1
|
Xu Z, Yi W, Guan L, Tang J, Feng D, Zou Y. Deciphering the Inhibitory Mechanism of ALS-Associated N352S and S352p Variants against TDP-43 Aggregation and Its Destabilization Effect on TDP-43 Protofibrils. ACS Chem Neurosci 2025; 16:1898-1908. [PMID: 40311013 DOI: 10.1021/acschemneuro.5c00045] [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] [Indexed: 05/03/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is closely related to ubiquitin-positive inclusions formed by transactive response deoxyribonucleic acid (DNA) binding protein of 43 kDa (TDP-43). Previous experiments identified that the ALS-linked familial variant, N352S (asparagine substituted by serine), and subsequent phosphorylation of S352 (S352p) are associated with the aggregation of TDP-43. However, the underlying molecular mechanisms are still not fully understood. By performing all-atom explicit-solvent replica exchange molecular dynamics (REMD) simulations with a total simulation time of 100.8 μs, we scrutinized the impact of the N352S mutation and its phosphorylation variant S352p on the conformational ensembles of the TDP-43342-366 dimer. Our simulation results show that both the N352S and S352p variants could promote the formation of unstructured conformation and impede the formation of β-structure and helix content, and the inhibitive effect of S352P is more obvious. Further analyses suggest that the H-bonding and hydrophobic interaction among TDP-43342-366 peptides, as well as the R361-E362 salt bridge, are attenuated by N352S and S352p variants. Additional MD simulations show that N352S and S352p variants reduce the structural stability of the hydrophobic region and lower the number of H-bonds and contacts of two hydrophobic clusters, thus possessing a destabilization effect on the TDP-43282-360 protofibrils. Our results unmask the molecular mechanism of the N352S mutation and its phosphorylation variant S352p toward the inhibition of TDP-43342-366 aggregation and prove the protofibril-destabilizing effects of these two variants, which may be helpful for designing drugs for the treatment of ALS.
Collapse
Affiliation(s)
- Zhengdong Xu
- Department of Physical Education, Shanghai University of Engineering Science, 333 Long Teng Road, Shanghai 201620, People's Republic of China
| | - Wenjuan Yi
- Department Sport and Exercise Science, College of Education, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310007, People's Republic of China
| | - Lulu Guan
- Department Sport and Exercise Science, College of Education, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310007, People's Republic of China
| | - Jiaxing Tang
- School of Physical Education, Xiangnan University, 889 Chenzhou Road, Chenzhou 423000, People's Republic of China
| | - Dushuo Feng
- Department Sport and Exercise Science, College of Education, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310007, People's Republic of China
| | - Yu Zou
- Department Sport and Exercise Science, College of Education, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310007, People's Republic of China
| |
Collapse
|
2
|
Fernandes APM, de Holanda LJ, de Lucena LC, da Silva KER, Lopes ACSM, Borges DT, Nagem DAP, Valentim RADM, Bougrain L, Rodrigues Lindquist AR. Electromyography as a tool to motion analysis for people with Amyotrophic Lateral Sclerosis: A protocol for a systematic review. PLoS One 2024; 19:e0302479. [PMID: 38805448 PMCID: PMC11132455 DOI: 10.1371/journal.pone.0302479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 03/30/2024] [Indexed: 05/30/2024] Open
Abstract
Biomechanical analysis of human movement plays an essential role in understanding functional changes in people with Amyotrophic Lateral Sclerosis (ALS), providing information on muscle impairment. Studies suggest that surface electromyography (sEMG) may be able to quantify muscle activity, identify levels of fatigue, assess muscle strength, and monitor variation in limb movement. In this article, a systematic review protocol will analyze the psychometric properties of the sEMG regarding the clinical data on the skeletal muscles of people with ALS. This protocol uses the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) methodological tool. A specific field structure was defined to reach each phase. Nine scientific databases (PubMed, Web of Science, Embase, Elsevier, IEEE, Google Scholar, SciELO, PEDro, LILACS E CENTRAL) were searched. The framework developed will extract data (i.e. study information, sample information, sEMG information, intervention, and outcomes) from the selected studies using a rigorous approach. The data will be described quantitatively using frequency and trend analysis methods, and heterogeneity between the included studies will be assessed using the I2 test. The risk of bias will be summarized using the most recent prediction model risk of bias assessment tool. Be sure to include relevant statistics here, such as sample sizes, response rates, P values or Confidence Intervals. Be specific (by stating the value) rather than general (eg, "there were differences between the groups"). This protocol will map out the construction of a systematic review that will identify and synthesize the advances in movement analysis of people with ALS through sEMG, using data extracted from articles.
Collapse
Affiliation(s)
- Ana Paula Mendonça Fernandes
- Department of Physical Therapy, Federal University of Rio Grande do Norte, Natal, Brazil
- Laboratory of Technological Innovation in Health, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Larissa Coutinho de Lucena
- Laboratory of Technological Innovation in Health, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Kelly Evangelista Rodrigues da Silva
- Department of Physical Therapy, Federal University of Rio Grande do Norte, Natal, Brazil
- Laboratory of Technological Innovation in Health, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Daniel Tezoni Borges
- Department of Physical Therapy, Federal University of Rio Grande do Norte, Natal, Brazil
- Laboratory of Technological Innovation in Health, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Danilo A. P. Nagem
- Laboratory of Technological Innovation in Health, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Biomedical Engineering, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Ricardo A. de M. Valentim
- Laboratory of Technological Innovation in Health, Federal University of Rio Grande do Norte, Natal, Brazil
- Department of Biomedical Engineering, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Laurent Bougrain
- Dept. of complex system, Artificial intelligence and robotics at Loria, University of Lorraine, Nancy, France
| | - Ana Raquel Rodrigues Lindquist
- Department of Physical Therapy, Federal University of Rio Grande do Norte, Natal, Brazil
- Laboratory of Technological Innovation in Health, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
3
|
Gonzalez D, Cuenca X, Allende ML. Knockdown of tgfb1a partially improves ALS phenotype in a transient zebrafish model. Front Cell Neurosci 2024; 18:1384085. [PMID: 38644973 PMCID: PMC11032012 DOI: 10.3389/fncel.2024.1384085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/15/2024] [Indexed: 04/23/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) corresponds to a neurodegenerative disorder marked by the progressive degeneration of both upper and lower motor neurons located in the brain, brainstem, and spinal cord. ALS can be broadly categorized into two main types: sporadic ALS (sALS), which constitutes approximately 90% of all cases, and familial ALS (fALS), which represents the remaining 10% of cases. Transforming growth factor type-β (TGF-β) is a cytokine involved in various cellular processes and pathological contexts, including inflammation and fibrosis. Elevated levels of TGF-β have been observed in the plasma and cerebrospinal fluid (CSF) of both ALS patients and mouse models. In this perspective, we explore the impact of the TGF-β signaling pathway using a transient zebrafish model for ALS. Our findings reveal that the knockdown of tgfb1a lead to a partial prevention of motor axon abnormalities and locomotor deficits in a transient ALS zebrafish model at 48 h post-fertilization (hpf). In this context, we delve into the proposed distinct roles of TGF-β in the progression of ALS. Indeed, some evidence suggests a dual role for TGF-β in ALS progression. Initially, it seems to exert a neuroprotective effect in the early stages, but paradoxically, it may contribute to disease progression in later stages. Consequently, we suggest that the TGF-β signaling pathway emerges as an attractive therapeutic target for treating ALS. Nevertheless, further research is crucial to comprehensively understand the nuanced role of TGF-β in the pathological context.
