a Mice Model. Neuroscience 2019; 415:31-43. [DOI: 10.1016/j.neuroscience.2019.06.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/15/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022]
|
276
|
Neelagandan N, Gonnella G, Dang S, Janiesch PC, Miller KK, Küchler K, Marques RF, Indenbirken D, Alawi M, Grundhoff A, Kurtz S, Duncan KE. TDP-43 enhances translation of specific mRNAs linked to neurodegenerative disease. Nucleic Acids Res 2019; 47:341-361. [PMID: 30357366 PMCID: PMC6326785 DOI: 10.1093/nar/gky972] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
The RNA-binding protein TDP-43 is heavily implicated in neurodegenerative disease. Numerous patient mutations in TARDBP, the gene encoding TDP-43, combined with data from animal and cell-based models, imply that altered RNA regulation by TDP-43 causes Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. However, underlying mechanisms remain unresolved. Increased cytoplasmic TDP-43 levels in diseased neurons suggest a possible role in this cellular compartment. Here, we examined the impact on translation of overexpressing human TDP-43 and the TDP-43A315T patient mutant protein in motor neuron-like cells and primary cultures of cortical neurons. In motor-neuron like cells, TDP-43 associates with ribosomes without significantly affecting global translation. However, ribosome profiling and additional assays revealed enhanced translation and direct binding of Camta1, Mig12, and Dennd4a mRNAs. Overexpressing either wild-type TDP-43 or TDP-43A315T stimulated translation of Camta1 and Mig12 mRNAs via their 5'UTRs and increased CAMTA1 and MIG12 protein levels. In contrast, translational enhancement of Dennd4a mRNA required a specific 3'UTR region and was specifically observed with the TDP-43A315T patient mutant allele. Our data reveal that TDP-43 can function as an mRNA-specific translational enhancer. Moreover, since CAMTA1 and DENND4A are linked to neurodegeneration, they suggest that this function could contribute to disease.
Collapse
Affiliation(s)
- Nagammal Neelagandan
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Giorgio Gonnella
- Universität Hamburg, MIN-Fakultät, ZBH-Center for Bioinformatics, Hamburg 20146, Germany
| | - Stefan Dang
- Universität Hamburg, MIN-Fakultät, ZBH-Center for Bioinformatics, Hamburg 20146, Germany
| | - Philipp C Janiesch
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Katharine K Miller
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Katrin Küchler
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Rita F Marques
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Daniela Indenbirken
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Malik Alawi
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany.,Bioinformatics Core, University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| | - Adam Grundhoff
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg 20251, Germany
| | - Stefan Kurtz
- Universität Hamburg, MIN-Fakultät, ZBH-Center for Bioinformatics, Hamburg 20146, Germany
| | - Kent E Duncan
- Neuronal Translational Control Research Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg 20251, Germany
| |
Collapse
|
277
|
Gao J, Wang L, Yan T, Perry G, Wang X. TDP-43 proteinopathy and mitochondrial abnormalities in neurodegeneration. Mol Cell Neurosci 2019; 100:103396. [PMID: 31445085 DOI: 10.1016/j.mcn.2019.103396] [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] [Received: 04/10/2019] [Revised: 07/03/2019] [Accepted: 08/17/2019] [Indexed: 12/12/2022] Open
Abstract
Genetic mutations in TAR DNA-binding protein 43 (TDP-43) cause amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Importantly, TDP-43 proteinopathy, characterized by aberrant phosphorylation, ubiquitination, cleavage or nuclear depletion of TDP-43 in neurons and glial cells, is a common prominent pathological feature of various major neurodegenerative diseases including ALS, FTD, and Alzheimer's disease (AD). Although the pathomechanisms underlying TDP-43 proteinopathy remain elusive, pathologically relevant TDP-43 has been repeatedly shown to be present in either the inside or outside of mitochondria, and functionally involved in the regulation of mitochondrial morphology, trafficking, and function, suggesting mitochondria as likely targets of TDP-43 proteinopathy. In this review, we first describe the current knowledge of the association of TDP-43 with mitochondria. We then review in detail multiple mitochondrial pathways perturbed by pathological TDP-43, including mitochondrial fission and fusion dynamics, mitochondrial trafficking, bioenergetics, and mitochondrial quality control. Lastly, we briefly discuss how the study of TDP-43 proteinopathy and mitochondrial abnormalities may provide new avenues for neurodegeneration therapeutics.
Collapse
Affiliation(s)
- Ju Gao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Luwen Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Tingxiang Yan
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Xinglong Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
278
|
Cobos SN, Bennett SA, Torrente MP. The impact of histone post-translational modifications in neurodegenerative diseases. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1982-1991. [PMID: 30352259 PMCID: PMC6475498 DOI: 10.1016/j.bbadis.2018.10.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/05/2018] [Accepted: 10/11/2018] [Indexed: 02/08/2023]
Abstract
Every year, neurodegenerative disorders take more than 5000 lives in the US alone. Cures have not yet been found for many of the multitude of neuropathies. The majority of amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD) and Parkinson's disease (PD) cases have no known genetic basis. Thus, it is evident that contemporary genetic approaches have failed to explain the etiology or etiologies of ALS/FTD and PD. Recent investigations have explored the potential role of epigenetic mechanisms in disease development. Epigenetics comprises heritable changes in gene utilization that are not derived from changes in the genome. A main epigenetic mechanism involves the post-translational modification of histones. Increased knowledge of the epigenomic landscape of neurodegenerative diseases would not only further our understanding of the disease pathologies, but also lead to the development of treatments able to halt their progress. Here, we review recent advances on the association of histone post-translational modifications with ALS, FTD, PD and several ataxias.
