1
|
Wang H, Zeng R. Aberrant protein aggregation in amyotrophic lateral sclerosis. J Neurol 2024; 271:4826-4851. [PMID: 38869826 DOI: 10.1007/s00415-024-12485-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease. As its pathological mechanisms are not well understood, there are no efficient therapeutics for it at present. While it is highly heterogenous both etiologically and clinically, it has a common salient hallmark, i.e., aberrant protein aggregation (APA). The upstream pathogenesis and the downstream effects of APA in ALS are sophisticated and the investigation of this pathology would be of consequence for understanding ALS. In this paper, the pathomechanism of APA in ALS and the candidate treatment strategies for it are discussed.
Collapse
Affiliation(s)
- Huaixiu Wang
- Department Neurology, Shanxi Provincial Peoples Hospital: Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China.
- Beijing Ai-Si-Kang Medical Technology Co. Ltd., No. 18 11th St Economical & Technological Development Zone, Beijing, 100176, China.
| | - Rong Zeng
- Department Neurology, Shanxi Provincial Peoples Hospital: Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| |
Collapse
|
2
|
Moreno-Martinez L, Macías-Redondo S, Strunk M, Guillén-Antonini MI, Lunetta C, Tarlarini C, Penco S, Calvo AC, Osta R, Schoorlemmer J. New Insights into Endogenous Retrovirus-K Transcripts in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2024; 25:1549. [PMID: 38338823 PMCID: PMC10855536 DOI: 10.3390/ijms25031549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Retroviral reverse transcriptase activity and the increased expression of human endogenous retroviruses (HERVs) are associated with amyotrophic lateral sclerosis (ALS). We were interested in confirming HERVK overexpression in the ALS brain, its use as an accessory diagnostic marker for ALS, and its potential interplay with neuroinflammation. Using qPCR to analyze HERVK expression in peripheral blood mononuclear cells (PBMCs) and in postmortem brain samples from ALS patients, no significant differences were observed between patients and control subjects. By contrast, we report alterations in the expression patterns of specific HERVK copies, especially in the brainstem. Out of 27 HERVK copies sampled, the relative expression of 17 loci was >1.2-fold changed in samples from ALS patients. In particular, the relative expression of two HERVK copies (Chr3-3 and Chr3-5) was significantly different in brainstem samples from ALS patients compared with controls. Further qPCR analysis of inflammation markers in brain samples revealed a significant increase in NLRP3 levels, while TNFA, IL6, and GZMB showed slight decreases. We cannot confirm global HERVK overexpression in ALS, but we can report the ALS-specific overexpression of selected HERVK copies in the ALS brain. Our data are compatible with the requirement for better patient stratification and support the potential importance of particular HERVK copies in ALS.
Collapse
Affiliation(s)
- Laura Moreno-Martinez
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain; (L.M.-M.); (R.O.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Instituto Agroalimentario de Aragón (IA2), University of Zaragoza-CITA, C/Miguel, Servet 177, 50013 Zaragoza, Spain
| | - Sofía Macías-Redondo
- Instituto Aragonés de Ciencias de la Salud (IACS), Centro de Investigación Biomédica de Aragón (CIBA), 50009 Zaragoza, Spain; (S.M.-R.)
| | - Mark Strunk
- Instituto Aragonés de Ciencias de la Salud (IACS), Centro de Investigación Biomédica de Aragón (CIBA), 50009 Zaragoza, Spain; (S.M.-R.)
| | | | - Christian Lunetta
- NEMO (NEuroMuscular Omnicentre) Clinical Center, Fondazione Serena Onlus, 20162 Milan, Italy
- Neurorehabilitation Department of Milano Institute, Istituti Clinici Scientifici Maugeri IRCCS, 20138 Milan, Italy
- Medical Genetics Unit, Department of Laboratory Medicine, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (C.T.)
| | - Claudia Tarlarini
- Medical Genetics Unit, Department of Laboratory Medicine, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (C.T.)
| | - Silvana Penco
- Medical Genetics Unit, Department of Laboratory Medicine, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (C.T.)
| | - Ana Cristina Calvo
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain; (L.M.-M.); (R.O.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Instituto Agroalimentario de Aragón (IA2), University of Zaragoza-CITA, C/Miguel, Servet 177, 50013 Zaragoza, Spain
| | - Rosario Osta
- Laboratory of Genetics and Biochemistry (LAGENBIO), Faculty of Veterinary, University of Zaragoza, Miguel Servet 177, 50013 Zaragoza, Spain; (L.M.-M.); (R.O.)
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Instituto Agroalimentario de Aragón (IA2), University of Zaragoza-CITA, C/Miguel, Servet 177, 50013 Zaragoza, Spain
| | - Jon Schoorlemmer
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), Centro de Investigación Biomédica de Aragón (CIBA), 50009 Zaragoza, Spain; (S.M.-R.)
- ARAID Foundation, 50009 Zaragoza, Spain
| |
Collapse
|
3
|
Ruf WP, Boros M, Freischmidt A, Brenner D, Grozdanov V, de Meirelles J, Meyer T, Grehl T, Petri S, Grosskreutz J, Weyen U, Guenther R, Regensburger M, Hagenacker T, Koch JC, Emmer A, Roediger A, Steinbach R, Wolf J, Weishaupt JH, Lingor P, Deschauer M, Cordts I, Klopstock T, Reilich P, Schoeberl F, Schrank B, Zeller D, Hermann A, Knehr A, Günther K, Dorst J, Schuster J, Siebert R, Ludolph AC, Müller K. Spectrum and frequency of genetic variants in sporadic amyotrophic lateral sclerosis. Brain Commun 2023; 5:fcad152. [PMID: 37223130 PMCID: PMC10202555 DOI: 10.1093/braincomms/fcad152] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/24/2023] [Accepted: 05/05/2023] [Indexed: 05/25/2023] Open
Abstract
Therapy of motoneuron diseases entered a new phase with the use of intrathecal antisense oligonucleotide therapies treating patients with specific gene mutations predominantly in the context of familial amyotrophic lateral sclerosis. With the majority of cases being sporadic, we conducted a cohort study to describe the mutational landscape of sporadic amyotrophic lateral sclerosis. We analysed genetic variants in amyotrophic lateral sclerosis-associated genes to assess and potentially increase the number of patients eligible for gene-specific therapies. We screened 2340 sporadic amyotrophic lateral sclerosis patients from the German Network for motor neuron diseases for variants in 36 amyotrophic lateral sclerosis-associated genes using targeted next-generation sequencing and for the C9orf72 hexanucleotide repeat expansion. The genetic analysis could be completed on 2267 patients. Clinical data included age at onset, disease progression rate and survival. In this study, we found 79 likely pathogenic Class 4 variants and 10 pathogenic Class 5 variants (without the C9orf72 hexanucleotide repeat expansion) according to the American College of Medical Genetics and Genomics guidelines, of which 31 variants are novel. Thus, including C9orf72 hexanucleotide repeat expansion, Class 4, and Class 5 variants, 296 patients, corresponding to ∼13% of our cohort, could be genetically resolved. We detected 437 variants of unknown significance of which 103 are novel. Corroborating the theory of oligogenic causation in amyotrophic lateral sclerosis, we found a co-occurrence of pathogenic variants in 10 patients (0.4%) with 7 being C9orf72 hexanucleotide repeat expansion carriers. In a gene-wise survival analysis, we found a higher hazard ratio of 1.47 (95% confidence interval 1.02-2.1) for death from any cause for patients with the C9orf72 hexanucleotide repeat expansion and a lower hazard ratio of 0.33 (95% confidence interval 0.12-0.9) for patients with pathogenic SOD1 variants than for patients without a causal gene mutation. In summary, the high yield of 296 patients (∼13%) harbouring a pathogenic variant and oncoming gene-specific therapies for SOD1/FUS/C9orf72, which would apply to 227 patients (∼10%) in this cohort, corroborates that genetic testing should be made available to all sporadic amyotrophic lateral sclerosis patients after respective counselling.
Collapse
Affiliation(s)
- Wolfgang P Ruf
- Correspondence to: Dr Wolfgang P. Ruf Department of Neurology Medical Faculty, Ulm University Albert-Einstein-Allee 23, Ulm 89081, Germany E-mail:
| | - Matej Boros
- Institute of Human Genetics, Ulm University & Ulm University Medical Center, Ulm 89081, Germany
| | - Axel Freischmidt
- Department of Neurology, Ulm University, Ulm 89081, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), German Center for Neurodegenerative Diseases, Ulm 89081, Germany
| | - David Brenner
- Department of Neurology, Ulm University, Ulm 89081, Germany
| | | | - Joao de Meirelles
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), German Center for Neurodegenerative Diseases, Ulm 89081, Germany
| | - Thomas Meyer
- Department of Neurology, Center for ALS and other Motor Neuron Disorders, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin 13353, Germany
| | - Torsten Grehl
- Department of Neurology, Alfried Krupp Hospital, Essen 45131, Germany
| | - Susanne Petri
- Department of Neurology, Medizinische Hochschule Hannover, Hannover 30625, Germany
| | | | - Ute Weyen
- Department of Neurology, University Hospital Bochum, Bochum 44789, Germany
| | - Rene Guenther
- Department of Neurology, Technische Universität Dresden, Dresden 01307, Germany
| | - Martin Regensburger
- Department of Neurology, University Hospital Erlangen, Erlangen 91054, Germany
| | - Tim Hagenacker
- Department of Neurology Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, Essen 45147, Germany
| | - Jan C Koch
- Department of Neurology, University Medical Center Goettingen, Goettingen 37075, Germany
| | - Alexander Emmer
- University Clinic and Polyclinic for Neurology, University Hospital Halle, Halle 06120, Germany
| | | | - Robert Steinbach
- Department of Neurology, University Hospital Jena, Jena 07747, Germany
| | - Joachim Wolf
- Department of Neurology, Diako Mannheim, Mannheim 68163, Germany
| | - Jochen H Weishaupt
- Department of Neurology, University Hospital Mannheim, Mannheim 68167, Germany
| | - Paul Lingor
- Department of Neurology, Technical University Munich, Munich 80333, Germany
| | - Marcus Deschauer
- Department of Neurology, Technical University Munich, Munich 80333, Germany
| | - Isabell Cordts
- Department of Neurology, Technical University Munich, Munich 80333, Germany
| | - Thomas Klopstock
- Department of Neurology with Friedrich-Baur-Institute, University Hospital of Ludwig-Maximilians-University, München 80336, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), German Center for Neurodegenerative Diseases, Munich 81377, Germany
| | - Peter Reilich
- Department of Neurology with Friedrich-Baur-Institute, University Hospital of Ludwig-Maximilians-University, München 80336, Germany
| | - Florian Schoeberl
- Department of Neurology with Friedrich-Baur-Institute, University Hospital of Ludwig-Maximilians-University, München 80336, Germany
| | - Berthold Schrank
- Department of Neurology, DKD Helios Clinics, Wiesbaden 65191, Germany
| | - Daniel Zeller
- Department of Neurology, University Hospital Wuerzburg, Wuerzburg 97080, Germany
| | - Andreas Hermann
- Translational Neurodegeneration Section ‘Albrecht Kossel’, University Medical Center Rostock, Rostock 18146, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), German Center for Neurodegenerative Diseases, Rostock/Greifswald 17489, Germany
| | - Antje Knehr
- Department of Neurology, Ulm University, Ulm 89081, Germany
| | | | - Johannes Dorst
- Department of Neurology, Ulm University, Ulm 89081, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), German Center for Neurodegenerative Diseases, Ulm 89081, Germany
| | - Joachim Schuster
- Department of Neurology, Ulm University, Ulm 89081, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), German Center for Neurodegenerative Diseases, Ulm 89081, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University & Ulm University Medical Center, Ulm 89081, Germany
| | - Albert C Ludolph
- Department of Neurology, Ulm University, Ulm 89081, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), German Center for Neurodegenerative Diseases, Ulm 89081, Germany
| | - Kathrin Müller
- Department of Neurology, Ulm University, Ulm 89081, Germany
- Institute of Human Genetics, Ulm University & Ulm University Medical Center, Ulm 89081, Germany
| |
Collapse
|
4
|
Ketabforoush AHME, Chegini R, Barati S, Tahmasebi F, Moghisseh B, Joghataei MT, Faghihi F, Azedi F. Masitinib: The promising actor in the next season of the Amyotrophic Lateral Sclerosis treatment series. Biomed Pharmacother 2023; 160:114378. [PMID: 36774721 DOI: 10.1016/j.biopha.2023.114378] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/25/2023] [Accepted: 02/05/2023] [Indexed: 02/12/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a neurodegenerative disease with high mortality and morbidity rate affecting both upper and lower motor neurons (MN). Muscle force reduction, behavioral change, pseudobulbar affect, and cognitive impairments are the most common clinical manifestations of ALS. The main physiopathology of ALS is still unclear, though several studies have identified that oxidative stress, proteinopathies, glutamate-related excitotoxicity, microglial activation, and neuroinflammation may be involved in the pathogenesis of ALS. From 1995 until October 2022, only Riluzole, Dextromethorphan Hydrobromide (DH) with Quinidine sulfate (Q), Edaravone, and Sodium phenylbutyrate with Taurursodiol (PB/TUDCO) have achieved FDA approval for ALS treatment. Despite the use of these four approved agents, the survival rate and quality of life of ALS patients are still low. Thus, finding novel treatments for ALS patients is an urgent requirement. Masitinib, a tyrosine kinase inhibitor, emphasizes the neuro-inflammatory activity of ALS by targeting macrophages, mast cells, and microglia cells. Masitinib downregulates the proinflammatory cytokines, indirectly reduces inflammation, and induces neuroprotection. Also, it was effective in phase 2/3 and 3 clinical trials (CTs) by increasing overall survival and delaying motor, bulbar, and respiratory function deterioration. This review describes the pathophysiology of ALS, focusing on Masitinib's mechanism of action and explaining why Masitinib could be a promising actor in the treatment of ALS patients. In addition, Masitinib CTs and other competitor drugs in phase 3 CTs have been discussed.