Collapse
Affiliation(s)
- David Gonzalez
- Departamento de Ciencias Químicas y Biológicas, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago, Chile
- Escuela de Terapia Ocupacional, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago, Chile
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Xiomara Cuenca
- Escuela de Terapia Ocupacional, Facultad de Ciencias de la Salud, Universidad Bernardo O’Higgins, Santiago, Chile
| | - Miguel L. Allende
- Millennium Institute Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
4
|
Sahota VK, Stone A, Woodling NS, Spiers JG, Steinert JR, Partridge L, Augustin H. Plum modulates Myoglianin and regulates synaptic function in D. melanogaster. Open Biol 2023; 13:230171. [PMID: 37699519 PMCID: PMC10497343 DOI: 10.1098/rsob.230171] [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: 06/03/2023] [Accepted: 08/14/2023] [Indexed: 09/14/2023] Open
Abstract
Alterations in the neuromuscular system underlie several neuromuscular diseases and play critical roles in the development of sarcopenia, the age-related loss of muscle mass and function. Mammalian Myostatin (MST) and GDF11, members of the TGF-β superfamily of growth factors, are powerful regulators of muscle size in both model organisms and humans. Myoglianin (MYO), the Drosophila homologue of MST and GDF11, is a strong inhibitor of synaptic function and structure at the neuromuscular junction in flies. Here, we identified Plum, a transmembrane cell surface protein, as a modulator of MYO function in the larval neuromuscular system. Reduction of Plum in the larval body-wall muscles abolishes the previously demonstrated positive effect of attenuated MYO signalling on both muscle size and neuromuscular junction structure and function. In addition, downregulation of Plum on its own results in decreased synaptic strength and body weight, classifying Plum as a (novel) regulator of neuromuscular function and body (muscle) size. These findings offer new insights into possible regulatory mechanisms behind ageing- and disease-related neuromuscular dysfunctions in humans and identify potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Virender K. Sahota
- Department of Biological Sciences, Centre for Biomedical Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Aelfwin Stone
- Faculty of Medicine & Health Sciences, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Nathaniel S. Woodling
- Department of Biological Sciences, Centre for Biomedical Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Jereme G. Spiers
- Faculty of Medicine & Health Sciences, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Joern R. Steinert
- Faculty of Medicine & Health Sciences, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - Linda Partridge
- Institute of Healthy Ageing, and GEE, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne 50931, Germany
| | - Hrvoje Augustin
- Department of Biological Sciences, Centre for Biomedical Sciences, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
- Institute of Healthy Ageing, and GEE, University College London, Darwin Building, Gower Street, London WC1E 6BT, UK
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne 50931, Germany
| |
Collapse
|
5
|
Colasuonno F, Price R, Moreno S. Upper and Lower Motor Neurons and the Skeletal Muscle: Implication for Amyotrophic Lateral Sclerosis (ALS). ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:111-129. [PMID: 37955773 DOI: 10.1007/978-3-031-38215-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The relationships between motor neurons and the skeletal muscle during development and in pathologic contexts are addressed in this Chapter.We discuss the developmental interplay of muscle and nervous tissue, through neurotrophins and the activation of differentiation and survival pathways. After a brief overview on muscular regulatory factors, we focus on the contribution of muscle to early and late neurodevelopment. Such a role seems especially intriguing in relation to the epigenetic shaping of developing motor neuron fate choices. In this context, emphasis is attributed to factors regulating energy metabolism, which may concomitantly act in muscle and neural cells, being involved in common pathways.We then review the main features of motor neuron diseases, addressing the cellular processes underlying clinical symptoms. The involvement of different muscle-associated neurotrophic factors for survival of lateral motor column neurons, innervating MyoD-dependent limb muscles, and of medial motor column neurons, innervating Myf5-dependent back musculature is discussed. Among the pathogenic mechanisms, we focus on oxidative stress, that represents a common and early trait in several neurodegenerative disorders. The role of organelles primarily involved in reactive oxygen species scavenging and, more generally, in energy metabolism-namely mitochondria and peroxisomes-is discussed in the frame of motor neuron degeneration.We finally address muscular involvement in amyotrophic lateral sclerosis (ALS), a multifactorial degenerative disorder, hallmarked by severe weight loss, caused by imbalanced lipid metabolism. Even though multiple mechanisms have been recognized to play a role in the disease, current literature generally assumes that the primum movens is neuronal degeneration and that muscle atrophy is only a consequence of such pathogenic event. However, several lines of evidence point to the muscle as primarily involved in the disease, mainly through its role in energy homeostasis. Data from different ALS mouse models strongly argue for an early mitochondrial dysfunction in muscle tissue, possibly leading to motor neuron disturbances. Detailed understanding of skeletal muscle contribution to ALS pathogenesis will likely lead to the identification of novel therapeutic strategies.
Collapse
Affiliation(s)
- Fiorella Colasuonno
- Department of Experimental Medicine , University of Rome "Tor Vergata", Rome, Italy
- Department of Science, LIME, University Roma Tre, Rome, Italy
| | - Rachel Price
- Department of Science, LIME, University Roma Tre, Rome, Italy
- Laboratory of Neurodevelopmental Biology, Neurogenetics and Molecular Neurobiology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Sandra Moreno
- Department of Science, LIME, University Roma Tre, Rome, Italy.
- Laboratory of Neurodevelopmental Biology, Neurogenetics and Molecular Neurobiology, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
6
|
Molendijk J, Blazev R, Mills RJ, Ng YK, Watt KI, Chau D, Gregorevic P, Crouch PJ, Hilton JBW, Lisowski L, Zhang P, Reue K, Lusis AJ, Hudson JE, James DE, Seldin MM, Parker BL. Proteome-wide systems genetics identifies UFMylation as a regulator of skeletal muscle function. eLife 2022; 11:e82951. [PMID: 36472367 PMCID: PMC9833826 DOI: 10.7554/elife.82951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Improving muscle function has great potential to improve the quality of life. To identify novel regulators of skeletal muscle metabolism and function, we performed a proteomic analysis of gastrocnemius muscle from 73 genetically distinct inbred mouse strains, and integrated the data with previously acquired genomics and >300 molecular/phenotypic traits via quantitative trait loci mapping and correlation network analysis. These data identified thousands of associations between protein abundance and phenotypes and can be accessed online (https://muscle.coffeeprot.com/) to identify regulators of muscle function. We used this resource to prioritize targets for a functional genomic screen in human bioengineered skeletal muscle. This identified several negative regulators of muscle function including UFC1, an E2 ligase for protein UFMylation. We show UFMylation is up-regulated in a mouse model of amyotrophic lateral sclerosis, a disease that involves muscle atrophy. Furthermore, in vivo knockdown of UFMylation increased contraction force, implicating its role as a negative regulator of skeletal muscle function.
Collapse
Affiliation(s)
- Jeffrey Molendijk
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
- Centre for Muscle Research, University of MelbourneMelbourneAustralia
| | - Ronnie Blazev
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
- Centre for Muscle Research, University of MelbourneMelbourneAustralia
| | | | - Yaan-Kit Ng
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
- Centre for Muscle Research, University of MelbourneMelbourneAustralia
| | - Kevin I Watt
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
- Centre for Muscle Research, University of MelbourneMelbourneAustralia
| | - Daryn Chau
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, IrvineIrvineUnited States
| | - Paul Gregorevic
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
- Centre for Muscle Research, University of MelbourneMelbourneAustralia
| | - Peter J Crouch
- Department of Biochemistry and Pharmacology, University of MelbourneMelbourneAustralia
| | - James BW Hilton
- Department of Biochemistry and Pharmacology, University of MelbourneMelbourneAustralia
| | - Leszek Lisowski
- Children's Medical Research Institute, University of SydneySydneyAustralia
- Military Institute of MedicineWarszawaPoland
| | - Peixiang Zhang
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Karen Reue
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Aldons J Lusis
- Department of Human Genetics/Medicine, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los AngelesLos AngelesUnited States
| | - James E Hudson
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Science, School of Medical Science, University of SydneySydneyAustralia
| | - Marcus M Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, IrvineIrvineUnited States
| | - Benjamin L Parker
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
- Centre for Muscle Research, University of MelbourneMelbourneAustralia
| |
Collapse
|
7
|
Anakor E, Duddy WJ, Duguez S. The Cellular and Molecular Signature of ALS in Muscle. J Pers Med 2022; 12:1868. [PMID: 36579600 PMCID: PMC9692882 DOI: 10.3390/jpm12111868] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022] Open
Abstract
Amyotrophic lateral sclerosis is a disease affecting upper and lower motor neurons. Although motor neuron death is the core event of ALS pathology, it is increasingly recognized that other tissues and cell types are affected in the disease, making potentially major contributions to the occurrence and progression of pathology. We review here the known cellular and molecular characteristics of muscle tissue affected by ALS. Evidence of toxicity in skeletal muscle tissue is considered, including metabolic dysfunctions, impaired proteostasis, and deficits in muscle regeneration and RNA metabolism. The role of muscle as a secretory organ, and effects on the skeletal muscle secretome are also covered, including the increase in secretion of toxic factors or decrease in essential factors that have consequences for neuronal function and survival.
Collapse
Affiliation(s)
| | | | - Stephanie Duguez
- Northern Ireland Center for Personalised Medicine, School of Medicine, Ulster University, Derry-Londonderry BT47 6SB, UK
| |
Collapse
|
8
|
Kolasa M, Czerczak K, Fraczyk J, Szymanski L, Lewicki S, Bednarowicz A, Tarzynska N, Sikorski D, Szparaga G, Draczynski Z, Cierniak S, Brzoskowska U, Galita G, Majsterek I, Bociaga D, Krol P, Kolesinska B. Evaluation of Polysaccharide-Peptide Conjugates Containing the RGD Motif for Potential Use in Muscle Tissue Regeneration. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6432. [PMID: 36143745 PMCID: PMC9503514 DOI: 10.3390/ma15186432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023]
Abstract
New scaffold materials composed of biodegradable components are of great interest in regenerative medicine. These materials should be: stable, nontoxic, and biodegrade slowly and steadily, allowing the stable release of biodegradable and biologically active substances. We analyzed peptide-polysaccharide conjugates derived from peptides containing RGD motif (H-RGDS-OH (1), H-GRGDS-NH2 (2), and cyclo(RGDfC) (3)) and polysaccharides as scaffolds to select the most appropriate biomaterials for application in regenerative medicine. Based on the results of MTT and Ki-67 assays, we can state that the conjugates containing calcium alginate and the ternary nonwoven material were the most supportive of muscle tissue regeneration. Scanning electron microscopy imaging and light microscopy studies with hematoxylin-eosin staining showed that C2C12 cells were able to interact with the tested peptide-polysaccharide conjugates. The release factor (Q) varied depending on both the peptide and the structure of the polysaccharide matrix. LDH, Alamarblue®, Ki-67, and cell cycle assays indicated that peptides 1 and 2 were characterized by the best biological properties. Conjugates containing chitosan and the ternary polysaccharide nonwoven with peptide 1 exhibited very high antibacterial activity against Staphylococcus aureus and Klebsiella pneumoniae. Overall, the results of the study suggested that polysaccharide conjugates with peptides 1 and 2 can be potentially used in regenerative medicine.