Collapse
Affiliation(s)
- Samantha N Cobos
- Chemistry Department of Brooklyn College, Brooklyn, New York 11210, United States; Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States
| | - Seth A Bennett
- Chemistry Department of Brooklyn College, Brooklyn, New York 11210, United States; Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, United States
| | - Mariana P Torrente
- Chemistry Department of Brooklyn College, Brooklyn, New York 11210, United States; Ph.D. Programs in Chemistry, Biochemistry, and Biology, The Graduate Center of the City University of New York, New York 10016, United States.
| |
Collapse
|
279
|
Formicola N, Vijayakumar J, Besse F. Neuronal ribonucleoprotein granules: Dynamic sensors of localized signals. Traffic 2019; 20:639-649. [PMID: 31206920 DOI: 10.1111/tra.12672] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/03/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022]
Abstract
Membrane-less organelles, because of their capacity to dynamically, selectively and reversibly concentrate molecules, are very well adapted for local information processing and rapid response to environmental fluctuations. These features are particularly important in the context of neuronal cells, where synapse-specific activation, or localized extracellular cues, induce signaling events restricted to specialized axonal or dendritic subcompartments. Neuronal ribonucleoprotein (RNP) particles, or granules, are nonmembrane bound macromolecular condensates that concentrate specific sets of mRNAs and regulatory proteins, promoting their long-distance transport to axons or dendrites. Neuronal RNP granules also have a dual function in regulating the translation of associated mRNAs: while preventing mRNA translation at rest, they fuel local protein synthesis upon activation. As revealed by recent work, rapid and reversible switches between these two functional modes are triggered by modifications of the networks of interactions underlying RNP granule assembly. Such flexible properties also come with a cost, as neuronal RNP granules are prone to transition into pathological aggregates in response to mutations, aging, or cellular stresses, further emphasizing the need to better understand the mechanistic principles governing their dynamic assembly and regulation in living systems.
Collapse
|
280
|
Renaud L, Picher-Martel V, Codron P, Julien JP. Key role of UBQLN2 in pathogenesis of amyotrophic lateral sclerosis and frontotemporal dementia. Acta Neuropathol Commun 2019; 7:103. [PMID: 31319884 PMCID: PMC6889556 DOI: 10.1186/s40478-019-0758-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Accepted: 06/22/2019] [Indexed: 12/11/2022] Open
Abstract
Ubiquilin-2 (UBQLN2) is a member of the ubiquilin family, actively implicated in the degradation of misfolded and redundant proteins through the ubiquitin-proteasome system and macroautophagy. UBQLN2 received much attention after the discovery of gene mutations in amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD). The abnormal presence of positive UBQLN2 inclusion in the cytosol of degenerating motor neurons of familial and sporadic forms of ALS patients has been newly related to neurodegeneration. Only recently, data have emerged on its role in liquid-liquid phase separation, in stress granule development and in the formation of secondary amyloid structures. Furthermore, several animal models are available to investigate its involvement in TDP-43 pathology and neuroinflammation in ALS. This review addresses the molecular pathogenetic pathways involving UBQLN2 abnormalities which are converging toward defects in clearance mechanisms. UBQLN2.
Collapse
|
281
|
Kim H, Lim J, Bao H, Jiao B, Canon SM, Epstein MP, Xu K, Jiang J, Parameswaran J, Li Y, Moberg KH, Landers JE, Fournier C, Allen EG, Glass JD, Wingo TS, Jin P. Rare variants in MYH15 modify amyotrophic lateral sclerosis risk. Hum Mol Genet 2019; 28:2309-2318. [PMID: 30985904 PMCID: PMC6606848 DOI: 10.1093/hmg/ddz063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/14/2019] [Accepted: 03/21/2019] [Indexed: 11/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurological disorder characterized by progressive muscular atrophy and respiratory failure. The G4C2 repeat expansion in the C9orf72 gene is the most prevalent genetic risk for ALS. Mutation carriers (C9ALS) display variability in phenotypes such as age-at-onset and duration, suggesting the existence of additional genetic factors. Here we introduce a three-step gene discovery strategy to identify genetic factors modifying the risk of both C9ALS and sporadic ALS (sALS) using limited samples. We first identified 135 candidate genetic modifiers of C9ALS using whole-genome sequencing (WGS) of extreme C9ALS cases diagnosed ~30 years apart. We then performed an unbiased genetic screen using a Drosophila model of the G4C2 repeat expansion with the genes identified from WGS analysis. This genetic screen identified the novel genetic interaction between G4C2 repeat-associated toxicity and 18 genetic factors, suggesting their potential association with C9ALS risk. We went on to test if 14 out of the 18 genes, those which were not known to be risk factors for ALS previously, are also associated with ALS risk in sALS cases. Gene-based-statistical analyses of targeted resequencing and WGS were performed. These analyses together reveal that rare variants in MYH15 represent a likely genetic risk factor for ALS. Furthermore, we show that MYH15 could modulate the toxicity of dipeptides produced from expanded G4C2 repeat. Our study presented here demonstrates the power of combining WGS with fly genetics to facilitate the discovery of fundamental genetic components of complex traits with a limited number of samples.