Collapse
Affiliation(s)
| | - Rojin Chegini
- Metabolic Liver Disease Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - Fatemeh Tahmasebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bardia Moghisseh
- Student Research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Fereshteh Azedi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Afridi R, Rahman MH, Suk K. Implications of glial metabolic dysregulation in the pathophysiology of neurodegenerative diseases. Neurobiol Dis 2022; 174:105874. [PMID: 36154877 DOI: 10.1016/j.nbd.2022.105874] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 08/28/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Glial cells are the most abundant cells of the brain, outnumbering neurons. These multifunctional cells are crucial for maintaining brain homeostasis by providing trophic and nutritional support to neurons, sculpting synapses, and providing an immune defense. Glia are highly plastic and undergo both structural and functional alterations in response to changes in the brain microenvironment. Glial phenotypes are intimately regulated by underlying metabolic machinery, which dictates the effector functions of these cells. Altered brain energy metabolism and chronic neuroinflammation are common features of several neurodegenerative diseases. Microglia and astrocytes are the major glial cells fueling the ongoing neuroinflammatory process, exacerbating neurodegeneration. Distinct metabolic perturbations in microglia and astrocytes, including altered carbohydrate, lipid, and amino acid metabolism have been documented in neurodegenerative diseases. These disturbances aggravate the neurodegenerative process by potentiating the inflammatory activation of glial cells. This review covers the recent advances in the molecular aspects of glial metabolic changes in the pathophysiology of neurodegenerative diseases. Finally, we discuss studies exploiting glial metabolism as a potential therapeutic avenue in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu 41944, Republic of Korea.
| |
Collapse
|
6
|
Bazán-Rodríguez L, Ruíz-Avalos JA, Bernal-López O, Benitez-Alonso EO, Villaseñor-Sánchez J, Fernandez-Valverde F, Vargas-Cañas S. FUS as a cause of familial Amyotrophic lateral sclerosis, a case report in a pregnant patient. Neurocase 2022; 28:323-330. [PMID: 35833217 DOI: 10.1080/13554794.2022.2100265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease. In 10% the disease is familial and rarely occurs in childbearing age women. A 28-year-old female pregnancy patient presented a two-month history of dropped head syndrome, dysphagia, muscle weakness, atrophy, and lingual wasting. Electromyography supported the diagnosis of ALS. Due to family history and background, we carried out molecular genetic testing. We identified a novel variant of uncertain significance: c. 1566 G > C (p.Arg522Ser) in exon 15 in FUS gene. Our findings provide the first case of ALS onset during pregnancy with a novel mutation in FUS gene reported in Mexico.
Collapse
Affiliation(s)
- Lisette Bazán-Rodríguez
- Neuromuscular Disorders Clinic, National Institute of Neurology and Neurosurgery Dr. Manuel Velasco Suarez, Mexico City, Mexico
| | - Jesús A Ruíz-Avalos
- Neuromuscular Disorders Clinic, National Institute of Neurology and Neurosurgery Dr. Manuel Velasco Suarez, Mexico City, Mexico
| | - Oscar Bernal-López
- Department of Neurology, Mexican Institute of Social Security, La Raza National Medical Center Specialty Hospital, Mexico City, Mexico
| | - Edmar O Benitez-Alonso
- Department of Neurogenetics, National Institute of Neurology and Neurosurgery Dr. Manuel Velasco Suarez, Mexico City, Mexico
| | - Jesica Villaseñor-Sánchez
- Department of Neurology, Mexican Institute of Social Security, La Raza National Medical Center Specialty Hospital, Mexico City, Mexico
| | - Francisca Fernandez-Valverde
- Neuromuscular Pathology Laboratory, National Institute of Neurology and Neurosurgery Dr. Manuel Velasco Suarez, Mexico City, Mexico
| | - Steven Vargas-Cañas
- Neuromuscular Disorders Clinic, National Institute of Neurology and Neurosurgery Dr. Manuel Velasco Suarez, Mexico City, Mexico
| |
Collapse
|
7
|
Ahangaran M, Chiò A, D'Ovidio F, Manera U, Vasta R, Canosa A, Moglia C, Calvo A, Minaei-Bidgoli B, Jahed-Motlagh MR. Causal associations of genetic factors with clinical progression in amyotrophic lateral sclerosis. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 216:106681. [PMID: 35151113 DOI: 10.1016/j.cmpb.2022.106681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 01/08/2022] [Accepted: 02/02/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND AND OBJECTIVE Recent advances in the genetic causes of ALS reveals that about 10% of ALS patients have a genetic origin and that more than 30 genes are likely to contribute to this disease. However, four genes are more frequently associated with ALS: C9ORF72, TARDBP, SOD1, and FUS. The relationship between genetic factors and ALS progression rate is not clear. In this study, we carried out a causal analysis of ALS disease with a genetics perspective in order to assess the contribution of the four mentioned genes to the progression rate of ALS. METHODS In this work, we applied a novel causal learning model to the CRESLA dataset which is a longitudinal clinical dataset of ALS patients including genetic information of such patients. This study aims to discover the relationship between four mentioned genes and ALS progression rate from a causation perspective using machine learning and probabilistic methods. RESULTS The results indicate a meaningful association between genetic factors and ALS progression rate with causality viewpoint. Our findings revealed that causal relationships between ALSFRS-R items associated with bulbar regions have the strongest association with genetic factors, especially C9ORF72; and other three genes have the greatest contribution to the respiratory ALSFRS-R items with a causation point of view. CONCLUSIONS The findings revealed that genetic factors have a significant causal effect on the rate of ALS progression. Since C9ORF72 patients have higher proportion compared to those carrying other three gene mutations in the CRESLA cohort, we need a large multi-centric study to better analyze SOD1, TARDBP and FUS contribution to the ALS clinical progression. We conclude that causal associations between ALSFRS-R clinical factors is a suitable predictor for designing a prognostic model of ALS.
Collapse
Affiliation(s)
- Meysam Ahangaran
- Department of Computer Engineering, Iran University of Science and Technology, Tehran, Iran; Department of Computer Engineering, Mazandaran University of Science and Technology, Babol, Iran.
| | - Adriano Chiò
- Department of Computer Engineering, Mazandaran University of Science and Technology, Babol, Iran; 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy; Neurology 1, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza of Torino, Turin, Italy; National Research Council, Institute of Cognitive Sciences and Technologies, Rome, Italy.
| | - Fabrizio D'Ovidio
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Umberto Manera
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Rosario Vasta
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy
| | - Antonio Canosa
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy; Neurology 1, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza of Torino, Turin, Italy
| | - Cristina Moglia
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy; Neurology 1, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza of Torino, Turin, Italy
| | - Andrea Calvo
- 'Rita Levi Montalcini' Department of Neuroscience, University of Turin, Turin, Italy; Neurology 1, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza of Torino, Turin, Italy
| | - Behrouz Minaei-Bidgoli
- Department of Computer Engineering, Iran University of Science and Technology, Tehran, Iran
| | | |
Collapse
|
8
|
Zhao X, Feng X, Li X, Mou J, Liu H, Chen J, Wu J. The G41D mutation in SOD1-related amyotrophic lateral sclerosis exhibits phenotypic heterogeneity among individuals: A case report and literature review. Medicine (Baltimore) 2022; 101:e28771. [PMID: 35147103 PMCID: PMC8830837 DOI: 10.1097/md.0000000000028771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/19/2022] [Indexed: 12/11/2022] Open
Abstract
RATIONALE Amyotrophic lateral sclerosis (ALS) is a progressive and fatal neurodegenerative disease. However, the misdiagnosis of ALS always occurs because of atypical clinical manifestations. Since mutations in Cu/Zn superoxide dismutase 1 (SOD1) have been implicated as causative and account for 20% of fALS cases, early genetic sequencing of suspected individuals in ALS pedigrees could be helpful. PATIENT CONCERNS Here we report a Chinese family spanning three generations with fALS. A heterozygous c.125G>A (p.Gly42Asp) missense mutation in exon 2 of SOD1 gene was detected in our proband as well as her 2 siblings and next generation. Phenotypic diversity was also reported among symptomatic individuals. DIAGNOSES Peripheral blood samples from the proband were collected and sent for polymerase chain reaction (PCR) and Sanger sequencing of the SOD1 gene at Sanvalley Diagnostics. The other 11 members in the studied family then underwent locus verification. INTERVENTIONS Butylphthalide, Vitamin B12, Coenzyme Q10 and mouse nerve growth factor is given to the symptomatic members. OUTCOMES The symptoms of our proband was not improved by treatments at a late stage. She passed away the fourth year of the disease due to respiratory failure. Two siblings of the proband were given active treatments once verified as carrier. Their symptoms are still limited to limb weakness. LESSONS This study suggests genetic sequencing is a powerful tool for the diagnosis of familial ALS. Phenotypic heterogeneity exists among G41D-mutated individuals, which further highlights the importance of genomic strategies for early diagnosis.