Collapse
Affiliation(s)
- Marcin Kolasa
- Military Institute of Hygiene and Epidemiology, Department of Pharmacology and Toxicology, Kozielska 4, 01-163 Warsaw, Poland
| | - Katarzyna Czerczak
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Justyna Fraczyk
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Lukasz Szymanski
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland
| | - Slawomir Lewicki
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland
| | - Anna Bednarowicz
- Institute of Material Sciences of Textiles and Polymer Composites, Faculty of Material Technologies and Textile Design, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Nina Tarzynska
- Institute of Material Sciences of Textiles and Polymer Composites, Faculty of Material Technologies and Textile Design, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Dominik Sikorski
- Institute of Material Sciences of Textiles and Polymer Composites, Faculty of Material Technologies and Textile Design, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Grzegorz Szparaga
- Institute of Material Sciences of Textiles and Polymer Composites, Faculty of Material Technologies and Textile Design, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Zbigniew Draczynski
- Institute of Material Sciences of Textiles and Polymer Composites, Faculty of Material Technologies and Textile Design, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | | | | | - Grzegorz Galita
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland
| | - Dorota Bociaga
- Institute of Materials Science and Engineering, Lodz University of Technology, Stefanowskiego 1/15, 90-537 Lodz, Poland
| | - Paulina Krol
- Lukasiewicz Research Network-Textile Research Institute, Brzezinska 5/15, 92-103 Lodz, Poland
| | - Beata Kolesinska
- Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| |
Collapse
|
9
|
Peggion C, Scalcon V, Massimino ML, Nies K, Lopreiato R, Rigobello MP, Bertoli A. SOD1 in ALS: Taking Stock in Pathogenic Mechanisms and the Role of Glial and Muscle Cells. Antioxidants (Basel) 2022; 11:614. [PMID: 35453299 PMCID: PMC9032988 DOI: 10.3390/antiox11040614] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/04/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by the loss of motor neurons in the brain and spinal cord. While the exact causes of ALS are still unclear, the discovery that familial cases of ALS are related to mutations in the Cu/Zn superoxide dismutase (SOD1), a key antioxidant enzyme protecting cells from the deleterious effects of superoxide radicals, suggested that alterations in SOD1 functionality and/or aberrant SOD1 aggregation strongly contribute to ALS pathogenesis. A new scenario was opened in which, thanks to the generation of SOD1 related models, different mechanisms crucial for ALS progression were identified. These include excitotoxicity, oxidative stress, mitochondrial dysfunctions, and non-cell autonomous toxicity, also implicating altered Ca2+ metabolism. While most of the literature considers motor neurons as primary target of SOD1-mediated effects, here we mainly discuss the effects of SOD1 mutations in non-neuronal cells, such as glial and skeletal muscle cells, in ALS. Attention is given to the altered redox balance and Ca2+ homeostasis, two processes that are strictly related with each other. We also provide original data obtained in primary myocytes derived from hSOD1(G93A) transgenic mice, showing perturbed expression of Ca2+ transporters that may be responsible for altered mitochondrial Ca2+ fluxes. ALS-related SOD1 mutants are also responsible for early alterations of fundamental biological processes in skeletal myocytes that may impinge on skeletal muscle functions and the cross-talk between muscle cells and motor neurons during disease progression.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (C.P.); (V.S.); (K.N.); (R.L.)
| | - Valeria Scalcon
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (C.P.); (V.S.); (K.N.); (R.L.)
| | | | - Kelly Nies
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (C.P.); (V.S.); (K.N.); (R.L.)
- Department of Radiology, CARIM School for Cardiovascular Diseases, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Raffaele Lopreiato
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (C.P.); (V.S.); (K.N.); (R.L.)
| | - Maria Pia Rigobello
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (C.P.); (V.S.); (K.N.); (R.L.)
| | - Alessandro Bertoli
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; (C.P.); (V.S.); (K.N.); (R.L.)
- CNR—Neuroscience Institute, 35131 Padova, Italy;
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy
| |
Collapse
|
10
|
Edgar JA, Molyneux RJ, Colegate SM. 1,2-Dehydropyrrolizidine Alkaloids: Their Potential as a Dietary Cause of Sporadic Motor Neuron Diseases. Chem Res Toxicol 2022; 35:340-354. [PMID: 35238548 DOI: 10.1021/acs.chemrestox.1c00384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Sporadic motor neuron diseases (MNDs), such as amyotrophic lateral sclerosis (ALS), can be caused by spontaneous genetic mutations. However, many sporadic cases of ALS and other debilitating neurodegenerative diseases (NDDs) are believed to be caused by environmental factors, subject to considerable debate and requiring intensive research. A common pathology associated with MND development involves progressive mitochondrial dysfunction and oxidative stress in motor neurons and glial cells of the central nervous system (CNS), leading to apoptosis. Consequent degeneration of skeletal and respiratory muscle cells can lead to death from respiratory failure. A significant number of MND cases present with cancers and liver and lung pathology. This Perspective explores the possibility that MNDs could be caused by intermittent, low-level dietary exposure to 1,2-dehydropyrrolizidine alkaloids (1,2-dehydroPAs) that are increasingly recognized as contaminants of many foods consumed throughout the world. Nontoxic, per se, 1,2-dehydroPAs are metabolized, by particular cytochrome P450 (CYP450) isoforms, to 6,7-dihydropyrrolizines that react with nucleophilic groups (-NH, -SH, -OH) on DNA, proteins, and other vital biochemicals, such as glutathione. Many factors, including aging, gender, smoking, and alcohol consumption, influence CYP450 isoform activity in a range of tissues, including glial cells and neurons of the CNS. Activation of 1,2-dehydroPAs in CNS cells can be expected to cause gene mutations and oxidative stress, potentially leading to the development of MNDs and other NDDs. While relatively high dietary exposure to 1,2-dehydroPAs causes hepatic sinusoidal obstruction syndrome, pulmonary venoocclusive disease, neurotoxicity, and diverse cancers, this Perspective suggests that, at current intermittent, low levels of dietary exposure, neurotoxicity could become the primary pathology that develops over time in susceptible individuals, along with a tendency for some of them to also display liver and lung pathology and diverse cancers co-occurring with some MND/NDD cases. Targeted research is recommended to investigate this proposal.
Collapse
Affiliation(s)
- John A Edgar
- CSIRO Agriculture and Food, 11 Julius Avenue, North Ryde, New South Wales 2113, Australia
| | - Russell J Molyneux
- Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, 200 West Kawili Street, Hilo, Hawaii 96720, United States
| | - Steven M Colegate
- Poisonous Plant Research Laboratory, ARS/USDA, 1150 East 1400 North, Logan, Utah 84341, United States
| |
Collapse
|
11
|
Badu-Mensah A, Guo X, Hickman JJ. ALS Skeletal Muscle: Victim or Culprit. THE NEUROSCIENCE CHRONICLES 2022; 2:31-33. [PMID: 35098252 PMCID: PMC8793963 DOI: 10.46439/neuroscience.2.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Agnes Badu-Mensah
- University of Central Florida College of Medicine, Burnett School of Biomedical Sciences, Orlando, Florida, USA.,Hickman Hybrid Systems Laboratory NanoScience Technology Center University of Central Florida Orlando, Florida, USA
| | - Xiufang Guo
- University of Central Florida College of Medicine, Burnett School of Biomedical Sciences, Orlando, Florida, USA.,Hickman Hybrid Systems Laboratory NanoScience Technology Center University of Central Florida Orlando, Florida, USA
| | - James J Hickman
- University of Central Florida College of Medicine, Burnett School of Biomedical Sciences, Orlando, Florida, USA.,Hickman Hybrid Systems Laboratory NanoScience Technology Center University of Central Florida Orlando, Florida, USA
| |
Collapse
|
12
|
Harlaar L, Ciet P, van Tulder G, Brusse E, Timmermans RGM, Janssen WGM, de Bruijne M, van der Ploeg AT, Tiddens HAWM, van Doorn PA, van der Beek NAME. Diaphragmatic dysfunction in neuromuscular disease, an MRI study. Neuromuscul Disord 2021; 32:15-24. [PMID: 34973872 DOI: 10.1016/j.nmd.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/03/2021] [Accepted: 11/02/2021] [Indexed: 11/19/2022]
Abstract
The aim of this exploratory study was to evaluate diaphragmatic function across various neuromuscular diseases using spirometry-controlled MRI. We measured motion of the diaphragm relative to that of the thoracic wall (cranial-caudal ratio vs. anterior posterior ratio; CC-AP ratio), and changes in the diaphragmatic curvature (diaphragm height and area ratio) during inspiration in 12 adults with a neuromuscular disease having signs of respiratory muscle weakness, 18 healthy controls, and 35 adult Pompe patients - a group with prominent diaphragmatic weakness. CC-AP ratio was lower in patients with myopathies (n=7, 1.25±0.30) and motor neuron diseases (n=5, 1.30±0.10) than in healthy controls (1.37±0.14; p=0.001 and p=0.008), but not as abnormal as in Pompe patients (1.12±0.18; p=0.011 and p=0.024). The mean diaphragm height ratio was 1.17±0.33 in patients with myopathies, pointing at an insufficient diaphragmatic contraction. This was also seen in patients with Pompe disease (1.28±0.36), but not in healthy controls (0.82±0.33) or patients with motor neuron disease (0.82±0.24). We conclude that spirometry-controlled MRI enables us to investigate respiratory dysfunction across neuromuscular diseases, suggesting that the diaphragm is affected in a different way in myopathies and motor neuron diseases. Whether MRI can also be used to evaluate progression of diaphragmatic dysfunction requires additional studies.