Collapse
Affiliation(s)
- Hyerim Kim
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
- Cancer Biology Program, Emory University, Atlanta, GA, USA
| | - Junghwa Lim
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Han Bao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Bin Jiao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Se Min Canon
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Keqin Xu
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jie Jiang
- Department of Cell Biology, Emory University and Emory University School of Medicine, Atlanta, GA, USA
| | - Janani Parameswaran
- Department of Cell Biology, Emory University and Emory University School of Medicine, Atlanta, GA, USA
| | - Yingjie Li
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Kenneth H Moberg
- Department of Cell Biology, Emory University and Emory University School of Medicine, Atlanta, GA, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christina Fournier
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Division of Neurology, Atlanta VA Medical Center, Decatur, GA, USA
| | - Emily G Allen
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Thomas S Wingo
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Division of Neurology, Atlanta VA Medical Center, Decatur, GA, USA
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
282
|
Sugai A, Kato T, Koyama A, Koike Y, Konno T, Ishihara T, Onodera O. Non-genetically modified models exhibit TARDBP mRNA increase due to perturbed TDP-43 autoregulation. Neurobiol Dis 2019; 130:104534. [PMID: 31310801 DOI: 10.1016/j.nbd.2019.104534] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/22/2019] [Accepted: 07/12/2019] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by accumulation of fragmented insoluble TDP-43 and loss of TDP-43 from the nucleus. Increased expression of exogenous TARDBP (encoding TDP-43) induces TDP-43 pathology and cytotoxicity, suggesting the involvement of aberrant expression of TDP-43 in the pathogenesis of ALS. In normal conditions, however, the amount of TDP-43 is tightly regulated by the autoregulatory mechanism involving alternative splicing of TARDBP mRNA. To investigate the influence of autoregulation dysfunction, we inhibited the splicing of cryptic intron 6 using antisense oligonucleotides in vivo. This inhibition doubled the Tardbp mRNA expression, increased the fragmented insoluble TDP-43, and reduced the number of motor neurons in the mouse spinal cord. In human induced pluripotent stem cell-derived neurons, the splicing inhibition of intron 6 increased TARDBP mRNA and decreased nuclear TDP-43. These non-genetically modified models exhibiting rise in the TARDBP mRNA levels suggest that TDP-43 autoregulation turbulence might be linked to the pathogenesis of ALS.
Collapse
Affiliation(s)
- Akihiro Sugai
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Taisuke Kato
- Department of System Pathology for Neurological Disorders, Brain Science Branch, Center for Bioresource-based Research, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Akihide Koyama
- Division of Legal Medicine, Graduate School of Medicine and Dental Science, Niigata University, Niigata 951-8585, Japan
| | - Yuka Koike
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Takuya Konno
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Tomohiko Ishihara
- Department of Molecular Neuroscience, Resource Branch for Brain Disease Research, Center for Bioresource-based Research, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Osamu Onodera
- Department of Neurology, Clinical Neuroscience Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan.
| |
Collapse
|
283
|
Kim S, Chung AY, Na JE, Lee SJ, Jeong SH, Kim E, Sun W, Rhyu IJ, Park HC. Myelin degeneration induced by mutant superoxide dismutase 1 accumulation promotes amyotrophic lateral sclerosis. Glia 2019; 67:1910-1921. [PMID: 31290185 DOI: 10.1002/glia.23669] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 05/16/2019] [Accepted: 06/18/2019] [Indexed: 11/11/2022]
Abstract
Myelin is a specialized membrane that wraps around nerve fibers and is essential for normal axonal conduction in neurons. In the central nervous system, oligodendrocytes are responsible for myelin formation. Recent studies have reported pathological abnormalities in oligodendrocytes in human patients with amyotrophic lateral sclerosis (ALS) and a mouse model of ALS expressing the G93A mutation of the human superoxide dismutase 1 (mtSOD1). However, it is unclear whether oligodendrocyte pathology in ALS represents the primary dysfunction induced by mtSOD1 and how mtSOD1 contributes to oligodendrocyte degeneration and ALS pathogenesis. We analyzed GAL4-VP16-UAS transgenic zebrafish selectively expressing mtSOD1 in mature oligodendrocytes. We observed that mtSOD1 directly induced oligodendrocyte degeneration by disrupting the myelin sheath and downregulating monocarboxylate transporter 1 (MCT1), thereby causing spinal motor neuron degeneration. Pathological changes observed in this transgenic zebrafish were similar to the pathology observed in the SOD1G93A mouse model of ALS, which is characterized by expression of mtSOD1 in all cells. In addition, oligodendrocyte dysfunction induced by mtSOD1 was associated with anxiety-related behavioral abnormalities, learning impairments, and motor defects in the early symptomatic stage. We also found that treatment with potassium channel inhibitors rescued behavioral abnormalities without rescuing MCT1 expression, suggesting that myelin disruption induces behavioral abnormalities independently of MCT1. These results indicate that mtSOD1-induced dysfunction of mature oligodendrocytes is sufficient to induce motor neuron degeneration, thus informing future therapeutic strategies targeted at oligodendrocytes in ALS.