Collapse
Affiliation(s)
- Xinyi Zhao
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xinhong Feng
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Xiuli Li
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jingyu Mou
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Hongjing Liu
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jing Chen
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jian Wu
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- IDG/McGovern Institute for Brain Research at Tsinghua University, Beijing, China
| |
Collapse
|
9
|
Kannan A, Cuartas J, Gangwani P, Branzei D, Gangwani L. Mutation in senataxin alters the mechanism of R-loop resolution in amyotrophic lateral sclerosis 4. Brain 2022; 145:3072-3094. [PMID: 35045161 PMCID: PMC9536298 DOI: 10.1093/brain/awab464] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/08/2021] [Accepted: 12/03/2021] [Indexed: 11/22/2022] Open
Abstract
Mutation in the senataxin (SETX) gene causes an autosomal dominant neuromuscular disorder, amyotrophic lateral sclerosis 4 (ALS4), characterized by degeneration of motor neurons, muscle weakness and atrophy. SETX is an RNA-DNA helicase that mediates resolution of co-transcriptional RNA:DNA hybrids (R-loops). The process of R-loop resolution is essential for the normal functioning of cells, including neurons. The molecular basis of ALS4 pathogenesis and the mechanism of R-loop resolution are unclear. We report that the zinc finger protein ZPR1 binds to RNA:DNA hybrids, recruits SETX onto R-loops and is critical for R-loop resolution. ZPR1 deficiency disrupts the integrity of R-loop resolution complexes containing SETX and causes increased R-loop accumulation throughout gene transcription. We uncover that SETX is a downstream target of ZPR1 and that overexpression of ZPR1 can rescue R-loop resolution complexe assembly in SETX-deficient cells but not vice versa. To uncover the mechanism of R-loop resolution, we examined the function of SETX-ZPR1 complexes using two genetic motor neuron disease models with altered R-loop resolution. Notably, chronic low levels of SETX-ZPR1 complexes onto R-loops result in a decrease of R-loop resolution activity causing an increase in R-loop levels in spinal muscular atrophy. ZPR1 overexpression increases recruitment of SETX onto R-loops, decreases R-loops and rescues the spinal muscular atrophy phenotype in motor neurons and patient cells. Strikingly, interaction of SETX with ZPR1 is disrupted in ALS4 patients that have heterozygous SETX (L389S) mutation. ZPR1 fails to recruit the mutant SETX homodimer but recruits the heterodimer with partially disrupted interaction between SETX and ZPR1. Interestingly, disruption of SETX-ZPR1 complexes causes increase in R-loop resolution activity leading to fewer R-loops in ALS4. Modulation of ZPR1 levels regulates R-loop accumulation and rescues the pathogenic R-loop phenotype in ALS4 patient cells. These findings originate a new concept, ‘opposite alterations in a cell biological activity (R-loop resolution) result in similar pathogenesis (neurodegeneration) in different genetic motor neuron disorders’. We propose that ZPR1 collaborates with SETX and may function as a molecular brake to regulate SETX-dependent R-loop resolution activity critical for the normal functioning of motor neurons.
Collapse
Affiliation(s)
- Annapoorna Kannan
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905, USA
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905, USA
| | - Juliana Cuartas
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905, USA
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905, USA
| | - Pratik Gangwani
- Automated Driving Compute System Architecture, GM Global Technical Center - Sloan Engineering Center, Warren, Michigan 48092, USA
| | - Dana Branzei
- The FIRC Institute of Molecular Oncology Foundation, IFOM Foundation, Via Adamello 16, Milan 20139, Italy
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche (IGM-CNR), Via Abbiategrasso 207, 27100, Pavia, Italy
| | - Laxman Gangwani
- Center of Emphasis in Neurosciences, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905, USA
- Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas 79905, USA
| |
Collapse
|
10
|
Lu Y, Chen W, Wei C, Zhu Y, Xu R. Potential Common Genetic Risks of Sporadic Parkinson's Disease and Amyotrophic Lateral Sclerosis in the Han Population of Mainland China. Front Neurosci 2021; 15:753870. [PMID: 34707478 PMCID: PMC8542930 DOI: 10.3389/fnins.2021.753870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
Sporadic Parkinson’s disease (sPD) and sporadic amyotrophic lateral sclerosis (sALS) are neurodegenerative diseases characterized by progressive and selective neuron death, with some genetic similarities. In order to investigate the genetic risk factors common to both sPD and sALS, we carried out a screen of risk alleles for sALS and related loci in 530 sPD patients and 530 controls from the Han population of Mainland China (HPMC). We selected 27 single-nucleotide polymorphisms in 10 candidate genes associated with sALS, and we performed allelotyping and genotyping to determine their frequencies in the study population as well as bioinformatics analysis to assess their functional significance in these diseases. The minor alleles of rs17115303 in DAB adaptor protein 1 (DAB1) gene and rs6030462 in protein tyrosine phosphatase receptor type T (PTPRT) gene were correlated with increased risk of both sPD and sALS. Polymorphisms of rs17115303 and rs6030462 were associated with alterations in transcription factor binding sites, secondary structures, long non-coding RNA interactions, and nervous system regulatory networks; these changes involved biological processes associated with neural cell development, differentiation, neurogenesis, migration, axonogenesis, cell adhesion, and metabolism of phosphate-containing compounds. Thus, variants of DAB1 gene (rs17115303) and PTPRT gene (rs6030462) are risk factors common to sPD and sALS in the HPMC. These findings provide insight into the molecular pathogenesis of both diseases and can serve as a basis for the development of targeted therapies.
Collapse
Affiliation(s)
- Yi Lu
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenzhi Chen
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, China
| | - Caihui Wei
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, China
| | - Yu Zhu
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Affiliated People's Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
11
|
Bartoletti-Stella A, Vacchiano V, De Pasqua S, Mengozzi G, De Biase D, Bartolomei I, Avoni P, Rizzo G, Parchi P, Donadio V, Chiò A, Pession A, Oppi F, Salvi F, Liguori R, Capellari S. Targeted sequencing panels in Italian ALS patients support different etiologies in the ALS/FTD continuum. J Neurol 2021; 268:3766-3776. [PMID: 33770234 PMCID: PMC8463338 DOI: 10.1007/s00415-021-10521-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/12/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND 5-10% of amyotrophic lateral sclerosis (ALS) patients presented a positive family history (fALS). More than 30 genes have been identified in association with ALS/frontotemporal dementia (FTD) spectrum, with four major genes accounting for 60-70% of fALS. In this paper, we aimed to assess the contribution to the pathogenesis of major and rare ALS/FTD genes in ALS patients. METHODS We analyzed ALS and ALS/FTD associated genes by direct sequencing or next-generation sequencing multigene panels in ALS patients. RESULTS Genetic abnormalities in ALS major genes included repeated expansions of hexanucleotide in C9orf72 gene (7.3%), mutations in SOD1 (4.9%), FUS (2.1%), and TARDBP (2.4%), whereas variants in rare ALS/FTD genes affected 15.5% of subjects overall, most frequently involving SQSTM1 (3.4%), and CHMP2B (1.9%). We found clustering of variants in ALS major genes in patients with a family history for "pure" ALS, while ALS/FTD related genes mainly occurred in patients with a family history for other neurodegenerative diseases (dementia and/or parkinsonism). CONCLUSIONS Our data support the presence of two different genetic components underlying ALS pathogenesis, related to the presence of a family history for ALS or other neurodegenerative diseases. Thus, family history may help in optimizing the genetic screening protocol to be applied.
Collapse
Affiliation(s)
- Anna Bartoletti-Stella
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
| | - Veria Vacchiano
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
| | - Silvia De Pasqua
- Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), Università Di Bologna, 40123, Bologna, Italy
| | - Giacomo Mengozzi
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
| | - Dario De Biase
- Department of Pharmacy and Biotechnology, Molecular Diagnostic Unit, University of Bologna, viale Ercolani 4/2, 40138, Bologna, Italy
| | - Ilaria Bartolomei
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
| | - Patrizia Avoni
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), Università Di Bologna, 40123, Bologna, Italy
| | - Giovanni Rizzo
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), Università Di Bologna, 40123, Bologna, Italy
| | - Piero Parchi
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138, Bologna, Italy
| | - Vincenzo Donadio
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
| | - Adriano Chiò
- Rita Levi Montalcini Department of Neuroscience, University of Turin, Turin, Italy
- Azienda Ospedaliero Universitaria Citta Della Salute E Della Scienza Di Torino, Turin, Italy
- Neuroscience Institute of Turin, Turin, Italy
| | - Annalisa Pession
- Department of Pharmacy and Biotechnology, Molecular Diagnostic Unit, University of Bologna, viale Ercolani 4/2, 40138, Bologna, Italy
| | - Federico Oppi
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
| | - Fabrizio Salvi
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
| | - Rocco Liguori
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy
- Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), Università Di Bologna, 40123, Bologna, Italy
| | - Sabina Capellari
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bellaria Hospital, 40139, Bologna, Italy.
- Dipartimento di Scienze Biomediche e Neuromotorie (DIBINEM), Università Di Bologna, 40123, Bologna, Italy.
| | | |
Collapse
|
12
|
Bonet LFS, Loureiro JP, Pereira GRC, Da Silva ANR, De Mesquita JF. Molecular dynamics and protein frustration analysis of human fused in Sarcoma protein variants in Amyotrophic Lateral Sclerosis type 6: An In Silico approach. PLoS One 2021; 16:e0258061. [PMID: 34587215 PMCID: PMC8480726 DOI: 10.1371/journal.pone.0258061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/16/2021] [Indexed: 11/18/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most frequent adult-onset motor neuron disorder. The disease is characterized by degeneration of upper and lower motor neurons, leading to death usually within five years after the onset of symptoms. While most cases are sporadic, 5%-10% of cases can be associated with familial inheritance, including ALS type 6, which is associated with mutations in the Fused in Sarcoma (FUS) gene. This work aimed to evaluate how the most frequent ALS-related mutations in FUS, R521C, R521H, and P525L affect the protein structure and function. We used prediction algorithms to analyze the effects of the non-synonymous single nucleotide polymorphisms and performed evolutionary conservation analysis, protein frustration analysis, and molecular dynamics simulations. Most of the prediction algorithms classified the three mutations as deleterious. All three mutations were predicted to reduce protein stability, especially the mutation R521C, which was also predicted to increase chaperone binding tendency. The protein frustration analysis showed an increase in frustration in the interactions involving the mutated residue 521C. Evolutionary conservation analysis showed that residues 521 and 525 of human FUS are highly conserved sites. The molecular dynamics results indicate that protein stability could be compromised in all three mutations. They also affected the exposed surface area and protein compactness. The analyzed mutations also displayed high flexibility in most residues in all variants, most notably in the interaction site with the nuclear import protein of FUS.