Collapse
Affiliation(s)
- Laurike Harlaar
- Erasmus MC, University Medical Center Rotterdam, Center for Lysosomal and Metabolic Diseases, Department of Neurology, Rotterdam, the Netherlands; Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Center for Lysosomal and Metabolic Diseases, Department of Paediatrics, Rotterdam, the Netherlands
| | - Pierluigi Ciet
- Erasmus MC, University Medical Center Rotterdam, Department of Radiology and Nuclear Medicine, Rotterdam, the Netherlands; Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Department of Respiratory Medicine and Allergology, Rotterdam, the Netherlands
| | - Gijs van Tulder
- Erasmus MC, University Medical Center Rotterdam, Department of Radiology and Nuclear Medicine, Rotterdam, the Netherlands
| | - Esther Brusse
- Erasmus MC, University Medical Center Rotterdam, Center for Lysosomal and Metabolic Diseases, Department of Neurology, Rotterdam, the Netherlands
| | - Remco G M Timmermans
- Rijndam Rehabilitation Centre Rotterdam, location Erasmus MC, Rotterdam, the Netherlands
| | - Wim G M Janssen
- Rijndam Rehabilitation Centre Rotterdam, location Erasmus MC, Rotterdam, the Netherlands
| | - Marleen de Bruijne
- Erasmus MC, University Medical Center Rotterdam, Department of Radiology and Nuclear Medicine, Rotterdam, the Netherlands; University of Copenhagen, Department of Computer Science, Copenhagen, Denmark
| | - Ans T van der Ploeg
- Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Center for Lysosomal and Metabolic Diseases, Department of Paediatrics, Rotterdam, the Netherlands
| | - Harm A W M Tiddens
- Erasmus MC, University Medical Center Rotterdam, Department of Radiology and Nuclear Medicine, Rotterdam, the Netherlands; Erasmus MC - Sophia Children's Hospital, University Medical Center Rotterdam, Department of Respiratory Medicine and Allergology, Rotterdam, the Netherlands
| | - Pieter A van Doorn
- Erasmus MC, University Medical Center Rotterdam, Center for Lysosomal and Metabolic Diseases, Department of Neurology, Rotterdam, the Netherlands
| | - Nadine A M E van der Beek
- Erasmus MC, University Medical Center Rotterdam, Center for Lysosomal and Metabolic Diseases, Department of Neurology, Rotterdam, the Netherlands.
| |
Collapse
|
13
|
Carton F, Di Francesco D, Fusaro L, Zanella E, Apostolo C, Oltolina F, Cotella D, Prat M, Boccafoschi F. Myogenic Potential of Extracellular Matrix Derived from Decellularized Bovine Pericardium. Int J Mol Sci 2021; 22:ijms22179406. [PMID: 34502309 PMCID: PMC8431302 DOI: 10.3390/ijms22179406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscles represent 40% of body mass and its native regenerative capacity can be permanently lost after a traumatic injury, congenital diseases, or tumor ablation. The absence of physiological regeneration can hinder muscle repair preventing normal muscle tissue functions. To date, tissue engineering (TE) represents one promising option for treating muscle injuries and wasting. In particular, hydrogels derived from the decellularized extracellular matrix (dECM) are widely investigated in tissue engineering applications thanks to their essential role in guiding muscle regeneration. In this work, the myogenic potential of dECM substrate, obtained from decellularized bovine pericardium (Tissuegraft Srl), was evaluated in vitro using C2C12 murine muscle cells. To assess myotubes formation, the width, length, and fusion indexes were measured during the differentiation time course. Additionally, the ability of dECM to support myogenesis was assessed by measuring the expression of specific myogenic markers: α-smooth muscle actin (α-sma), myogenin, and myosin heavy chain (MHC). The results obtained suggest that the dECM niche was able to support and enhance the myogenic potential of C2C12 cells in comparison of those grown on a plastic standard surface. Thus, the use of extracellular matrix proteins, as biomaterial supports, could represent a promising therapeutic strategy for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
- Flavia Carton
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy; (F.C.); (D.D.F.); (E.Z.); (C.A.); (F.O.); (D.C.); (M.P.)
| | - Dalila Di Francesco
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy; (F.C.); (D.D.F.); (E.Z.); (C.A.); (F.O.); (D.C.); (M.P.)
| | | | - Emma Zanella
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy; (F.C.); (D.D.F.); (E.Z.); (C.A.); (F.O.); (D.C.); (M.P.)
| | - Claudio Apostolo
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy; (F.C.); (D.D.F.); (E.Z.); (C.A.); (F.O.); (D.C.); (M.P.)
| | - Francesca Oltolina
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy; (F.C.); (D.D.F.); (E.Z.); (C.A.); (F.O.); (D.C.); (M.P.)
| | - Diego Cotella
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy; (F.C.); (D.D.F.); (E.Z.); (C.A.); (F.O.); (D.C.); (M.P.)
| | - Maria Prat
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy; (F.C.); (D.D.F.); (E.Z.); (C.A.); (F.O.); (D.C.); (M.P.)
| | - Francesca Boccafoschi
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, 28100 Novara, Italy; (F.C.); (D.D.F.); (E.Z.); (C.A.); (F.O.); (D.C.); (M.P.)
- Correspondence: ; Tel.: +39-0321-660-556
| |
Collapse
|
14
|
Rebolledo DL, Lipson KE, Brandan E. Driving fibrosis in neuromuscular diseases: Role and regulation of Connective tissue growth factor (CCN2/CTGF). Matrix Biol Plus 2021; 11:100059. [PMID: 34435178 PMCID: PMC8377001 DOI: 10.1016/j.mbplus.2021.100059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Connective tissue growth factor or cellular communication network 2 (CCN2/CTGF) is a matricellular protein member of the CCN family involved in several crucial biological processes. In skeletal muscle, CCN2/CTGF abundance is elevated in human muscle biopsies and/or animal models for diverse neuromuscular pathologies, including muscular dystrophies, neurodegenerative disorders, muscle denervation, and muscle overuse. In this context, CCN2/CTGF is deeply involved in extracellular matrix (ECM) modulation, acting as a strong pro-fibrotic factor that promotes excessive ECM accumulation. Reducing CCN2/CTGF levels or biological activity in pathological conditions can decrease fibrosis, improve muscle architecture and function. In this work, we summarize information about the role of CCN2/CTGF in fibrosis associated with neuromuscular pathologies and the mechanisms and signaling pathways that regulate their expression in skeletal muscle.
Collapse
Affiliation(s)
- Daniela L Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Punta Arenas, Chile
| | | | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Chile.,Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile.,Fundación Ciencia y Vida, Santiago, Chile
| |
Collapse
|
15
|
Méndez-López I, Sancho-Bielsa FJ, Engel T, García AG, Padín JF. Progressive Mitochondrial SOD1 G93A Accumulation Causes Severe Structural, Metabolic and Functional Aberrations through OPA1 Down-Regulation in a Mouse Model of Amyotrophic Lateral Sclerosis. Int J Mol Sci 2021; 22:ijms22158194. [PMID: 34360957 PMCID: PMC8347639 DOI: 10.3390/ijms22158194] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 01/20/2023] Open
Abstract
In recent years, the “non-autonomous motor neuron death” hypothesis has become more consolidated behind amyotrophic lateral sclerosis (ALS). It postulates that cells other than motor neurons participate in the pathology. In fact, the involvement of the autonomic nervous system is fundamental since patients die of sudden death when they become unable to compensate for cardiorespiratory arrest. Mitochondria are thought to play a fundamental role in the physiopathology of ALS, as they are compromised in multiple ALS models in different cell types, and it also occurs in other neurodegenerative diseases. Our study aimed to uncover mitochondrial alterations in the sympathoadrenal system of a mouse model of ALS, from a structural, bioenergetic and functional perspective during disease instauration. We studied the adrenal chromaffin cell from mutant SOD1G93A mouse at pre-symptomatic and symptomatic stages. The mitochondrial accumulation of the mutated SOD1G93A protein and the down-regulation of optic atrophy protein-1 (OPA1) provoke mitochondrial ultrastructure alterations prior to the onset of clinical symptoms. These changes affect mitochondrial fusion dynamics, triggering mitochondrial maturation impairment and cristae swelling, with increased size of cristae junctions. The functional consequences are a loss of mitochondrial membrane potential and changes in the bioenergetics profile, with reduced maximal respiration and spare respiratory capacity of mitochondria, as well as enhanced production of reactive oxygen species. This study identifies mitochondrial dynamics regulator OPA1 as an interesting therapeutic target in ALS. Additionally, our findings in the adrenal medulla gland from presymptomatic stages highlight the relevance of sympathetic impairment in this disease. Specifically, we show new SOD1G93A toxicity pathways affecting cellular energy metabolism in non-motor neurons, which offer a possible link between cell specific metabolic phenotype and the progression of ALS.