Collapse
Affiliation(s)
- Suhyun Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, South Korea
| | - Ah-Young Chung
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, South Korea
| | - Ji E Na
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Se J Lee
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Sang H Jeong
- Biomedical Research Center, Korea University Ansan hospital, Ansan, South Korea
| | - Eunmi Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, South Korea
| | - Woong Sun
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Im J Rhyu
- Department of Anatomy, College of Medicine, Korea University, Seoul, South Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, South Korea
| |
Collapse
|
284
|
Extracellular vesicles and their diagnostic potential in amyotrophic lateral sclerosis. Clin Chim Acta 2019; 497:27-34. [PMID: 31301281 DOI: 10.1016/j.cca.2019.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/09/2019] [Accepted: 07/09/2019] [Indexed: 01/15/2023]
Abstract
Extracellular vesicles, small reservoirs that carry various biomolecules, have gained significant interest from the clinical field in recent years based on the diagnostic, therapeutic and prognostic possibilities they offer. While information abound regarding the clinical potential of such vesicles in diverse conditions, the information demonstrating their likely importance in amyotrophic lateral sclerosis (ALS) is more limited. This review will thus provide a brief introduction to extracellular vesicles, highlight their diagnostic significance in various diseases with a focus on ALS and explore additional applications of extracellular vesicles in the medical field. Overall, this work sheds further light on the clinical importance of extracellular vesicles in diagnostic applications as well as supports the need to better characterize their roles and signatures in patients diagnosed with ALS.
Collapse
|
285
|
Vega MV, Nigro A, Luti S, Capitini C, Fani G, Gonnelli L, Boscaro F, Chiti F. Isolation and characterization of soluble human full‐length TDP‐43 associated with neurodegeneration. FASEB J 2019; 33:10780-10793. [DOI: 10.1096/fj.201900474r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Mirella Vivoli Vega
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Alessia Nigro
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Simone Luti
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Claudia Capitini
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Giulia Fani
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| | - Leonardo Gonnelli
- Centro di Ricerca di Risonanze Magnetiche (CERM)University of FlorenceFlorenceItaly
| | | | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical SciencesUniversity of FlorenceFlorenceItaly
| |
Collapse
|
286
|
DNA repair and neurological disease: From molecular understanding to the development of diagnostics and model organisms. DNA Repair (Amst) 2019; 81:102669. [PMID: 31331820 DOI: 10.1016/j.dnarep.2019.102669] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In both replicating and non-replicating cells, the maintenance of genomic stability is of utmost importance. Dividing cells can repair DNA damage during cell division, tolerate the damage by employing potentially mutagenic DNA polymerases or die via apoptosis. However, the options for accurate DNA repair are more limited in non-replicating neuronal cells. If DNA damage is left unresolved, neuronal cells die causing neurodegenerative disorders. A number of pathogenic variants of DNA repair proteins have been linked to multiple neurological diseases. The current challenge is to harness our knowledge of fundamental properties of DNA repair to improve diagnosis, prognosis and treatment of such debilitating disorders. In this perspective, we will focus on recent efforts in identifying novel DNA repair biomarkers for the diagnosis of neurological disorders and their use in monitoring the patient response to therapy. These efforts are greatly facilitated by the development of model organisms such as zebrafish, which will also be summarised.
Collapse
|
287
|
Foster A, Scott D, Layfield R, Rea S. An FTLD-associated SQSTM1 variant impacts Nrf2 and NF-κB signalling and is associated with reduced phosphorylation of p62. Mol Cell Neurosci 2019; 98:32-45. [DOI: 10.1016/j.mcn.2019.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022] Open
|
288
|
ALS and FTD: Where RNA metabolism meets protein quality control. Semin Cell Dev Biol 2019; 99:183-192. [PMID: 31254610 DOI: 10.1016/j.semcdb.2019.06.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 12/12/2022]
Abstract
Recent genetic and biochemical evidence has improved our understanding of the pathomechanisms that lead to amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), two devastating neurodegenerative diseases with overlapping symptoms and causes. Impaired RNA metabolism, enhanced aggregation of protein-RNA complexes, aberrant formation of ribonucleoprotein (RNP) granules and dysfunctional protein clearance via autophagy are emerging as crucial events in ALS/FTD pathogenesis. Importantly, these processes interact at the molecular level, converging on a common pathogenic cascade. In this review, we summarize key principles underlying ALS and FTD, and we discuss how mutations in genes involved in RNA metabolism, protein quality control and protein degradation meet mechanistically to impair the functionality and dynamics of RNP granules, and how this leads to cellular toxicity and death. Finally, we describe recent advances in understanding signaling pathways that become dysfunctional in ALS/FTD, partly due to altered RNP granule dynamics, but also with stress granule-independent mechanisms and, thus could be promising targets for future therapeutic intervention.