Collapse
Affiliation(s)
- L. F. S. Bonet
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J. P. Loureiro
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - G. R. C. Pereira
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - A. N. R. Da Silva
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J. F. De Mesquita
- Department of Genetics and Molecular Biology, Laboratory of Bioinformatics and Computational Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Salucci S, Bartoletti Stella A, Battistelli M, Burattini S, Bavelloni A, Cocco LI, Gobbi P, Faenza I. How Inflammation Pathways Contribute to Cell Death in Neuro-Muscular Disorders. Biomolecules 2021; 11:1109. [PMID: 34439778 PMCID: PMC8391499 DOI: 10.3390/biom11081109] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
Neuro-muscular disorders include a variety of diseases induced by genetic mutations resulting in muscle weakness and waste, swallowing and breathing difficulties. However, muscle alterations and nerve depletions involve specific molecular and cellular mechanisms which lead to the loss of motor-nerve or skeletal-muscle function, often due to an excessive cell death. Morphological and molecular studies demonstrated that a high number of these disorders seem characterized by an upregulated apoptosis which significantly contributes to the pathology. Cell death involvement is the consequence of some cellular processes that occur during diseases, including mitochondrial dysfunction, protein aggregation, free radical generation, excitotoxicity and inflammation. The latter represents an important mediator of disease progression, which, in the central nervous system, is known as neuroinflammation, characterized by reactive microglia and astroglia, as well the infiltration of peripheral monocytes and lymphocytes. Some of the mechanisms underlying inflammation have been linked to reactive oxygen species accumulation, which trigger mitochondrial genomic and respiratory chain instability, autophagy impairment and finally neuron or muscle cell death. This review discusses the main inflammatory pathways contributing to cell death in neuro-muscular disorders by highlighting the main mechanisms, the knowledge of which appears essential in developing therapeutic strategies to prevent the consequent neuron loss and muscle wasting.
Collapse
Affiliation(s)
- Sara Salucci
- Department of Biomolecular Sciences (DiSB), Urbino University Carlo Bo, 61029 Urbino, Italy; (M.B.); (S.B.); (P.G.)
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (L.I.C.); (I.F.)
| | - Anna Bartoletti Stella
- Department of Diagnostic Experimental and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy;
| | - Michela Battistelli
- Department of Biomolecular Sciences (DiSB), Urbino University Carlo Bo, 61029 Urbino, Italy; (M.B.); (S.B.); (P.G.)
| | - Sabrina Burattini
- Department of Biomolecular Sciences (DiSB), Urbino University Carlo Bo, 61029 Urbino, Italy; (M.B.); (S.B.); (P.G.)
| | - Alberto Bavelloni
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Lucio Ildebrando Cocco
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (L.I.C.); (I.F.)
| | - Pietro Gobbi
- Department of Biomolecular Sciences (DiSB), Urbino University Carlo Bo, 61029 Urbino, Italy; (M.B.); (S.B.); (P.G.)
| | - Irene Faenza
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy; (L.I.C.); (I.F.)
| |
Collapse
|
14
|
Lynch EM, Robertson S, FitzGibbons C, Reilly M, Switalski C, Eckardt A, Tey SR, Hayakawa K, Suzuki M. Transcriptome analysis using patient iPSC-derived skeletal myocytes: Bet1L as a new molecule possibly linked to neuromuscular junction degeneration in ALS. Exp Neurol 2021; 345:113815. [PMID: 34310943 DOI: 10.1016/j.expneurol.2021.113815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 06/23/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disease in which patients gradually become paralyzed due to loss of motor function. Many genetically inheritable mutations have been linked to ALS; however, the majority of ALS patients are considered sporadic. Therefore, there is a need for a common therapy that is effective for all ALS patients. Although there is evidence of the disease beginning in the periphery at the neuromuscular junction (NMJ), the specific processes involved in skeletal muscle and at the NMJ are still largely unknown. To study common disease mechanisms in ALS skeletal muscle, we performed RNA sequencing of skeletal myocytes differentiated from induced pluripotent stem cells (iPSCs) derived from familial ALS (with C9ORF72, SOD1, or TARDBP mutations) and sporadic ALS patients. Compared to healthy control lines, the myocytes from all ALS lines showed downregulation of four genes: BET1L, DCX, GPC3, and HNRNPK. We next measured the expression levels of these four genes in hind limb muscle samples from a rat model of familial ALS (SOD1G93A transgenic) and found that only the Bet1L gene, which encodes Bet1 Golgi Vesicular Membrane Trafficking Protein Like, was commonly downregulated. Bet1L protein appeared to be localized to the basal lamina of the NMJ, with decreased expression over time in SOD1G93A transgenic rats. Importantly, the expression levels began to decrease early in the disease process. Our results indicate that loss of Bet1L at the NMJ could be of interest for better understanding ALS disease progression.
Collapse
Affiliation(s)
- Eileen M Lynch
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Claire FitzGibbons
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Megan Reilly
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Colton Switalski
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Adam Eckardt
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Sin-Ruow Tey
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA
| | - Koji Hayakawa
- Department of Toxicology, Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin-Madison, WI, USA; Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, WI, USA.
| |
Collapse
|
15
|
Lin M, Hu X, Chang S, Chang Y, Bian W, Hu R, Wang J, Zhu Q, Qiu J. Advances of Antisense Oligonucleotide Technology in the Treatment of Hereditary Neurodegenerative Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6678422. [PMID: 34211575 PMCID: PMC8211492 DOI: 10.1155/2021/6678422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/13/2021] [Accepted: 05/29/2021] [Indexed: 12/11/2022]
Abstract
Antisense nucleic acids are single-stranded oligonucleotides that have been specially chemically modified, which can bind to RNA expressed by target genes through base complementary pairing and affect protein synthesis at the level of posttranscriptional processing or protein translation. In recent years, the application of antisense nucleic acid technology in the treatment of neuromuscular diseases has made remarkable progress. In 2016, the US FDA approved two antisense nucleic acid drugs for the treatment of Duchenne muscular dystrophy (DMD) and spinal muscular atrophy (SMA), and the development to treat other neurodegenerative diseases has also entered the clinical stage. Therefore, ASO represents a treatment with great potential. The article will summarize ASO therapies in terms of mechanism of action, chemical modification, and administration methods and analyze their role in several common neurodegenerative diseases, such as SMA, DMD, and amyotrophic lateral sclerosis (ALS). This article systematically summarizes the great potential of antisense nucleic acid technology in the treatment of hereditary neurodegenerative diseases.
Collapse
Affiliation(s)
- Mengsi Lin
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Xinyi Hu
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Shiyi Chang
- School of Medicine, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yan Chang
- School of Life Sciences, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Wenjun Bian
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Ruikun Hu
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jing Wang
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Qingwen Zhu
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Jiaying Qiu
- Department of Prenatal Screening and Diagnosis Center, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, Jiangsu Province 226001, China
| |
Collapse
|
16
|
Chua JP, De Calbiac H, Kabashi E, Barmada SJ. Autophagy and ALS: mechanistic insights and therapeutic implications. Autophagy 2021; 18:254-282. [PMID: 34057020 PMCID: PMC8942428 DOI: 10.1080/15548627.2021.1926656] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mechanisms of protein homeostasis are crucial for overseeing the clearance of misfolded and toxic proteins over the lifetime of an organism, thereby ensuring the health of neurons and other cells of the central nervous system. The highly conserved pathway of autophagy is particularly necessary for preventing and counteracting pathogenic insults that may lead to neurodegeneration. In line with this, mutations in genes that encode essential autophagy factors result in impaired autophagy and lead to neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS). However, the mechanistic details underlying the neuroprotective role of autophagy, neuronal resistance to autophagy induction, and the neuron-specific effects of autophagy-impairing mutations remain incompletely defined. Further, the manner and extent to which non-cell autonomous effects of autophagy dysfunction contribute to ALS pathogenesis are not fully understood. Here, we review the current understanding of the interplay between autophagy and ALS pathogenesis by providing an overview of critical steps in the autophagy pathway, with special focus on pivotal factors impaired by ALS-causing mutations, their physiologic effects on autophagy in disease models, and the cell type-specific mechanisms regulating autophagy in non-neuronal cells which, when impaired, can contribute to neurodegeneration. This review thereby provides a framework not only to guide further investigations of neuronal autophagy but also to refine therapeutic strategies for ALS and related neurodegenerative diseases.Abbreviations: ALS: amyotrophic lateral sclerosis; Atg: autophagy-related; CHMP2B: charged multivesicular body protein 2B; DPR: dipeptide repeat; FTD: frontotemporal dementia; iPSC: induced pluripotent stem cell; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; PINK1: PTEN induced kinase 1; RNP: ribonuclear protein; sALS: sporadic ALS; SPHK1: sphingosine kinase 1; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK-binding kinase 1; TFEB: transcription factor EB; ULK: unc-51 like autophagy activating kinase; UPR: unfolded protein response; UPS: ubiquitin-proteasome system; VCP: valosin containing protein.
Collapse
Affiliation(s)
- Jason P Chua
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Hortense De Calbiac
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Edor Kabashi
- Recherche translationnelle sur les maladies neurologiques, Institut Imagine, UMR-1163 INSERM et Université Paris Descartes, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Sami J Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
17
|
Cappella M, Pradat PF, Querin G, Biferi MG. Beyond the Traditional Clinical Trials for Amyotrophic Lateral Sclerosis and The Future Impact of Gene Therapy. J Neuromuscul Dis 2021; 8:25-38. [PMID: 33074186 PMCID: PMC7902976 DOI: 10.3233/jnd-200531] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating and incurable motor neuron (MN) disorder affecting both upper and lower MNs. Despite impressive advances in the understanding of the disease’s pathological mechanism, classical pharmacological clinical trials failed to provide an efficient cure for ALS over the past twenty years. Two different gene therapy approaches were recently approved for the monogenic disease Spinal muscular atrophy, characterized by degeneration of lower MNs. This milestone suggests that gene therapy-based therapeutic solutions could be effective for the treatment of ALS. This review summarizes the possible reasons for the failure of traditional clinical trials for ALS. It provides then a focus on the advent of gene therapy approaches for hereditary forms of ALS. Specifically, it describes clinical use of antisense oligonucleotides in three familial forms of ALS, caused by mutations in SOD1, C9orf72 and FUS genes, respectively.. Clinical and pre-clinical studies based on AAV-mediated gene therapy approaches for both familial and sporadic ALS cases are presented as well. Overall, this overview highlights the potential of gene therapy as a transforming technology that will have a huge impact on treatment perspective for ALS patients and on the design of future clinical trials.