Collapse
Affiliation(s)
- Iago Méndez-López
- Instituto Teófilo Hernando and Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain; (I.M.-L.); (A.G.G.)
| | - Francisco J. Sancho-Bielsa
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha (UCLM), 13005 Ciudad Real, Spain;
| | - Tobias Engel
- Department of Physiology & Medical Physics, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- FutureNeuro SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland
| | - Antonio G. García
- Instituto Teófilo Hernando and Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain; (I.M.-L.); (A.G.G.)
| | - Juan Fernando Padín
- Instituto Teófilo Hernando and Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain; (I.M.-L.); (A.G.G.)
- Departamento de Ciencias Médicas, Facultad de Medicina, Universidad de Castilla-La Mancha (UCLM), 13005 Ciudad Real, Spain;
- Correspondence:
| |
Collapse
|
16
|
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
|
17
|
Russell KL, Downie JM, Gibson SB, Tsetsou S, Keefe MD, Duran JA, Figueroa KP, Bromberg MB, Murtaugh LC, Bonkowsky JL, Pulst SM, Jorde LB. Pathogenic Effect of TP73 Gene Variants in People With Amyotrophic Lateral Sclerosis. Neurology 2021; 97:e225-e235. [PMID: 34135078 PMCID: PMC8302149 DOI: 10.1212/wnl.0000000000012285] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/13/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To identify novel disease associated loci for amyotrophic lateral sclerosis (ALS), we used sequencing data and performed in vitro and in vivo experiments to demonstrate pathogenicity of mutations identified in TP73. METHODS We analyzed exome sequences of 87 patients with sporadic ALS and 324 controls, with confirmatory sequencing in independent ALS cohorts of >2,800 patients. For the top hit, TP73, a regulator of apoptosis and differentiation and a binding partner and homolog of the tumor suppressor gene TP53, we assayed mutation effects using in vitro and in vivo experiments. C2C12 myoblast differentiation assays, characterization of myotube appearance, and immunoprecipitation of p53-p73 complexes were performed in vitro. In vivo, we used clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 targeting of zebrafish tp73 to assay motor neuron number and axon morphology. RESULTS Four heterozygous rare, nonsynonymous mutations in TP73 were identified in our sporadic ALS cohort. In independent ALS cohorts, we identified an additional 19 rare, deleterious variants in TP73. Patient TP73 mutations caused abnormal differentiation and increased apoptosis in the myoblast differentiation assay, with abnormal myotube appearance. Immunoprecipitation of mutant ΔN-p73 demonstrated that patient mutations hinder the ability of ΔN-p73 to bind p53. CRISPR/Cas9 knockout of tp73 in zebrafish led to impaired motor neuron development and abnormal axonal morphology, concordant with ALS pathology. CONCLUSION Together, these results strongly suggest that variants in TP73 correlate with risk for ALS and indicate a role for apoptosis in ALS disease pathology.
Collapse
Affiliation(s)
- Kristi L Russell
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT.
| | - Jonathan M Downie
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - Summer B Gibson
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - Spyridoula Tsetsou
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - Matthew D Keefe
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - Jerry A Duran
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - Karla P Figueroa
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - Mark B Bromberg
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - L Charles Murtaugh
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - Joshua L Bonkowsky
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - Stefan M Pulst
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| | - Lynn B Jorde
- From the Departments of Human Genetics (K.L.R., J.A.D., L.C.M., L.B.J.), Neurology (S.B.G., K.P.F., M.B.B., S.M.P.), and Pediatrics (M.D.K., J.L.B.), University of Utah School of Medicine, Salt Lake City; Department of Medicine (J.M.D.), Massachusetts General Hospital, Boston; Department of Neurosurgery (S.T.), Mount Sinai Hospital, Icahn School of Medicine, New York, NY; and Brain and Spine Center (J.L.B.), Primary Children's Hospital, Salt Lake City, UT
| |
Collapse
|
18
|
Bang S, Lee S, Choi N, Kim HN. Emerging Brain-Pathophysiology-Mimetic Platforms for Studying Neurodegenerative Diseases: Brain Organoids and Brains-on-a-Chip. Adv Healthc Mater 2021; 10:e2002119. [PMID: 34028201 DOI: 10.1002/adhm.202002119] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/25/2021] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases are a group of disorders characterized by progressive degeneration of the structural and functional integrity of the central and peripheral nervous systems. Millions of people suffer from degenerative brain diseases worldwide, and the mortality continues to increase every year, causing a growing demand for knowledge of the underlying mechanisms and development of therapeutic targets. Conventional 2D-based cell culture platforms and animal models cannot fully recapitulate the pathophysiology, and this has limited the capability for estimating drug efficacy. Recently, engineered platforms, including brain organoids and brain-on-a-chip, have emerged. They mimic the physiology of brain tissue and reflect the fundamental pathophysiological signatures of neurodegenerative diseases, such as the accumulation of neurotoxic proteins, structural abnormalities, and functional loss. In this paper, recent advances in brain-mimetic platforms and their potential for modeling features of neurodegenerative diseases in vitro are reviewed. The development of a physiologically relevant model should help overcome unresolved neurodegenerative diseases.
Collapse
Affiliation(s)
- Seokyoung Bang
- Brain Science Institute Korea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
| | - Songhyun Lee
- Department of Medical Engineering Yonsei University College of Medicine Seoul 03722 Republic of Korea
| | - Nakwon Choi
- Brain Science Institute Korea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
- KU‐KIST Graduate School of Converging Science and Technology Korea University Seoul 02841 Republic of Korea
| | - Hong Nam Kim
- Brain Science Institute Korea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
- Division of Bio‐Medical Science & Technology KIST School Korea University of Science and Technology (UST) Seoul 02792 Republic of Korea
| |
Collapse
|
19
|
Yu H, Chen L, Zhang S, He J, Fan D. Early Axonal Dysfunction of the Peripheral Nervous System Influences Disease Progression of ALS: Evidence From Clinical Neuroelectrophysiology. Front Neurol 2021; 12:574919. [PMID: 33643181 PMCID: PMC7905229 DOI: 10.3389/fneur.2021.574919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 01/20/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: To assess the prognostic value of the decrement in compound muscle action potential amplitude within 12 months of symptom onset (CMAP-12 amplitude) for the survival of patients with amyotrophic lateral sclerosis (ALS). Methods: Patients were stratified into 4 groups according to the decrement of the CMAP-12 amplitudes: normal (≥the lower limit of normal, LLN), mild (<LLN but ≥50% of LLN), moderate (<50% but ≥30% of LLN) and severe (<30% of LLN). All patients were followed up every 3 months. Survival was analyzed using the Kaplan-Meier method and Cox proportional hazards regression. Results: A total of 149 patients were included in the analysis [90 males (60.4%); mean age at onset, 50.7 years]. The decrement of CMAP-12 amplitudes was normal in 24.2% of patients, mild in 22.1%, moderate in 15.4% and severe in 38.3%. Kaplan–Meier analysis showed there was a significant difference in the overall survival across the 4 groups (p < 0.05). Further pairwise comparisons identified significant differences in survival between the normal vs. the moderate group (p < 0.05) and the normal vs. the severe group (p < 0.01). There was a significant inverse correlation between the CMAP-12 amplitude and overall survival. Compared to that in the normal group, survival in the moderately and severely decreased groups was significantly shorter (HR 3.394, 95% CI 1.292–8.917, p = 0.013; and HR 4.732, 95% CI 2.032–11.017; p = 0.000, respectively). Conclusions: Our results suggest that CMAP-12 amplitude could be a prognostic indicator of disease progression in ALS. More importantly, our findings provide clinical evidence for the viewpoint that early axonal dysfunction of the peripheral nervous system accelerates disease progression of ALS.
Collapse
Affiliation(s)
- Huiyan Yu
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Department of Neurology, National Center of Gerontology, Beijing Hospital, Beijing, China
| | - Lu Chen
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Shuo Zhang
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Jing He
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Department of Neurology, National Center of Gerontology, Beijing Hospital, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China.,Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China.,Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China
| |
Collapse
|
20
|
de Jongh R, Spijkers XM, Pasteuning-Vuhman S, Vulto P, Pasterkamp RJ. Neuromuscular junction-on-a-chip: ALS disease modeling and read-out development in microfluidic devices. J Neurochem 2021; 157:393-412. [PMID: 33382092 DOI: 10.1111/jnc.15289] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 12/21/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and progressive neurodegenerative disease affecting upper and lower motor neurons with no cure available. Clinical and animal studies reveal that the neuromuscular junction (NMJ), a synaptic connection between motor neurons and skeletal muscle fibers, is highly vulnerable in ALS and suggest that NMJ defects may occur at the early stages of the disease. However, mechanistic insight into how NMJ dysfunction relates to the onset and progression of ALS is incomplete, which hampers therapy development. This is, in part, caused by a lack of robust in vitro models. The ability to combine microfluidic and induced pluripotent stem cell (iPSC) technologies has opened up new avenues for studying molecular and cellular ALS phenotypes in vitro. Microfluidic devices offer several advantages over traditional culture approaches when modeling the NMJ, such as the spatial separation of different cell types and increased control over the cellular microenvironment. Moreover, they are compatible with 3D cell culture, which enhances NMJ functionality and maturity. Here, we review how microfluidic technology is currently being employed to develop more reliable in vitro NMJ models. To validate and phenotype such models, various morphological and functional read-outs have been developed. We describe and discuss the relevance of these read-outs and specifically illustrate how these read-outs have enhanced our understanding of NMJ pathology in ALS. Finally, we share our view on potential future directions and challenges.