Collapse
|
289
|
Taylor LM, McMillan PJ, Kraemer BC, Liachko NF. Tau tubulin kinases in proteinopathy. FEBS J 2019; 286:2434-2446. [PMID: 31034749 PMCID: PMC6936727 DOI: 10.1111/febs.14866] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/23/2019] [Accepted: 04/25/2019] [Indexed: 12/12/2022]
Abstract
A number of neurodegenerative diseases are characterized by deposition of abnormally phosphorylated tau or TDP-43 in disease-affected neurons. These diseases include Alzheimer's disease, frontotemporal lobar degeneration, and amyotrophic lateral sclerosis. No disease-modifying therapeutics is available to treat these disorders, and we have a limited understanding of the cellular and molecular factors integral to disease initiation or progression. Phosphorylated tau and TDP-43 are important markers of pathology in dementia disorders and directly contribute to tau- and TDP-43-related neurotoxicity and neurodegeneration. Here, we review the scope of tau and TDP-43 phosphorylation in neurodegenerative disease and discuss recent work demonstrating the kinases TTBK1 and TTBK2 phosphorylate both tau and TDP-43, promoting neurodegeneration.
Collapse
Affiliation(s)
- Laura M Taylor
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Pamela J McMillan
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
| | - Brian C Kraemer
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, Washington, USA
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Nicole F Liachko
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| |
Collapse
|
290
|
Huntley ML, Gao J, Termsarasab P, Wang L, Zeng S, Thammongkolchai T, Liu Y, Cohen ML, Wang X. Association between TDP-43 and mitochondria in inclusion body myositis. J Transl Med 2019; 99:1041-1048. [PMID: 30742062 PMCID: PMC6609472 DOI: 10.1038/s41374-019-0233-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/12/2022] Open
Abstract
Inclusion body myositis (IBM) is the most common cause of primary myopathy in individuals aged 50 years and over, and is pathologically characterized by protein aggregates of p62 and mislocalized cytoplasmic TDP-43, as well as mitochondrial abnormalities in affected muscle fibers. Our recent studies have shown the accumulation of TDP-43 in mitochondria in neurons from patients with amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD), and revealed mitochondria as critical mediators of TDP-43 neurotoxicity. In this study, we investigated the association between mitochondria and TDP-43 in biopsied skeletal muscle samples from IBM patients. We found that IBM pathological markers TDP-43, phosphorylated TDP-43, and p62 all coexisted with intensively stained key subunits of mitochondrial oxidative phosphorylation complexes I-V in the same skeletal muscle fibers of patients with IBM. Further immunoblot analysis showed increased levels of TDP-43, truncated TDP-43, phosphorylated TDP-43, and p62, but decreased levels of key subunits of mitochondrial oxidative phosphorylation complexes I and III in IBM patients compared to aged matched control subjects. This is the first demonstration of the close association of TDP-43 accumulation with mitochondria in degenerating muscle fibers in IBM and this association may contribute to the development of mitochondrial dysfunction and pathological protein aggregates.
Collapse
Affiliation(s)
- Mikayla L. Huntley
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Ju Gao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Pichet Termsarasab
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Luwen Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Sophia Zeng
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | | | - Ying Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Mark L. Cohen
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Xinglong Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA. .,Center for Mitochondrial Diseases, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
291
|
Sawada H. Considerations for pharmacotherapy use in patients with amyotrophic lateral sclerosis: the earlier it starts, the better the results. Expert Opin Pharmacother 2019; 20:1671-1674. [PMID: 31256690 DOI: 10.1080/14656566.2019.1636965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Hideyuki Sawada
- Chair of Department of Neurology, Utano National Hospital, National Hospital Organization , Kyoto , Japan
| |
Collapse
|
292
|
Ragagnin AMG, Shadfar S, Vidal M, Jamali MS, Atkin JD. Motor Neuron Susceptibility in ALS/FTD. Front Neurosci 2019; 13:532. [PMID: 31316328 PMCID: PMC6610326 DOI: 10.3389/fnins.2019.00532] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/08/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the death of both upper and lower motor neurons (MNs) in the brain, brainstem and spinal cord. The neurodegenerative mechanisms leading to MN loss in ALS are not fully understood. Importantly, the reasons why MNs are specifically targeted in this disorder are unclear, when the proteins associated genetically or pathologically with ALS are expressed ubiquitously. Furthermore, MNs themselves are not affected equally; specific MNs subpopulations are more susceptible than others in both animal models and human patients. Corticospinal MNs and lower somatic MNs, which innervate voluntary muscles, degenerate more readily than specific subgroups of lower MNs, which remain resistant to degeneration, reflecting the clinical manifestations of ALS. In this review, we discuss the possible factors intrinsic to MNs that render them uniquely susceptible to neurodegeneration in ALS. We also speculate why some MN subpopulations are more vulnerable than others, focusing on both their molecular and physiological properties. Finally, we review the anatomical network and neuronal microenvironment as determinants of MN subtype vulnerability and hence the progression of ALS.