Collapse
Affiliation(s)
- Marisa Cappella
- INSERM, Institute of Myology, Centre of Research in Myology, Sorbonne Université, Paris, France
| | - Pierre-François Pradat
- INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, Sorbonne Université, Paris, France.,APHP, Département de Neurologie, Hôpital Pitié-Salpêtrière, Centre référent SLA, Paris, France.,Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute Ulster University, C-TRIC, Altnagelvin Hospital, Derry/Londonderry, United Kingdom
| | - Giorgia Querin
- INSERM, Institute of Myology, Centre of Research in Myology, Sorbonne Université, Paris, France.,Association Institut de Myologie, Plateforme Essais Cliniques Adultes, Paris, France.,APHP, Service de Neuromyologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Maria Grazia Biferi
- INSERM, Institute of Myology, Centre of Research in Myology, Sorbonne Université, Paris, France
| |
Collapse
|
18
|
A novel method for predicting the progression rate of ALS disease based on automatic generation of probabilistic causal chains. Artif Intell Med 2020; 107:101879. [PMID: 32828438 DOI: 10.1016/j.artmed.2020.101879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/17/2020] [Accepted: 05/12/2020] [Indexed: 01/22/2023]
Abstract
Causal discovery is considered as a major concept in biomedical informatics contributing to diagnosis, therapy, and prognosis of diseases. Probabilistic causality approaches in epidemiology and medicine is a common method for finding relationships between pathogen and disease, environment and disease, and adverse events and drugs. Bayesian Network (BN) is one of the common approaches for probabilistic causality, which is widely used in health-care and biomedical science. Since in many biomedical applications we deal with temporal dataset, the temporal extension of BNs called Dynamic Bayesian network (DBN) is used for such applications. DBNs define probabilistic relationships between parameters in consecutive time points in the form of a graph and have been successfully used in many biomedical applications. In this paper, a novel method was introduced for finding probabilistic causal chains from a temporal dataset with the help of entropy and causal tendency measures. In this method, first, Causal Features Dependency (CFD) matrix is created on the basis of parameters changes in consecutive events of a phenomenon, and then the probabilistic causal graph is constructed from this matrix based on entropy criteria. At the next step, a set of probabilistic causal chains of the corresponding causal graph is constructed by a novel polynomial-time heuristic. Finally, the causal chains are used for predicting the future trend of the phenomenon. The proposed model was applied to the Pooled Resource Open-Access Clinical Trials (PRO-ACT) dataset related to Amyotrophic Lateral Sclerosis (ALS) disease, in order to predict the progression rate of this disease. The results of comparison with Bayesian tree, random forest, support vector regression, linear regression, and multivariate regression show that the proposed algorithm can compete with these methods and in some cases outperforms other algorithms. This study revealed that probabilistic causality is an appropriate approach for predicting the future states of chronic diseases with unknown cause.
Collapse
|
19
|
Ramesh N, Kour S, Anderson EN, Rajasundaram D, Pandey UB. RNA-recognition motif in Matrin-3 mediates neurodegeneration through interaction with hnRNPM. Acta Neuropathol Commun 2020; 8:138. [PMID: 32811564 PMCID: PMC7437177 DOI: 10.1186/s40478-020-01021-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is an adult-onset, fatal neurodegenerative disease characterized by progressive loss of upper and lower motor neurons. While pathogenic mutations in the DNA/RNA-binding protein Matrin-3 (MATR3) are linked to ALS and distal myopathy, the molecular mechanisms underlying MATR3-mediated neuromuscular degeneration remain unclear. Methods We generated Drosophila lines with transgenic insertion of human MATR3 wildtype, disease-associated variants F115C and S85C, and deletion variants in functional domains, ΔRRM1, ΔRRM2, ΔZNF1 and ΔZNF2. We utilized genetic, behavioral and biochemical tools for comprehensive characterization of our models in vivo and in vitro. Additionally, we employed in silico approaches to find transcriptomic targets of MATR3 and hnRNPM from publicly available eCLIP datasets. Results We found that targeted expression of MATR3 in Drosophila muscles or motor neurons shorten lifespan and produces progressive motor defects, muscle degeneration and atrophy. Strikingly, deletion of its RNA-recognition motif (RRM2) mitigates MATR3 toxicity. We identified rump, the Drosophila homolog of human RNA-binding protein hnRNPM, as a modifier of mutant MATR3 toxicity in vivo. Interestingly, hnRNPM physically and functionally interacts with MATR3 in an RNA-dependent manner in mammalian cells. Furthermore, common RNA targets of MATR3 and hnRNPM converge in biological processes important for neuronal health and survival. Conclusions We propose a model of MATR3-mediated neuromuscular degeneration governed by its RNA-binding domains and modulated by interaction with splicing factor hnRNPM. Electronic supplementary material The online version of this article (10.1186/s40478-020-01021-5) contains supplementary material, which is available to authorized users.
Collapse
|
20
|
Ryu SW, Stewart R, Pectol DC, Ender NA, Wimalarathne O, Lee JH, Zanini CP, Harvey A, Huibregtse JM, Mueller P, Paull TT. Proteome-wide identification of HSP70/HSC70 chaperone clients in human cells. PLoS Biol 2020; 18:e3000606. [PMID: 32687490 PMCID: PMC7392334 DOI: 10.1371/journal.pbio.3000606] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 07/30/2020] [Accepted: 06/29/2020] [Indexed: 12/25/2022] Open
Abstract
The 70 kDa heat shock protein (HSP70) family of chaperones are the front line of protection from stress-induced misfolding and aggregation of polypeptides in most organisms and are responsible for promoting the stability, folding, and degradation of clients to maintain cellular protein homeostasis. Here, we demonstrate quantitative identification of HSP70 and 71 kDa heat shock cognate (HSC70) clients using a ubiquitin-mediated proximity tagging strategy and show that, despite their high degree of similarity, these enzymes have largely nonoverlapping specificities. Both proteins show a preference for association with newly synthesized polypeptides, but each responds differently to changes in the stoichiometry of proteins in obligate multi-subunit complexes. In addition, expression of an amyotrophic lateral sclerosis (ALS)-associated superoxide dismutase 1 (SOD1) mutant protein induces changes in HSP70 and HSC70 client association and aggregation toward polypeptides with predicted disorder, indicating that there are global effects from a single misfolded protein that extend to many clients within chaperone networks. Together these findings show that the ubiquitin-activated interaction trap (UBAIT) fusion system can efficiently isolate the complex interactome of HSP chaperone family proteins under normal and stress conditions. Development of a ubiquitin-based system to comprehensively identify substrates of HSP70 enzymes in human cells reveals that constitutive HSC70 and stress-induced HSP70 have different binding preferences and respond dynamically to changes in misfolded protein levels.
Collapse
Affiliation(s)
- Seung W. Ryu
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Rose Stewart
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - D. Chase Pectol
- The Department of Chemistry, Texas A&M University, College Station, Texas, United States of America
| | - Nicolette A. Ender
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Oshadi Wimalarathne
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Ji-Hoon Lee
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Carlos P. Zanini
- Department of Statistics & Data Sciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Antony Harvey
- Thermo Fisher Scientific, Austin, Texas, United States of America
| | - Jon M. Huibregtse
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Peter Mueller
- Department of Statistics & Data Sciences, University of Texas at Austin, Austin, Texas, United States of America
| | - Tanya T. Paull
- The Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
21
|
Yun Y, Ha Y. CRISPR/Cas9-Mediated Gene Correction to Understand ALS. Int J Mol Sci 2020; 21:E3801. [PMID: 32471232 PMCID: PMC7312396 DOI: 10.3390/ijms21113801] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/21/2020] [Accepted: 05/21/2020] [Indexed: 12/24/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease caused by the death of motor neurons in the spinal cord and brainstem. ALS has a diverse genetic origin; at least 20 genes have been shown to be related to ALS. Most familial and sporadic cases of ALS are caused by variants of the SOD1, C9orf72, FUS, and TARDBP genes. Genome editing using clustered regularly interspaced short palindromic repeats/CRISPR-associated system 9 (CRISPR/Cas9) can provide insights into the underlying genetics and pathophysiology of ALS. By correcting common mutations associated with ALS in animal models and patient-derived induced pluripotent stem cells (iPSCs), CRISPR/Cas9 has been used to verify the effects of ALS-associated mutations and observe phenotype differences between patient-derived and gene-corrected iPSCs. This technology has also been used to create mutations to investigate the pathophysiology of ALS. Here, we review recent studies that have used CRISPR/Cas9 to understand the genetic underpinnings of ALS.
Collapse
Affiliation(s)
- Yeomin Yun
- Department of Neurosurgery, Spine and Spinal Cord Institute, College of Medicine, Yonsei University, Seoul 03722, Korea;
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul 03722, Korea
| | - Yoon Ha
- Department of Neurosurgery, Spine and Spinal Cord Institute, College of Medicine, Yonsei University, Seoul 03722, Korea;
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
22
|
Abstract
Objectives: The beneficial effects of many substances have been discovered because of regular dietary consumption. This is also the case with curcumin, whose effects have been known for more than 4,000 years in Eastern countries such as China and India. A curcumin-rich diet has been known to counteract many human diseases, including cancer and diabetes, and has been shown to reduce inflammation. The effect of a curcumin treatment for neurological diseases, such as spinal muscular atrophy; Alzheimer's disease; Parkinson's disease; amyotrophic lateral sclerosis; multiple sclerosis; and others, has only recently been brought to the attention of researchers and the wider population.Methods: In this paper, we summarise the studies on this natural product, from its isolation two centuries ago to its characterisation a century later.Results: We describe its role in the treatment of neurological diseases, including its cellular and common molecular mechanisms, and we report on the clinical trials of curcumin with healthy people and patients.Discussion: Commenting on the different approaches adopted by the efforts made to increase its bioavailability.
Collapse
Affiliation(s)
- Raffaella Adami
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Daniele Bottai
- Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
23
|
Perri ER, Parakh S, Vidal M, Mehta P, Ma Y, Walker AK, Atkin JD. The Cysteine (Cys) Residues Cys-6 and Cys-111 in Mutant Superoxide Dismutase 1 (SOD1) A4V Are Required for Induction of Endoplasmic Reticulum Stress in Amyotrophic Lateral Sclerosis. J Mol Neurosci 2020; 70:1357-1368. [PMID: 32445072 DOI: 10.1007/s12031-020-01551-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the degeneration of motor neurons. Between 12 and 20% of inherited cases and approximately 1-2% of all cases are caused by mutations in the gene encoding dismutase 1 (SOD1). Mutant SOD1 A4V (alanine to valine) induces endoplasmic reticulum (ER) stress, which is increasingly implicated as a pathway to motor neuron degeneration and death in ALS. However, it remains unclear how ER stress is induced by mutant SOD1 A4V. Previous studies have established that it is induced early in pathophysiology and it precedes the formation of mutant SOD1 inclusions. SOD1 contains four cysteine residues, two of which form an intra-subunit disulphide bond involving Cys-57 and Cys-146. The remaining two cysteines, Cys-6 and Cys-111, remain unpaired and have been implicated in mutant SOD1 aggregation. In this study, we examined the relationship between the SOD1 A4V cysteine residues and aggregation, ER stress induction and toxicity. We report here that mutation of Cys-6 and Cys-111 in mutant SOD1 A4V, but not Cys-57 or Cys-146, ameliorates ER stress, inclusion formation and apoptosis in neuronal cell lines. These results imply that protein misfolding, induced by Cys-6 and Cys-111, is required for these pathological events in neuronal cells.