Collapse
Affiliation(s)
- Rianne de Jongh
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Xandor M Spijkers
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.,Mimetas B.V., Organ-on-a-chip Company, Leiden, The Netherlands
| | - Svetlana Pasteuning-Vuhman
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Paul Vulto
- Mimetas B.V., Organ-on-a-chip Company, Leiden, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
21
|
Li A, Zhou J, Widelitz RB, Chow RH, Chuong CM. Integrating Bioelectrical Currents and Ca 2+ Signaling with Biochemical Signaling in Development and Pathogenesis. Bioelectricity 2020; 2:210-220. [PMID: 34476353 PMCID: PMC8370337 DOI: 10.1089/bioe.2020.0001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Roles of bioelectrical signals are increasingly recognized in excitable and nonexcitable non-neural tissues. Diverse ion-selective channels, pumps, and gap junctions participate in bioelectrical signaling, including those transporting calcium ions (Ca2+). Ca2+ is the most versatile transported ion, because it serves as an electrical charge carrier and a biochemical regulator for multiple molecular binding, enzyme, and transcription activities. We aspire to learn how bioelectrical signals crosstalk to biochemical/biomechanical signals. In this study, we review four recent studies showing how bioelectrical currents and Ca2+ signaling affect collective dermal cell migration during feather bud elongation, affect chondrogenic differentiation in limb development, couple with mechanical tension in aligning gut smooth muscle, and affect mitochondrial function and skeletal muscle atrophy. We observe bioelectrical signals involved in several developmental and pathological conditions in chickens and mice at multiple spatial scales: cellular, cellular collective, and subcellular. These examples inspire novel concept and approaches for future basic and translational studies.
Collapse
Affiliation(s)
- Ang Li
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas, USA
| | - Jingsong Zhou
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas, USA
| | - Randall B. Widelitz
- Department of Pathology and Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Robert H. Chow
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Cheng-Ming Chuong
- Department of Pathology and Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
22
|
Badu-Mensah A, Guo X, McAleer CW, Rumsey JW, Hickman JJ. Functional skeletal muscle model derived from SOD1-mutant ALS patient iPSCs recapitulates hallmarks of disease progression. Sci Rep 2020; 10:14302. [PMID: 32868812 PMCID: PMC7459299 DOI: 10.1038/s41598-020-70510-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Recent findings suggest a pathologic role of skeletal muscle in amyotrophic lateral sclerosis (ALS) onset and progression. However, the exact mechanism by which this occurs remains elusive due to limited human-based studies. To this end, phenotypic ALS skeletal muscle models were developed from induced pluripotent stem cells (iPSCs) derived from healthy individuals (WT) and ALS patients harboring mutations in the superoxide dismutase 1 (SOD1) gene. Although proliferative, SOD1 myoblasts demonstrated delayed and reduced fusion efficiency compared to WT. Additionally, SOD1 myotubes exhibited significantly reduced length and cross-section. Also, SOD1 myotubes had loosely arranged myosin heavy chain and reduced acetylcholine receptor expression per immunocytochemical analysis. Functional analysis indicated considerably reduced contractile force and synchrony in SOD1 myotubes. Mitochondrial assessment indicated reduced inner mitochondrial membrane potential (ΔΨm) and metabolic plasticity in the SOD1-iPSC derived myotubes. This work presents the first well-characterized in vitro iPSC-derived muscle model that demonstrates SOD1 toxicity effects on human muscle regeneration, contractility and metabolic function in ALS. Current findings align with previous ALS patient biopsy studies and suggest an active contribution of skeletal muscle in NMJ dysfunction. Further, the results validate this model as a human-relevant platform for ALS research and drug discovery studies.
Collapse
Affiliation(s)
- Agnes Badu-Mensah
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.,College of Medicine, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32816, USA
| | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | | | - John W Rumsey
- Hesperos Inc., 12501 Research Pkwy, Suite 100, Orlando, FL, 32826, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA. .,Hesperos Inc., 12501 Research Pkwy, Suite 100, Orlando, FL, 32826, USA.
| |
Collapse
|
23
|
Li S, Wu Z, Li Y, Tantray I, De Stefani D, Mattarei A, Krishnan G, Gao FB, Vogel H, Lu B. Altered MICOS Morphology and Mitochondrial Ion Homeostasis Contribute to Poly(GR) Toxicity Associated with C9-ALS/FTD. Cell Rep 2020; 32:107989. [PMID: 32755582 PMCID: PMC7433775 DOI: 10.1016/j.celrep.2020.107989] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 05/20/2020] [Accepted: 07/14/2020] [Indexed: 12/31/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) manifests pathological changes in motor neurons and various other cell types. Compared to motor neurons, the contribution of the other cell types to the ALS phenotypes is understudied. G4C2 repeat expansion in C9ORF72 is the most common genetic cause of ALS along with frontotemporal dementia (C9-ALS/FTD), with increasing evidence supporting repeat-encoded poly(GR) in disease pathogenesis. Here, we show in Drosophila muscle that poly(GR) enters mitochondria and interacts with components of the Mitochondrial Contact Site and Cristae Organizing System (MICOS), altering MICOS dynamics and intra-subunit interactions. This impairs mitochondrial inner membrane structure, ion homeostasis, mitochondrial metabolism, and muscle integrity. Similar mitochondrial defects are observed in patient fibroblasts. Genetic manipulation of MICOS components or pharmacological restoration of ion homeostasis with nigericin effectively rescue the mitochondrial pathology and disease phenotypes in both systems. These results implicate MICOS-regulated ion homeostasis in C9-ALS pathogenesis and suggest potential new therapeutic strategies.
Collapse
Affiliation(s)
- Shuangxi Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA,These authors contributed equally
| | - Zhihao Wu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA,These authors contributed equally,Present address: Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas TX 75275, USA
| | - Yu Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ishaq Tantray
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Diego De Stefani
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - Gopinath Krishnan
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA,Lead Contact,Correspondence:
| |
Collapse
|
24
|
McCombe PA, Garton FC, Katz M, Wray NR, Henderson RD. What do we know about the variability in survival of patients with amyotrophic lateral sclerosis? Expert Rev Neurother 2020; 20:921-941. [PMID: 32569484 DOI: 10.1080/14737175.2020.1785873] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION ALS is a fatal neurodegenerative disease. However, patients show variability in the length of survival after symptom onset. Understanding the mechanisms of long survival could lead to possible avenues for therapy. AREAS COVERED This review surveys the reported length of survival in ALS, the clinical features that predict survival in individual patients, and possible factors, particularly genetic factors, that could cause short or long survival. The authors also speculate on possible mechanisms. EXPERT OPINION a small number of known factors can explain some variability in ALS survival. However, other disease-modifying factors likely exist. Factors that alter motor neurone vulnerability and immune, metabolic, and muscle function could affect survival by modulating the disease process. Knowing these factors could lead to interventions to change the course of the disease. The authors suggest a broad approach is needed to quantify the proportion of variation survival attributable to genetic and non-genetic factors and to identify and estimate the effect size of specific factors. Studies of this nature could not only identify novel avenues for therapeutic research but also play an important role in clinical trial design and personalized medicine.
Collapse
Affiliation(s)
- Pamela A McCombe
- Centre for Clinical Research, The University of Queensland , Brisbane, Australia.,Department of Neurology, Royal Brisbane and Women's Hospital , Brisbane, Australia
| | - Fleur C Garton
- Institute for Molecular Biosciences, The University of Queensland , Brisbane, Australia
| | - Matthew Katz
- Department of Neurology, Royal Brisbane and Women's Hospital , Brisbane, Australia
| | - Naomi R Wray
- Institute for Molecular Biosciences, The University of Queensland , Brisbane, Australia.,Queensland Brain Institute, The University of Queensland , Brisbane, Australia
| | - Robert D Henderson
- Centre for Clinical Research, The University of Queensland , Brisbane, Australia
| |
Collapse
|
25
|
Gokuladhas S, Schierding W, Cameron-Smith D, Wake M, Scotter EL, O’Sullivan J. Shared Regulatory Pathways Reveal Novel Genetic Correlations Between Grip Strength and Neuromuscular Disorders. Front Genet 2020; 11:393. [PMID: 32391060 PMCID: PMC7194178 DOI: 10.3389/fgene.2020.00393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Muscle weakness is a common consequence of both aging (sarcopenia) and neuromuscular disorders (NMD). Whilst genome-wide association (GWA) studies have identified genetic variants associated with grip strength (GS; measure of muscle strength/weakness) and NMDs, including multiple sclerosis (MS), myasthenia gravis (MG) and amyotrophic lateral sclerosis (ALS), it is not known whether there are common mechanisms between these phenotypes. To examine this, we have integrated GS and NMD associated genetic variants (single nucleotide polymorphisms; SNPs) in a multimorbid analysis that leverages high-throughput chromatin interaction (Hi-C) data and expression quantitative trait loci data to identify target genes (i.e., SNP-mediated gene regulation). Biological pathways enriched by these genes were then identified using next-generation pathway enrichment analysis. Lastly, druggable genes were identified using drug gene interaction (DGI) database. We identified gene regulatory mechanisms associated with GS, MG, MS, and ALS. The SNPs associated with GS regulate a subset of genes that are also regulated by the SNPs of MS, MG, and ALS. Yet, we did not find any genes commonly regulated by all four phenotype associated SNPs. By contrast, we identified significant enrichment in three pathways (mTOR signaling, axon guidance, and alcoholism) that are commonly affected by the gene regulatory mechanisms associated with all four phenotypes. 13% of the genes we identified were known drug targets, and GS shares at least one druggable gene and pathway with each of the NMD phenotypes. We have identified significant biological overlaps between GS and NMD, demonstrating the potential for spatial genetic analysis to identify common mechanisms between potential multimorbid phenotypes. Collectively, our results form the foundation for a shift from a gene to a pathway-based approach to the rationale design of therapeutic interventions and treatments for NMD.