Collapse
Affiliation(s)
- Audrey M G Ragagnin
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sina Shadfar
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Marta Vidal
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Md Shafi Jamali
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Julie D Atkin
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
293
|
Gentile F, Scarlino S, Falzone YM, Lunetta C, Tremolizzo L, Quattrini A, Riva N. The Peripheral Nervous System in Amyotrophic Lateral Sclerosis: Opportunities for Translational Research. Front Neurosci 2019; 13:601. [PMID: 31293369 PMCID: PMC6603245 DOI: 10.3389/fnins.2019.00601] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/27/2019] [Indexed: 12/11/2022] Open
Abstract
Although amyotrophic lateral sclerosis (ALS) has been considered as a disorder of the motor neuron (MN) cell body, recent evidences show the non-cell-autonomous pathogenic nature of the disease. Axonal degeneration, loss of peripheral axons and destruction of nerve terminals are early events in the disease pathogenic cascade, anticipating MN degeneration, and the onset of clinical symptoms. Therefore, although ALS and peripheral axonal neuropathies should be differentiated in clinical practice, they also share damage to common molecular pathways, including axonal transport, RNA metabolism and proteostasis. Thus, an extensive evaluation of the molecular events occurring in the peripheral nervous system (PNS) could be fundamental to understand the pathogenic mechanisms of ALS, favoring the discovery of potential disease biomarkers, and new therapeutic targets.
Collapse
Affiliation(s)
- Francesco Gentile
- Experimental Neuropathology Unit, Division of Neuroscience, Institute of Experimental Neurology – San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Scarlino
- Experimental Neuropathology Unit, Division of Neuroscience, Institute of Experimental Neurology – San Raffaele Scientific Institute, Milan, Italy
| | - Yuri Matteo Falzone
- Experimental Neuropathology Unit, Division of Neuroscience, Institute of Experimental Neurology – San Raffaele Scientific Institute, Milan, Italy
- Department of Neurology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Lucio Tremolizzo
- Neurology Unit, ALS Clinic, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Angelo Quattrini
- Experimental Neuropathology Unit, Division of Neuroscience, Institute of Experimental Neurology – San Raffaele Scientific Institute, Milan, Italy
| | - Nilo Riva
- Experimental Neuropathology Unit, Division of Neuroscience, Institute of Experimental Neurology – San Raffaele Scientific Institute, Milan, Italy
- Department of Neurology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
294
|
Petel Légaré V, Harji ZA, Rampal CJ, Allard-Chamard X, Rodríguez EC, Armstrong GAB. Augmentation of spinal cord glutamatergic synaptic currents in zebrafish primary motoneurons expressing mutant human TARDBP (TDP-43). Sci Rep 2019; 9:9122. [PMID: 31235725 PMCID: PMC6591224 DOI: 10.1038/s41598-019-45530-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/06/2019] [Indexed: 12/12/2022] Open
Abstract
Though there is compelling evidence that de-innervation of neuromuscular junctions (NMJ) occurs early in amyotrophic lateral sclerosis (ALS), defects arising at synapses in the spinal cord remain incompletely understood. To investigate spinal cord synaptic dysfunction, we took advantage of a zebrafish larval model and expressed either wild type human TARDBP (wtTARDBP) or the ALS-causing G348C variant (mutTARDBP). The larval zebrafish is ideally suited to examine synaptic connectivity between descending populations of neurons and spinal cord motoneurons as a fully intact spinal cord is preserved during experimentation. Here we provide evidence that the tail-beat motor pattern is reduced in both frequency and duration in larvae expressing mutTARDBP. In addition, we report that motor-related synaptic depolarizations in primary motoneurons of the spinal cord are shorter in duration and fewer action potentials are evoked in larvae expressing mutTARDBP. To more thoroughly examine spinal cord synaptic dysfunction in our ALS model, we isolated AMPA/kainate-mediated glutamatergic miniature excitatory post-synaptic currents in primary motoneurons and found that in addition to displaying a larger amplitude, the frequency of quantal events was higher in larvae expressing mutTARDBP when compared to larvae expressing wtTARDBP. In a final series of experiments, we optogenetically drove neuronal activity in the hindbrain and spinal cord population of descending ipsilateral glutamatergic interneurons (expressing Chx10) using the Gal4-UAS system and found that larvae expressing mutTARDBP displayed abnormal tail-beat patterns in response to optogenetic stimuli and augmented synaptic connectivity with motoneurons. These findings indicate that expression of mutTARDBP results in functionally altered glutamatergic synapses in the spinal cord.