Collapse
Affiliation(s)
- Emma R Perri
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Sonam Parakh
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Marta Vidal
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Prachi Mehta
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Yi Ma
- Department of General Surgery, Monash Health, Melbourne, Victoria, Australia
| | - Adam K Walker
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia.,Neurodegeneration Pathobiology Laboratory, Queensland Brain Institute, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Julie D Atkin
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia. .,Department of General Surgery, Monash Health, Melbourne, Victoria, Australia.
| |
Collapse
|
24
|
Carrasco L, Pisa D, Alonso R. Polymicrobial Infections and Neurodegenerative Diseases. CURRENT CLINICAL MICROBIOLOGY REPORTS 2020. [DOI: 10.1007/s40588-020-00139-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
de Araújo Boleti AP, de Oliveira Flores TM, Moreno SE, Anjos LD, Mortari MR, Migliolo L. Neuroinflammation: An overview of neurodegenerative and metabolic diseases and of biotechnological studies. Neurochem Int 2020; 136:104714. [PMID: 32165170 DOI: 10.1016/j.neuint.2020.104714] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/19/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
Neuroinflammation is an important factor contributing to cognitive impairment and neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), ischemic injury, and multiple sclerosis (MS). These diseases are characterized by inexorable progressive injury of neuron cells, and loss of motor or cognitive functions. Microglia, which are the resident macrophages in the brain, play an important role in both physiological and pathological conditions. In this review, we provide an updated discussion on the role of ROS and metabolic disease in the pathological mechanisms of activation of the microglial cells and release of cytotoxins, leading to the neurodegenerative process. In addition, we also discuss in vivo models, such as zebrafish and Caenorhabditis elegans, and provide new insights into therapeutics bioinspired by neuropeptides from venomous animals, supporting high throughput drug screening in the near future, searching for a complementary approach to elucidating crucial mechanisms associated with neurodegenerative disorders.
Collapse
Affiliation(s)
- Ana Paula de Araújo Boleti
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil
| | - Taylla Michelle de Oliveira Flores
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil; Programa de Pós-graduação em Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Susana Elisa Moreno
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil
| | - Lilian Dos Anjos
- Laboratório de Neurofarmacologia, Departmento Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brazil
| | - Márcia Renata Mortari
- Laboratório de Neurofarmacologia, Departmento Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade de Brasília, Brazil
| | - Ludovico Migliolo
- S-InovaBiotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, 79117-900, Campo Grande, MS, Brazil; Programa de Pós-graduação em Biologia Celular e Molecular, Universidade Federal da Paraíba, João Pessoa, Brazil; Programa de Pós-graduação em Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, Brazil.
| |
Collapse
|
26
|
Gregory JM, McDade K, Livesey MR, Croy I, Marion de Proce S, Aitman T, Chandran S, Smith C. Spatial transcriptomics identifies spatially dysregulated expression of GRM3 and USP47 in amyotrophic lateral sclerosis. Neuropathol Appl Neurobiol 2020; 46:441-457. [PMID: 31925813 DOI: 10.1111/nan.12597] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/21/2019] [Indexed: 02/02/2023]
Abstract
AIMS The mechanisms underlying the selective degeneration of motor neurones in amyotrophic lateral sclerosis (ALS) are poorly understood. The aim of this study was to implement spatially resolved RNA sequencing in human post mortem cortical tissue from an ALS patient harbouring the C9orf72 hexanucleotide repeat expansion to identify dysregulated transcripts that may account for differential vulnerabilities of distinct (i) cell types and (ii) brain regions in the pathogenesis of ALS. METHODS Using spatial transcriptomics (ST) we analysed the transcriptome of post mortem brain tissue, with spatial resolution down to 100 μm. Validation of these findings was then performed using BaseScope, an adapted, in situ hybridization technique with single-transcript single-cell-resolution, providing extensive regional and cell-type specific confirmation of these dysregulated transcripts. The validation cohort was then extended to include multiple post mortem brain regions and spinal cord tissue from an extended cohort of C9orf72, sporadic ALS (sALS) and SOD1 ALS cases. RESULTS We identified sixteen dysregulated transcripts of proteins that have roles within six disease-related pathways. Furthermore, these complementary molecular pathology techniques converged to identify two spatially dysregulated transcripts, GRM3 and USP47, that are commonly dysregulated across sALS, SOD1 and C9orf72 cases alike. CONCLUSIONS This study presents the first description of ST in human post mortem cortical tissue from an ALS patient harbouring the C9orf72 hexanucleotide repeat expansion. These data taken together highlight the importance of preserving spatial resolution, facilitating the identification of genes whose dysregulation may in part underlie regional susceptibilities to ALS, crucially highlighting potential therapeutic and diagnostic targets.
Collapse
Affiliation(s)
- J M Gregory
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Edinburgh Pathology, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - K McDade
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Edinburgh Pathology, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - M R Livesey
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - I Croy
- Edinburgh Pathology, University of Edinburgh, Edinburgh, UK.,Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - S Marion de Proce
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - T Aitman
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - S Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - C Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,Edinburgh Pathology, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
27
|
Corcia P, Lumbroso S, Cazeneuve C, Mouzat K, Camu W, Vourc'h P. Pre-symptomatic diagnosis in ALS. Rev Neurol (Paris) 2020; 176:166-169. [PMID: 31932031 DOI: 10.1016/j.neurol.2019.07.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022]
Abstract
Pathophysiology of amyotrophic lateral sclerosis (ALS) remains partially understood even though it is accepted worldwide that motor neuron death results from a pluri-factorial process with a variable role of genetic factors. Although not distinguishable from a clinical point of view, familial forms of ALS (fALS, 10% of cases) and sporadic forms (sALS, 90% of cases) can be described. Since the identification of superoxide dismutase 1 gene (SOD1) mutations, more than 30 genes have been linked to fALS. Among these genes, five (C9ORF72, SOD1, TARDBP, FUS, TBK1) seem predominant with mutation frequencies of 40%, 20%, 5%, <5%, <5% in fALS and 6%, 3%, and <1% for the last three in sALS, respectively. The situation that classically leads to request genetic screening is the presence of a familial history of motor neuron disorders (MND) or fronto-temporal lobar dementia (FTLD). However, this dichotomy between fALS and sALS based on familial history can lead to mistakes since illegitimacy, ignorance of MND, FTD or psychiatric disorders within the family due to a familial censorship or lack of familial relationship, or a recessive autosomal inheritance could wrongly lead to failing to recognize a familial form. The significant development of genetic research and easier access to genetic tests in fALS increase the number of situations for which gene mutations are identified. The consequence is an increase in genetic requests from relatives of ALS patients who are eager to know their own genetic status and their own individual risk to develop ALS. Pre-symptomatic testing is thus becoming a daily issue in ALS Centers. This led us to propose a framework for such pre-symptomatic genetic testing for people at risk for developing ALS.
Collapse
Affiliation(s)
- P Corcia
- Centre Constitutif SLA, CHU Bretonneau, Tours, France; UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
| | - S Lumbroso
- Motoneuron Disease: Pathophysiology and Therapy, INM, University Montpellier, Montpellier, France; Laboratoire de Biochimie et Biologie Moléculaire, CHU Nîmes, Nîmes, France
| | - C Cazeneuve
- Unité Fonctionnelle de Neurogénétique Moléculaire et Cellulaire, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, France
| | - K Mouzat
- Motoneuron Disease: Pathophysiology and Therapy, INM, University Montpellier, Montpellier, France; Laboratoire de Biochimie et Biologie Moléculaire, CHU Nîmes, Nîmes, France
| | - W Camu
- Centre Consitutif SLA, Hopital Guy de Chauliac, Montpellier, France
| | - P Vourc'h
- Motoneuron Disease: Pathophysiology and Therapy, INM, University Montpellier, Montpellier, France; Laboratoire de Biochimie et Biologie Moléculaire, CHU Nîmes, Nîmes, France; Service de Biochimie et Biologie Moléculaire, CHRU Tours, France
| | | |
Collapse
|
28
|
Fernandopulle M, Wang G, Nixon-Abell J, Qamar S, Balaji V, Morihara R, St George-Hyslop PH. Inherited and Sporadic Amyotrophic Lateral Sclerosis and Fronto-Temporal Lobar Degenerations arising from Pathological Condensates of Phase Separating Proteins. Hum Mol Genet 2019; 28:R187-R196. [PMID: 31595953 PMCID: PMC6872449 DOI: 10.1093/hmg/ddz162] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022] Open
Abstract
Recent work on the biophysics of proteins with low complexity, intrinsically disordered domains that have the capacity to form biological condensates has profoundly altered the concepts about the pathogenesis of inherited and sporadic neurodegenerative disorders associated with pathological accumulation of these proteins. In the present review, we use the FUS, TDP-43 and A11 proteins as examples to illustrate how missense mutations and aberrant post-translational modifications of these proteins cause amyotrophic lateral sclerosis (ALS) and fronto-temporal lobar degeneration (FTLD).
Collapse
Affiliation(s)
- Michael Fernandopulle
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - GuoZhen Wang
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Jonathon Nixon-Abell
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Seema Qamar
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
| | - Varun Balaji
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| | - Ryuta Morihara
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| | - Peter H St George-Hyslop
- Cambridge Institute for Medical Research, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK, CB2 0XY
- Tanz Centre for Research in Neurodegenerative Diseases, and Departments of Medicine, University of Toronto, Toronto, Ontario, Canada, M5S 3H2
| |
Collapse
|
29
|
Lynch E, Semrad T, Belsito VS, FitzGibbons C, Reilly M, Hayakawa K, Suzuki M. C9ORF72-related cellular pathology in skeletal myocytes derived from ALS-patient induced pluripotent stem cells. Dis Model Mech 2019; 12:12/8/dmm039552. [PMID: 31439573 PMCID: PMC6737948 DOI: 10.1242/dmm.039552] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/10/2019] [Indexed: 12/25/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a late-onset neuromuscular disease with no cure and limited treatment options. Patients experience a gradual paralysis leading to death from respiratory complications on average only 2-5 years after diagnosis. There is increasing evidence that skeletal muscle is affected early in the disease process, yet the pathological processes occurring in the skeletal muscle of ALS patients are still mostly unknown. Specifically, the most common genetic cause of ALS, a hexanucleotide repeat expansion in the C9ORF72 gene, has yet to be fully characterized in the context of skeletal muscle. In this study, we used the protocol previously developed in our lab to differentiate skeletal myocytes from induced pluripotent stem cells (iPSCs) of C9ORF72 ALS (C9-ALS) patients in order to create an in vitro disease model of C9-ALS skeletal muscle pathology. Of the three C9ORF72 mutation hallmarks, we did not see any evidence of haploinsufficiency, but we did detect RNA foci and dipeptide repeat (DPR) proteins. Additional abnormalities included changes in the expression of mitochondrial genes and a susceptibility to oxidative stress, indicating that mitochondrial dysfunction may be a critical feature of C9-ALS skeletal muscle pathology. Finally, the C9-ALS myocytes had increased expression and aggregation of TDP-43. Together, these data show that skeletal muscle cells experience pathological changes due to the C9ORF72 mutation. Our in vitro model could facilitate further study of cellular and molecular pathology in ALS skeletal muscle in order to discover new therapeutic targets against this devastating disease. This article has an associated First Person interview with the first author of the paper. Summary: Evidence of protein aggregation and mitochondrial dysfunction were found in skeletal myocytes differentiated from ALS-patient induced pluripotent stem cells with the C9ORF72 mutation.