Collapse
Affiliation(s)
| | | | - David Cameron-Smith
- Liggins Institute, The University of Auckland, Auckland, New Zealand
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Melissa Wake
- Murdoch Children’s Research Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Emma L. Scotter
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Justin O’Sullivan
- Liggins Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
26
|
VAPB ER-Aggregates, A Possible New Biomarker in ALS Pathology. Cells 2020; 9:cells9010164. [PMID: 31936602 PMCID: PMC7017080 DOI: 10.3390/cells9010164] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/16/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022] Open
Abstract
A point mutation (P56S) in the gene-encoding vesicle-associated membrane-protein-associated protein B (VAPB) leads to an autosomal-dominant form of amyotrophic lateral sclerosis (ALS), classified as ALS-8. The mutant VAPB is characterized by ER-associated aggregates that lead to a complete reorganization of ER structures. Growing evidences suggest VAPB involvement in ALS pathomechanisms. In fact, numerous studies demonstrated VAPB alteration also in sporadic ALS (sALS) and showed the presence of its aggregates when others ALS-related gene are mutant. Recently, the identification of new biomarkers in peripheral blood mononuclear cells (PBMCs) has been proposed as a good noninvasive option for studying ALS. Here, we evaluated VAPB as a possible ALS pathologic marker analyzing PBMCs of sALS patients. Immunofluorescence analysis (IFA) showed a peculiar pattern of VAPB aggregates in sALS, not evident in healthy control (HC) subjects and in Parkinson’s disease (PD) PBMCs. This specific pattern led us to suppose that VAPB could be misfolded in sALS. The data indirectly confirmed by flow cytometry assay (FCA) showed a reduction of VAPB fluorescent signals in sALS. However, our observations were not associated with the presence of a genetic mutation or altered gene expression of VAPB. Our study brings further evidences of the VAPB role in ALS as a diagnostic biomarker.
Collapse
|
27
|
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
|
28
|
Rossaert E, Pollari E, Jaspers T, Van Helleputte L, Jarpe M, Van Damme P, De Bock K, Moisse M, Van Den Bosch L. Restoration of histone acetylation ameliorates disease and metabolic abnormalities in a FUS mouse model. Acta Neuropathol Commun 2019; 7:107. [PMID: 31277703 PMCID: PMC6612190 DOI: 10.1186/s40478-019-0750-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022] Open
Abstract
Dysregulation of epigenetic mechanisms is emerging as a central event in neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). In many models of neurodegeneration, global histone acetylation is decreased in the affected neuronal tissues. Histone acetylation is controlled by the antagonistic actions of two protein families -the histone acetyltransferases (HATs) and the histone deacetylases (HDACs). Drugs inhibiting HDAC activity are already used in the clinic as anti-cancer agents. The aim of this study was to explore the therapeutic potential of HDAC inhibition in the context of ALS. We discovered that transgenic mice overexpressing wild-type FUS ("Tg FUS+/+"), which recapitulate many aspects of human ALS, showed reduced global histone acetylation and alterations in metabolic gene expression, resulting in a dysregulated metabolic homeostasis. Chronic treatment of Tg FUS+/+ mice with ACY-738, a potent HDAC inhibitor that can cross the blood-brain barrier, ameliorated the motor phenotype and substantially extended the life span of the Tg FUS+/+ mice. At the molecular level, ACY-738 restored global histone acetylation and metabolic gene expression, thereby re-establishing metabolite levels in the spinal cord. Taken together, our findings link epigenetic alterations to metabolic dysregulation in ALS pathology, and highlight ACY-738 as a potential therapeutic strategy to treat this devastating disease.
Collapse
|
29
|
Role of Transforming Growth Factor-β in Skeletal Muscle Fibrosis: A Review. Int J Mol Sci 2019; 20:ijms20102446. [PMID: 31108916 PMCID: PMC6566291 DOI: 10.3390/ijms20102446] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/09/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor-beta (TGF-β) isoforms are cytokines involved in a variety of cellular processes, including myofiber repair and regulation of connective tissue formation. Activation of the TGF-β pathway contributes to pathologic fibrosis in most organs. Here, we have focused on examining the evidence demonstrating the involvement of TGF-β in the fibrosis of skeletal muscle particularly. The TGF-β pathway plays a role in different skeletal muscle myopathies, and TGF-β signaling is highly induced in these diseases. In this review, we discuss different molecular mechanisms of TGF-β-mediated skeletal muscle fibrosis and highlight different TGF-β-targeted treatments that target these relevant pathways.
Collapse
|
30
|
Mulbauer GD, Matthew HW. Biomimetic Scaffolds in Skeletal Muscle Regeneration. Discoveries (Craiova) 2019; 7:e90. [PMID: 32309608 PMCID: PMC7086065 DOI: 10.15190/d.2019.3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 03/31/2019] [Accepted: 03/31/2019] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle tissue has inherent capacity for regeneration in response to minor injuries. However, in the case of severe trauma, tumor ablations, or in congenital muscle defects, these myopathies can cause irreversible loss of muscle mass and function, a condition referred to as volumetric muscle loss (VML). The natural muscle repair mechanisms are overwhelmed, prompting the search for new muscle regenerative strategies, such as using biomaterials that can provide regenerative signals to either transplanted or host muscle cells. Recent studies involve the use of suitable biomaterials which may be utilized as a template to guide tissue reorganization and ultimately provide optimum micro-environmental conditions to cells. These strategies range from approaches that utilize biomaterials alone to those that combine materials with exogenous growth factors, and ex vivo cultured cells. A number of scaffold materials have been used in the development of grafts to treat VML. In this brief review, we outline the natural skeletal regeneration process, available treatments used in the clinic for muscle injury and promising tissue bioengineering and regenerative approaches for muscle loss treatment.
Collapse
Affiliation(s)
- Greta D. Mulbauer
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Howard W.T. Matthew
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| |
Collapse
|
31
|
Gulino R, Vicario N, Giunta MAS, Spoto G, Calabrese G, Vecchio M, Gulisano M, Leanza G, Parenti R. Neuromuscular Plasticity in a Mouse Neurotoxic Model of Spinal Motoneuronal Loss. Int J Mol Sci 2019; 20:ijms20061500. [PMID: 30917493 PMCID: PMC6471664 DOI: 10.3390/ijms20061500] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/19/2019] [Accepted: 03/24/2019] [Indexed: 12/11/2022] Open
Abstract
Despite the relevant research efforts, the causes of amyotrophic lateral sclerosis (ALS) are still unknown and no effective cure is available. Many authors suggest that ALS is a multi-system disease caused by a network failure instead of a cell-autonomous pathology restricted to motoneurons. Although motoneuronal loss is the critical hallmark of ALS given their specific vulnerability, other cell populations, including muscle and glial cells, are involved in disease onset and progression, but unraveling their specific role and crosstalk requires further investigation. In particular, little is known about the plastic changes of the degenerating motor system. These spontaneous compensatory processes are unable to halt the disease progression, but their elucidation and possible use as a therapeutic target represents an important aim of ALS research. Genetic animal models of disease represent useful tools to validate proven hypotheses or to test potential therapies, and the conception of novel hypotheses about ALS causes or the study of pathogenic mechanisms may be advantaged by the use of relatively simple in vivo models recapitulating specific aspects of the disease, thus avoiding the inclusion of too many confounding factors in an experimental setting. Here, we used a neurotoxic model of spinal motoneuron depletion induced by injection of cholera toxin-B saporin in the gastrocnemius muscle to investigate the possible occurrence of compensatory changes in both the muscle and spinal cord. The results showed that, following the lesion, the skeletal muscle became atrophic and displayed electromyographic activity similar to that observed in ALS patients. Moreover, the changes in muscle fiber morphology were different from that observed in ALS models, thus suggesting that some muscular effects of disease may be primary effects instead of being simply caused by denervation. Notably, we found plastic changes in the surviving motoneurons that can produce a functional restoration probably similar to the compensatory changes occurring in disease. These changes could be at least partially driven by glutamatergic signaling, and astrocytes contacting the surviving motoneurons may support this process.