Collapse
Affiliation(s)
- Virginie Petel Légaré
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University, Montreal, Canada
| | - Ziyaan A Harji
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University, Montreal, Canada
| | - Christian J Rampal
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University, Montreal, Canada
| | - Xavier Allard-Chamard
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University, Montreal, Canada
| | - Esteban C Rodríguez
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University, Montreal, Canada
| | - Gary A B Armstrong
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, Faculty of Medicine, McGill University, Montreal, Canada.
| |
Collapse
|
295
|
Sprovieri T, Ungaro C, Perrone B, Naimo GD, Spataro R, Cavallaro S, La Bella V, Conforti FL. A novel S379A TARDBP mutation associated to late-onset sporadic ALS. Neurol Sci 2019; 40:2111-2118. [DOI: 10.1007/s10072-019-03943-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/16/2019] [Indexed: 01/05/2023]
|
296
|
Yin P, Guo X, Yang W, Yan S, Yang S, Zhao T, Sun Q, Liu Y, Li S, Li XJ. Caspase-4 mediates cytoplasmic accumulation of TDP-43 in the primate brains. Acta Neuropathol 2019; 137:919-937. [PMID: 30810811 PMCID: PMC6531422 DOI: 10.1007/s00401-019-01979-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
The cytoplasmic accumulation of the nuclear TAR DNA-binding protein 43 (TDP-43) is a pathologic hallmark in amyotrophic lateral sclerosis, frontotemporal lobar degeneration, and other neurological disorders. However, most transgenic TDP-43 rodent models show predominant nuclear distribution of TDP-43 in the brain. By expressing mutant TDP-43 (M337V) in the brains of rhesus monkeys and mice, we verified that mutant TDP-43 is distributed in the cytoplasm of the monkey brain and that the majority of mutant TDP-43 remains in the nuclei of the mouse brain. The primate-specific caspase-4, but not mouse homologue caspase-11, could remove the NLS-containing N-terminal domain and generate fragmented TDP-43 that accumulates in the cytoplasm. Moreover, increased expression of caspase-4 in the monkey brain promotes the cytoplasmic accumulation of endogenous TDP-43, and suppressing caspase-4 reduces the cytoplasmic distribution of endogenous TDP-43 in cultured human neural cells. Our findings suggest that primate-specific caspase-4-mediated cleavage of TDP-43 accounts for its cytoplasmic mislocalization in the primate brains and may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Peng Yin
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Xiangyu Guo
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Weili Yang
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Sen Yan
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Su Yang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ting Zhao
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Qiang Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yunbo Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Shihua Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Xiao-Jiang Li
- Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
297
|
Gertsman I, Wuu J, McAlonis-Downes M, Ghassemian M, Ling K, Rigo F, Bennett F, Benatar M, Miller TM, Da Cruz S. An endogenous peptide marker differentiates SOD1 stability and facilitates pharmacodynamic monitoring in SOD1 amyotrophic lateral sclerosis. JCI Insight 2019; 4:122768. [PMID: 31092730 DOI: 10.1172/jci.insight.122768] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
The discovery of novel biomarkers has emerged as a critical need for therapeutic development in amyotrophic lateral sclerosis (ALS). For some subsets of ALS, such as the genetic superoxide dismutase 1 (SOD1) form, exciting new treatment strategies, such as antisense oligonucleotide-mediated (ASO-mediated) SOD1 silencing, are being tested in clinical trials, so the identification of pharmacodynamic biomarkers for therapeutic monitoring is essential. We identify increased levels of a 7-amino acid endogenous peptide of SOD1 in cerebrospinal fluid (CSF) of human SOD1 mutation carriers but not in other neurological cases or nondiseased controls. Levels of peptide elevation vary based on the specific SOD1 mutation (ranging from 1.1-fold greater than control in D90A to nearly 30-fold greater in V148G) and correlate with previously published measurements of SOD1 stability. Using a mass spectrometry-based method (liquid chromatography-mass spectrometry), we quantified peptides in both extracellular samples (CSF) and intracellular samples (spinal cord from rat) to demonstrate that the peptide distinguishes mutation-specific differences in intracellular SOD1 degradation. Furthermore, 80% and 63% reductions of the peptide were measured in SOD1G93A and SOD1H46R rat CSF samples, respectively, following treatment with ASO, with an improved correlation to mRNA levels in spinal cords compared with the ELISA measuring intact SOD1 protein. These data demonstrate the potential of this peptide as a pharmacodynamic biomarker.