Collapse
Affiliation(s)
- Eileen Lynch
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Theran Semrad
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Vincent S Belsito
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Claire FitzGibbons
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Megan Reilly
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Koji Hayakawa
- Department of Toxicology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA .,Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
30
|
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: 1.7] [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
|
31
|
Franz CK, Joshi D, Daley EL, Grant RA, Dalamagkas K, Leung A, Finan JD, Kiskinis E. Impact of traumatic brain injury on amyotrophic lateral sclerosis: from bedside to bench. J Neurophysiol 2019; 122:1174-1185. [PMID: 31116639 DOI: 10.1152/jn.00572.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the loss of upper and lower motor neurons, which manifests clinically as progressive weakness. Although several epidemiological studies have found an association between traumatic brain injury (TBI) and ALS, there is not a consensus on whether TBI is an ALS risk factor. It may be that it can cause ALS in a subset of susceptible patients, based on a history of repetitive mild TBI and genetic predisposition. This cannot be determined based on clinical observational studies alone. Better preclinical models are necessary to evaluate the effects of TBI on ALS onset and progression. To date, only a small number of preclinical studies have been performed, mainly in the superoxide dismutase 1 transgenic rodents, which, taken together, have mixed results and notable methodological limitations. The more recent incorporation of additional animal models such as Drosophila flies, as well as patient-induced pluripotent stem cell-derived neurons, should facilitate a better understanding of a potential functional interaction between TBI and ALS.
Collapse
Affiliation(s)
- Colin K Franz
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, Illinois.,Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Divya Joshi
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, Illinois
| | - Elizabeth L Daley
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Rogan A Grant
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kyriakos Dalamagkas
- Department of Physical Medicine and Rehabilitation, McGovern Medical School, TIRR Memorial Hermann, Houston, Texas
| | - Audrey Leung
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, Illinois.,Department of Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - John D Finan
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, Illinois
| | - Evangelos Kiskinis
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
32
|
Huai J, Zhang Z. Structural Properties and Interaction Partners of Familial ALS-Associated SOD1 Mutants. Front Neurol 2019; 10:527. [PMID: 31164862 PMCID: PMC6536575 DOI: 10.3389/fneur.2019.00527] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/02/2019] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common motor neuron degenerative disease in adults and has also been proven to be a type of conformational disease associated with protein misfolding and dysfunction. To date, more than 150 distinct genes have been found to be associated with ALS, among which Superoxide Dismutase 1 (SOD1) is the first and the most extensively studied gene. It has been well-established that SOD1 mutants-mediated toxicity is caused by a gain-of-function rather than the loss of the detoxifying activity of SOD1. Compared with the clear autosomal dominant inheritance of SOD1 mutants in ALS, the potential toxic mechanisms of SOD1 mutants in motor neurons remain incompletely understood. A large body of evidence has shown that SOD1 mutants may adopt a complex profile of conformations and interact with a wide range of client proteins. Here, in this review, we summarize the fundamental conformational properties and the gained interaction partners of the soluble forms of the SOD1 mutants which have been published in the past decades. Our goal is to find clues to the possible internal links between structural and functional anomalies of SOD1 mutants, as well as the relationships between their exposed epitopes and interaction partners, in order to help reveal and determine potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Jisen Huai
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Zhongjian Zhang
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
33
|
Parakh S, Perri ER, Jagaraj CJ, Ragagnin AMG, Atkin JD. Rab-dependent cellular trafficking and amyotrophic lateral sclerosis. Crit Rev Biochem Mol Biol 2019; 53:623-651. [PMID: 30741580 DOI: 10.1080/10409238.2018.1553926] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Rab GTPases are becoming increasingly implicated in neurodegenerative disorders, although their role in amyotrophic lateral sclerosis (ALS) has been somewhat overlooked. However, dysfunction of intracellular transport is gaining increasing attention as a pathogenic mechanism in ALS. Many previous studies have focused axonal trafficking, and the extreme length of axons in motor neurons may contribute to their unique susceptibility in this disorder. In contrast, the role of transport defects within the cell body has been relatively neglected. Similarly, whilst Rab GTPases control all intracellular membrane trafficking events, their role in ALS is poorly understood. Emerging evidence now highlights this family of proteins in ALS, particularly the discovery that C9orf72 functions in intra transport in conjunction with several Rab GTPases. Here, we summarize recent updates on cellular transport defects in ALS, with a focus on Rab GTPases and how their dysfunction may specifically target neurons and contribute to pathophysiology. We discuss the molecular mechanisms associated with dysfunction of Rab proteins in ALS. Finally, we also discuss dysfunction in other modes of transport recently implicated in ALS, including nucleocytoplasmic transport and the ER-mitochondrial contact regions (MAM compartment), and speculate whether these may also involve Rab GTPases.
Collapse
Affiliation(s)
- S Parakh
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia.,b Department of Biochemistry and Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia
| | - E R Perri
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia.,b Department of Biochemistry and Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia
| | - C J Jagaraj
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia
| | - A M G Ragagnin
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia
| | - J D Atkin
- a Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Centre for MND Research , Macquarie University , Sydney , Australia.,b Department of Biochemistry and Genetics , La Trobe Institute for Molecular Science, La Trobe University , Melbourne , Australia
| |
Collapse
|
34
|
Alonso R, Pisa D, Carrasco L. Searching for Bacteria in Neural Tissue From Amyotrophic Lateral Sclerosis. Front Neurosci 2019; 13:171. [PMID: 30863279 PMCID: PMC6399391 DOI: 10.3389/fnins.2019.00171] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/13/2019] [Indexed: 12/28/2022] Open
Abstract
Despite great efforts in the investigation, the exact etiology of amyotrophic lateral sclerosis (ALS) is a matter of intensive research. We recently advanced the idea that ALS might be caused by fungal infection. Indeed, fungal yeast and hyphal structures can be directly visualized in neural tissue of ALS patients, and a number of fungal species have been identified in the central nervous system (CNS). In the present work, we tested the possibility that bacterial infections can accompany these mycoses. Our findings establish the presence of bacterial DNA in different regions of the CNS from all ALS patients examined. Specifically, we used PCR and next generation sequencing (NGS) to precisely determine the bacterial species present in ALS tissue. Consistent with these findings, immunohistochemistry analysis of CNS sections using specific anti-bacterial antibodies identified prokaryotic cells in neural tissue. Finally, we assayed for the repeat expansion of the hexanucleotide repeat GGGGCC in C9orf72, which is considered the most common genetic cause of ALS in patients, using DNA extracted from ALS CNS tissue. We failed to find this repeated sequence in any of the eleven patients analyzed. Our results indicate that bacterial DNA and prokaryotic cells are present in CNS tissue, leading to the concept that both fungal and bacterial infections coexist in patients with ALS. These observations lay the groundwork for the use of appropriate therapies to eradicate the polymicrobial infections in ALS.
Collapse
Affiliation(s)
- Ruth Alonso
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Diana Pisa
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Carrasco
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
35
|
Tryptophan 32 mediates SOD1 toxicity in a in vivo motor neuron model of ALS and is a promising target for small molecule therapeutics. Neurobiol Dis 2018; 124:297-310. [PMID: 30528257 DOI: 10.1016/j.nbd.2018.11.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 11/11/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022] Open
Abstract
SOD1 misfolding, toxic gain of function, and spread are proposed as a pathological basis of amyotrophic lateral sclerosis (ALS), but the nature of SOD1 toxicity has been difficult to elucidate. Uniquely in SOD1 proteins from humans and other primates, and rarely in other species, a tryptophan residue at position 32 (W32) is predicted to be solvent exposed and to participate in SOD1 misfolding. We hypothesized that W32 is influential in SOD1 acquiring toxicity, as it is known to be important in template-directed misfolding. We tested if W32 contributes to SOD1 cytotoxicity and if it is an appropriate drug target to ameliorate ALS-like neuromuscular deficits in a zebrafish model of motor neuron axon morphology and function (swimming). Embryos injected with human SOD1 variant with W32 substituted for a serine (SOD1W32S) had reduced motor neuron axonopathy and motor deficits compared to those injected with wildtype or disease-associated SOD1. A library of FDA-approved small molecules was ranked with virtual screening based on predicted binding to W32, and subsequently filtered for analogues using a pharmacophore model based on molecular features of the uracil moiety of a small molecule previously predicted to interact with W32 (5'-fluorouridine or 5'-FUrd). Along with testing 5'-FUrd and uridine, a lead candidate from this list was selected based on its lower toxicity and improved blood brain barrier penetrance; telbivudine significantly rescued SOD1 toxicity in a dose-dependent manner. The mechanisms whereby the small molecules ameliorated motor neuron phenotypes were specifically mediated through human SOD1 and its residue W32, because these therapeutics had no measurable impact on the effects of UBQLN4D90A, EtOH, or tryptophan-deficient human SOD1W32S. By substituting W32 for a more evolutionarily conserved residue (serine), we confirmed the significant influence of W32 on human SOD1 toxicity to motor neuron morphology and function; further, we performed pharmaceutical targeting of the W32 residue for rescuing SOD1 toxicity. This unique residue offers future novel insights into SOD1 stability and toxic gain of function, and therefore poses an potential target for drug therapy.