Collapse
Affiliation(s)
- Ilya Gertsman
- Biochemical Genetics and Metabolomics Laboratory, Department of Pediatrics, UCSD, La Jolla, California, USA.,Clarus Analytical, LLC, San Diego, California, USA
| | - Joanne Wuu
- Department of Neurology, University of Miami, Miami, Florida, USA
| | | | - Majid Ghassemian
- Biomolecular/Proteomics Mass Spectrometry Facility, Department of Chemistry and Biochemistry, UCSD, La Jolla, California, USA
| | - Karen Ling
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | | | - Michael Benatar
- Department of Neurology, University of Miami, Miami, Florida, USA
| | - Timothy M Miller
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | | |
Collapse
|
298
|
Vijayakumar UG, Milla V, Cynthia Stafford MY, Bjourson AJ, Duddy W, Duguez SMR. A Systematic Review of Suggested Molecular Strata, Biomarkers and Their Tissue Sources in ALS. Front Neurol 2019; 10:400. [PMID: 31139131 PMCID: PMC6527847 DOI: 10.3389/fneur.2019.00400] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/02/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease, is an incurable neurodegenerative condition, characterized by the loss of upper and lower motor neurons. It affects 1-1.8/100,000 individuals worldwide, and the number of cases is projected to increase as the population ages. Thus, there is an urgent need to identify both therapeutic targets and disease-specific biomarkers-biomarkers that would be useful to diagnose and stratify patients into different sub-groups for therapeutic strategies, as well as biomarkers to follow the efficacy of any treatment tested during clinical trials. There is a lack of knowledge about pathogenesis and many hypotheses. Numerous "omics" studies have been conducted on ALS in the past decade to identify a disease-signature in tissues and circulating biomarkers. The first goal of the present review was to group the molecular pathways that have been implicated in monogenic forms of ALS, to enable the description of patient strata corresponding to each pathway grouping. This strategy allowed us to suggest 14 strata, each potentially targetable by different pharmacological strategies. The second goal of this review was to identify diagnostic/prognostic biomarker candidates consistently observed across the literature. For this purpose, we explore previous biomarker-relevant "omics" studies of ALS and summarize their findings, focusing on potential circulating biomarker candidates. We systematically review 118 papers on biomarkers published during the last decade. Several candidate markers were consistently shared across the results of different studies in either cerebrospinal fluid (CSF) or blood (leukocyte or serum/plasma). Although these candidates still need to be validated in a systematic manner, we suggest the use of combinations of biomarkers that would likely reflect the "health status" of different tissues, including motor neuron health (e.g., pNFH and NF-L, cystatin C, Transthyretin), inflammation status (e.g., MCP-1, miR451), muscle health (miR-338-3p, miR-206) and metabolism (homocysteine, glutamate, cholesterol). In light of these studies and because ALS is increasingly perceived as a multi-system disease, the identification of a panel of biomarkers that accurately reflect features of pathology is a priority, not only for diagnostic purposes but also for prognostic or predictive applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Stephanie Marie-Rose Duguez
- Northern Ireland Center for Stratified Medicine, Biomedical Sciences Research Institute, Londonderry, United Kingdom
| |
Collapse
|
299
|
McCauley ME, Baloh RH. Inflammation in ALS/FTD pathogenesis. Acta Neuropathol 2019; 137:715-730. [PMID: 30465257 PMCID: PMC6482122 DOI: 10.1007/s00401-018-1933-9] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are neurodegenerative diseases that overlap in their clinical presentation, pathology and genetics, and likely represent a spectrum of one underlying disease. In ALS/FTD patients, neuroinflammation characterized by innate immune responses of tissue-resident glial cells is uniformly present on end-stage pathology, and human imaging studies and rodent models support that neuroinflammation begins early in disease pathogenesis. Additionally, changes in circulating immune cell populations and cytokines are found in ALS/FTD patients, and there is evidence for an autoinflammatory state. However, despite the prominent role of neuro- and systemic inflammation in ALS/FTD, and experimental evidence in rodents that altering microglial function can mitigate pathology, therapeutic approaches to decrease inflammation have thus far failed to alter disease course in humans. Here, we review the characteristics of inflammation in ALS/FTD in both the nervous and peripheral immune systems. We further discuss evidence for direct influence on immune cell function by mutations in ALS/FTD genes including C9orf72, TBK1 and OPTN, and how this could lead to the altered innate immune system “tone” observed in these patients.
Collapse
|
300
|
Lewitt MS, Boyd GW. The Role of Insulin-Like Growth Factors and Insulin-Like Growth Factor-Binding Proteins in the Nervous System. BIOCHEMISTRY INSIGHTS 2019; 12:1178626419842176. [PMID: 31024217 PMCID: PMC6472167 DOI: 10.1177/1178626419842176] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 01/23/2023]
Abstract
The insulin-like growth factors (IGF-I and IGF-II) and their receptors are widely expressed in nervous tissue from early embryonic life. They also cross the blood brain barriers by active transport, and their regulation as endocrine factors therefore differs from other tissues. In brain, IGFs have paracrine and autocrine actions that are modulated by IGF-binding proteins and interact with other growth factor signalling pathways. The IGF system has roles in nervous system development and maintenance. There is substantial evidence for a specific role for this system in some neurodegenerative diseases, and neuroprotective actions make this system an attractive target for new therapeutic approaches. In developing new therapies, interaction with IGF-binding proteins and other growth factor signalling pathways should be considered. This evidence is reviewed, gaps in knowledge are highlighted, and recommendations are made for future research.
Collapse
Affiliation(s)
- Moira S Lewitt
- School of Health & Life Sciences, University of the West of Scotland, Paisley, UK
| | - Gary W Boyd
- School of Health & Life Sciences, University of the West of Scotland, Paisley, UK
| |
Collapse
|