Collapse
|
36
|
Gromicho M, Pinto S, Gisca E, Pronto-Laborinho AC, Andersen PM, de Carvalho M. Frequency of C9orf72 hexanucleotide repeat expansion and SOD1 mutations in Portuguese patients with amyotrophic lateral sclerosis. Neurobiol Aging 2018; 70:325.e7-325.e15. [DOI: 10.1016/j.neurobiolaging.2018.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 05/03/2018] [Accepted: 05/06/2018] [Indexed: 12/11/2022]
|
37
|
Senataxin mutations elicit motor neuron degeneration phenotypes and yield TDP-43 mislocalization in ALS4 mice and human patients. Acta Neuropathol 2018; 136:425-443. [PMID: 29725819 DOI: 10.1007/s00401-018-1852-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis type 4 (ALS4) is a rare, early-onset, autosomal dominant form of ALS, characterized by slow disease progression and sparing of respiratory musculature. Dominant, gain-of-function mutations in the senataxin gene (SETX) cause ALS4, but the mechanistic basis for motor neuron toxicity is unknown. SETX is a RNA-binding protein with a highly conserved helicase domain, but does not possess a low-complexity domain, making it unique among ALS-linked disease proteins. We derived ALS4 mouse models by expressing two different senataxin gene mutations (R2136H and L389S) via transgenesis and knock-in gene targeting. Both approaches yielded SETX mutant mice that develop neuromuscular phenotypes and motor neuron degeneration. Neuropathological characterization of SETX mice revealed nuclear clearing of TDP-43, accompanied by TDP-43 cytosolic mislocalization, consistent with the hallmark pathology observed in human ALS patients. Postmortem material from ALS4 patients exhibited TDP-43 mislocalization in spinal cord motor neurons, and motor neurons from SETX ALS4 mice displayed enhanced stress granule formation. Immunostaining analysis for nucleocytoplasmic transport proteins Ran and RanGAP1 uncovered nuclear membrane abnormalities in the motor neurons of SETX ALS4 mice, and nuclear import was delayed in SETX ALS4 cortical neurons, indicative of impaired nucleocytoplasmic trafficking. SETX ALS4 mice thus recapitulated ALS disease phenotypes in association with TDP-43 mislocalization and provided insight into the basis for TDP-43 histopathology, linking SETX dysfunction to common pathways of ALS motor neuron degeneration.
Collapse
|
38
|
Müller K, Brenner D, Weydt P, Meyer T, Grehl T, Petri S, Grosskreutz J, Schuster J, Volk AE, Borck G, Kubisch C, Klopstock T, Zeller D, Jablonka S, Sendtner M, Klebe S, Knehr A, Günther K, Weis J, Claeys KG, Schrank B, Sperfeld AD, Hübers A, Otto M, Dorst J, Meitinger T, Strom TM, Andersen PM, Ludolph AC, Weishaupt JH. Comprehensive analysis of the mutation spectrum in 301 German ALS families. J Neurol Neurosurg Psychiatry 2018; 89:817-827. [PMID: 29650794 DOI: 10.1136/jnnp-2017-317611] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/25/2018] [Accepted: 03/07/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Recent advances in amyotrophic lateral sclerosis (ALS) genetics have revealed that mutations in any of more than 25 genes can cause ALS, mostly as an autosomal-dominant Mendelian trait. Detailed knowledge about the genetic architecture of ALS in a specific population will be important for genetic counselling but also for genotype-specific therapeutic interventions. METHODS Here we combined fragment length analysis, repeat-primed PCR, Southern blotting, Sanger sequencing and whole exome sequencing to obtain a comprehensive profile of genetic variants in ALS disease genes in 301 German pedigrees with familial ALS. We report C9orf72 mutations as well as variants in consensus splice sites and non-synonymous variants in protein-coding regions of ALS genes. We furthermore estimate their pathogenicity by taking into account type and frequency of the respective variant as well as segregation within the families. RESULTS 49% of our German ALS families carried a likely pathogenic variant in at least one of the earlier identified ALS genes. In 45% of the ALS families, likely pathogenic variants were detected in C9orf72, SOD1, FUS, TARDBP or TBK1, whereas the relative contribution of the other ALS genes in this familial ALS cohort was 4%. We identified several previously unreported rare variants and demonstrated the absence of likely pathogenic variants in some of the recently described ALS disease genes. CONCLUSIONS We here present a comprehensive genetic characterisation of German familial ALS. The present findings are of importance for genetic counselling in clinical practice, for molecular research and for the design of diagnostic gene panels or genotype-specific therapeutic interventions in Europe.
Collapse
Affiliation(s)
| | - David Brenner
- Department of Neurology, Ulm University, Ulm, Germany
| | - Patrick Weydt
- Department of Neurology, Ulm University, Ulm, Germany.,Department of Neurodegenerative Diseases and Gerontopsychiatry, Bonn University, Bonn, Germany
| | - Thomas Meyer
- Department of Neurology, Charité Hospital, Humboldt University, Berlin, Germany
| | - Torsten Grehl
- Department of Neurology, Alfried Krupp Hospital, Essen, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | | | - Alexander E Volk
- Institute of Human Genetics, Ulm University, Ulm, Germany.,Institute of Human Genetics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Guntram Borck
- Institute of Human Genetics, Ulm University, Ulm, Germany
| | - Christian Kubisch
- Institute of Human Genetics, Ulm University, Ulm, Germany.,Institute of Human Genetics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institut, University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Daniel Zeller
- Department of Neurology, University of Würzburg, Würzburg, Germany
| | - Sibylle Jablonka
- Institute of Clinical Neurobiology, University Hospital of Würzburg, Würzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital of Würzburg, Würzburg, Germany
| | - Stephan Klebe
- Department of Neurology, University of Würzburg, Würzburg, Germany.,Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Antje Knehr
- Department of Neurology, Ulm University, Ulm, Germany
| | | | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Kristl G Claeys
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Neurology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium.,Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, Experimental Neurology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Berthold Schrank
- Department of Neurology, DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany
| | - Anne-Dorte Sperfeld
- Department of Neurology, Martin Luther University of Halle-Wittenberg, Halle/Saale, Germany
| | | | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Germany
| | | | - Thomas Meitinger
- Institute of Human Genetics, Technische Universität München, Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Munich Cluster for Systems Neurology (SNergy), Munich, Germany
| | - Tim M Strom
- Institute of Human Genetics, Technische Universität München, Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany.,Munich Cluster for Systems Neurology (SNergy), Munich, Germany
| | - Peter M Andersen
- Department of Neurology, Ulm University, Ulm, Germany.,Department of Pharmacology and Clinical Neuroscience, Umeå University, Umeå, Sweden
| | | | | | | |
Collapse
|
39
|
Nakamura S, Shimazawa M, Hara H. Physiological Roles of Metallothioneins in Central Nervous System Diseases. Biol Pharm Bull 2018; 41:1006-1013. [DOI: 10.1248/bpb.b17-00856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University
| |
Collapse
|
40
|
Lamp M, Origone P, Geroldi A, Verdiani S, Gotta F, Caponnetto C, Devigili G, Verriello L, Scialò C, Cabona C, Canosa A, Vanni I, Bellone E, Eleopra R, Mandich P. Twenty years of molecular analyses in amyotrophic lateral sclerosis: genetic landscape of Italian patients. Neurobiol Aging 2018. [DOI: 10.1016/j.neurobiolaging.2018.01.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Differential expression of microRNAs and other small RNAs in muscle tissue of patients with ALS and healthy age-matched controls. Sci Rep 2018; 8:5609. [PMID: 29618798 PMCID: PMC5884852 DOI: 10.1038/s41598-018-23139-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/05/2018] [Indexed: 02/08/2023] Open
Abstract
Amyotrophic lateral sclerosis is a late-onset disorder primarily affecting motor neurons and leading to progressive and lethal skeletal muscle atrophy. Small RNAs, including microRNAs (miRNAs), can serve as important regulators of gene expression and can act both globally and in a tissue-/cell-type-specific manner. In muscle, miRNAs called myomiRs govern important processes and are deregulated in various disorders. Several myomiRs have shown promise for therapeutic use in cellular and animal models of ALS; however, the exact miRNA species differentially expressed in muscle tissue of ALS patients remain unknown. Following small RNA-Seq, we compared the expression of small RNAs in muscle tissue of ALS patients and healthy age-matched controls. The identified snoRNAs, mtRNAs and other small RNAs provide possible molecular links between insulin signaling and ALS. Furthermore, the identified miRNAs are predicted to target proteins that are involved in both normal processes and various muscle disorders and indicate muscle tissue is undergoing active reinnervation/compensatory attempts thus providing targets for further research and therapy development in ALS.
Collapse
|
42
|
Sun YM, Dong Y, Wang J, Lu JH, Chen Y, Wu JJ. A novel mutation of VAPB in one Chinese familial amyotrophic lateral sclerosis pedigree and its clinical characteristics. J Neurol 2017; 264:2387-2393. [PMID: 28993872 DOI: 10.1007/s00415-017-8628-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022]
Abstract
The mutation of vesicle-associated membrane protein-associated protein B (VAPB) was proved to cause family amyotrophic lateral sclerosis (FALS). Only two mutations of VAPB associated with ALS have been reported (p.Pro56Ser and p.Thr46Ile). Here we reported a Chinese Han FALS family caused by a novel VAPB point mutation. The clinical materials of one Chinese Han FALS family were collected. The genetic analysis was carried out by target sequencing and further verified by Sanger sequencing. One novel mutation of c.167C>A (p.Pro56His) on VAPB was found in the proband. The age at onset of the proband was 48 with the onset symptoms of weakness in the right arm, followed by progressive limb and trunk weakness with decreased deep-tendon reflexes, muscular cramps and fasciculation. But the disease duration was more than 15 years. He was under the tracheotomy for 1 year at last visit. Electromyography showed widespread acute and chronic neurogenic damages. His mother presented weakness in her limbs in 50 s and died 15 years later. One of his younger sisters diagnosed as ALS for 6 years also carried the same mutation. She presented the similar symptoms on 41. No dominant upper motor neuron sign was showed. The clinical features were similar to the patients carrying the known mutation of p.Pro56Ser. A novel mutation of VAPB was found in one Chinese Han FALS pedigree. The affected patients presented a much slower progression and the lesions were limited in lower motor neurons.
Collapse
Affiliation(s)
- Yi-Min Sun
- Department of Neurology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Yi Dong
- Department of Neurology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Jian Wang
- Department of Neurology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Jia-Hong Lu
- Department of Neurology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Yan Chen
- Department of Neurology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| | - Jian-Jun Wu
- Department of Neurology, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
43
|
Liu J, Wang F. Role of Neuroinflammation in Amyotrophic Lateral Sclerosis: Cellular Mechanisms and Therapeutic Implications. Front Immunol 2017; 8:1005. [PMID: 28871262 PMCID: PMC5567007 DOI: 10.3389/fimmu.2017.01005] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/07/2017] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that affects upper motor neurons (MNs) comprising the corticospinal tract and lower MNs arising from the brain stem nuclei and ventral roots of the spinal cord, leading to fatal paralysis. Currently, there are no effective therapies for ALS. Increasing evidence indicates that neuroinflammation plays an important role in ALS pathogenesis. The neuroinflammation in ALS is characterized by infiltration of lymphocytes and macrophages, activation of microglia and reactive astrocytes, as well as the involvement of complement. In this review, we focus on the key cellular players of neuroinflammation during the pathogenesis of ALS by discussing not only their detrimental roles but also their immunomodulatory actions. We will summarize the pharmacological therapies for ALS that target neuroinflammation, as well as recent advances in the field of stem cell therapy aimed at modulating the inflammatory environment to preserve the remaining MNs in ALS patients and animal models of the disease.
Collapse
Affiliation(s)
- Jia Liu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fei Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|