Collapse
Affiliation(s)
- Rosario Gulino
- Laboratory of Neurophysiology, Department of Biomedical and Biotechnological Science, Section of Physiology, University of Catania, Catania 95123, Italy.
| | - Nunzio Vicario
- Laboratory of Cellular and Molecular Physiology, Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania 95123, Italy.
| | - Maria A S Giunta
- Laboratory of Cellular and Molecular Physiology, Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania 95123, Italy.
| | - Graziana Spoto
- Laboratory of Cellular and Molecular Physiology, Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania 95123, Italy.
| | - Giovanna Calabrese
- Laboratory of Cellular and Molecular Physiology, Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania 95123, Italy.
| | - Michele Vecchio
- Rehabilitation Unit, "AOU Policlinico Vittorio Emanuele" and Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania 95123, Italy.
| | - Massimo Gulisano
- Laboratory of Synthetic and Systems Biology, Department of Drug Sciences, University of Catania, Catania 95125, Italy.
| | - Giampiero Leanza
- Laboratory of Neurogenesis and Repair, Department of Drug Sciences, University of Catania, Catania 95125, Italy.
| | - Rosalba Parenti
- Laboratory of Cellular and Molecular Physiology, Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania 95123, Italy.
| |
Collapse
|
32
|
Zhou J, Li A, Li X, Yi J. Dysregulated mitochondrial Ca 2+ and ROS signaling in skeletal muscle of ALS mouse model. Arch Biochem Biophys 2019; 663:249-258. [PMID: 30682329 PMCID: PMC6506190 DOI: 10.1016/j.abb.2019.01.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/24/2018] [Accepted: 01/18/2019] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neuromuscular disease characterized by motor neuron loss and prominent skeletal muscle wasting. Despite more than one hundred years of research efforts, the pathogenic mechanisms underlying neuromuscular degeneration in ALS remain elusive. While the death of motor neuron is a defining hallmark of ALS, accumulated evidences suggested that in addition to being a victim of motor neuron axonal withdrawal, the intrinsic skeletal muscle degeneration may also actively contribute to ALS disease pathogenesis and progression. Examination of spinal cord and muscle autopsy/biopsy samples of ALS patients revealed similar mitochondrial abnormalities in morphology, quantity and disposition, which are accompanied by defective mitochondrial respiratory chain complex and elevated oxidative stress. Detailing the molecular/cellular mechanisms and the role of mitochondrial dysfunction in ALS relies on ALS animal model studies. This review article discusses the dysregulated mitochondrial Ca2+ and reactive oxygen species (ROS) signaling revealed in live skeletal muscle derived from ALS mouse models, and a potential role of the vicious cycle formed between the dysregulated mitochondrial Ca2+ signaling and excessive ROS production in promoting muscle wasting during ALS progression.
Collapse
Affiliation(s)
- Jingsong Zhou
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA.
| | - Ang Li
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Xuejun Li
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Jianxun Yi
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA.
| |
Collapse
|
33
|
Walker CL, Meadows RM, Merfeld-Clauss S, Du Y, March KL, Jones KJ. Adipose-derived stem cell conditioned medium impacts asymptomatic peripheral neuromuscular denervation in the mutant superoxide dismutase (G93A) transgenic mouse model of amyotrophic lateral sclerosis. Restor Neurol Neurosci 2018; 36:621-627. [PMID: 30010155 DOI: 10.3233/rnn-180820] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is devastating, leading to paralysis and death. Disease onset begins pre-symptomatically through spinal motor neuron (MN) axon die-back from musculature at ∼47 days of age in the mutant superoxide dismutase 1 (mSOD1G93A) transgenic ALS mouse model. This period may be optimal to assess potential therapies. We previously demonstrated that post-symptomatic adipose-derived stem cell conditioned medium (ASC-CM) treatment is neuroprotective in mSOD1G93A mice. We hypothesized that early disease onset treatment could ameliorate neuromuscular junction (NMJ) disruption. OBJECTIVE To determine whether pre-symptom administration of ASC-CM prevents early NMJ disconnection. METHODS We confirmed the NMJ denervation time course in mSOD1G93A mice using co-labeling of neurofilament and post-synaptic acetylcholine receptors (AchR) by α-bungarotoxin. We determined whether ASC-CM ameliorates early NMJ loss in mSOD1G93A mice by systemically administering 200μl ASC-CM or vehicle medium daily from post-natal days 35 to 47 and quantifying intact NMJs through co-labeling of neurofilament and synaptophysin with α-bungarotoxin in gastrocnemius muscle. RESULTS Intact NMJs were significantly decreased in 47 day old mSOD1G93A mice (p < 0.05), and daily systemic ASC-CM prevented disease-induced NMJ denervation compared to vehicle treated mice (p < 0.05). CONCLUSIONS Our results lay the foundation for testing the long-term neurological benefits of systemic ASC-CM therapy in the mSOD1G93A mouse model of ALS.
Collapse
Affiliation(s)
- Chandler L Walker
- Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, USA.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Rena M Meadows
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Stephanie Merfeld-Clauss
- Roudebush VA Medical Center, Indianapolis, IN, USA.,Division of Cardiovascular Medicine, Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Yansheng Du
- Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Keith L March
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA.,Division of Cardiovascular Medicine, Center for Regenerative Medicine, University of Florida, Gainesville, FL, USA
| | - Kathryn J Jones
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Roudebush VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
34
|
Bereman MS, Beri J, Enders JR, Nash T. Machine Learning Reveals Protein Signatures in CSF and Plasma Fluids of Clinical Value for ALS. Sci Rep 2018; 8:16334. [PMID: 30397248 PMCID: PMC6218542 DOI: 10.1038/s41598-018-34642-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/23/2018] [Indexed: 11/14/2022] Open
Abstract
We use shotgun proteomics to identify biomarkers of diagnostic and prognostic value in individuals diagnosed with amyotrophic lateral sclerosis. Matched cerebrospinal and plasma fluids were subjected to abundant protein depletion and analyzed by nano-flow liquid chromatography high resolution tandem mass spectrometry. Label free quantitation was used to identify differential proteins between individuals with ALS (n = 33) and healthy controls (n = 30) in both fluids. In CSF, 118 (p-value < 0.05) and 27 proteins (q-value < 0.05) were identified as significantly altered between ALS and controls. In plasma, 20 (p-value < 0.05) and 0 (q-value < 0.05) proteins were identified as significantly altered between ALS and controls. Proteins involved in complement activation, acute phase response and retinoid signaling pathways were significantly enriched in the CSF from ALS patients. Subsequently various machine learning methods were evaluated for disease classification using a repeated Monte Carlo cross-validation approach. A linear discriminant analysis model achieved a median area under the receiver operating characteristic curve of 0.94 with an interquartile range of 0.88–1.0. Three proteins composed a prognostic model (p = 5e-4) that explained 49% of the variation in the ALS-FRS scores. Finally we investigated the specificity of two promising proteins from our discovery data set, chitinase-3 like 1 protein and alpha-1-antichymotrypsin, using targeted proteomics in a separate set of CSF samples derived from individuals diagnosed with ALS (n = 11) and other neurological diseases (n = 15). These results demonstrate the potential of a panel of targeted proteins for objective measurements of clinical value in ALS.
Collapse
Affiliation(s)
- Michael S Bereman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, 27695, USA. .,Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA. .,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA.
| | - Joshua Beri
- Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Jeffrey R Enders
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
| | - Tara Nash
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
| |
Collapse
|
35
|
Bojungikgi-tang Improves Muscle and Spinal Cord Function in an Amyotrophic Lateral Sclerosis Model. Mol Neurobiol 2018; 56:2394-2407. [PMID: 30030751 DOI: 10.1007/s12035-018-1236-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a motor neuron disease characterized by progressive motor function impairment, dysphagia, and respiratory failure. Owing to the complexity of its pathogenic mechanisms, an effective therapy for ALS is lacking. Herbal medicines with multiple targets have good efficacy and low adverse reactions for the treatment of neurodegenerative diseases. In this study, the effects of Bojungikgi-tang (BJIGT), an herbal medicine with eight component herbs, on muscle and spinal cord function were evaluated in an ALS animal model. Animals were randomly divided into three groups: a non-transgenic group (nTg, n = 24), a hSOD1G93A transgenic group (Tg, n = 24), and a hSOD1G93A transgenic group in which 8-week-old mice were orally administered BJIGT (1 mg/g) once daily for 6 weeks (Tg+BJIGT, n = 24). The effects of BJIGT were evaluated using a rotarod test, foot-printing, and survival analyses based on Kaplan-Meier survival curves. To determine the biological mechanism underlying the effects of BJIGT in hSOD1G93A mice, western blotting, transmission electron microscopy, and Bungarotoxin staining were used. BJIGT improved motor function and extended the survival duration of hSOD1G93A mice. In addition, BJIGT had protective effects, including anti-oxidative and anti-inflammatory effects, in both the spinal cord and muscle of hSOD1G93A mice. Our results demonstrated that BJIGT causes muscle atrophy and the denervation of neuromuscular junctions in the gastrocnemius of hSOD1G93A mice. The components of BJIGT may alleviate the symptoms of ALS via different mechanisms, and accordingly, BJIGT treatment may be an effective therapeutic approach.
Collapse
|
36
|
Gonzalez D, Rebolledo DL, Correa LM, Court FA, Cerpa W, Lipson KE, van Zundert B, Brandan E. The inhibition of CTGF/CCN2 activity improves muscle and locomotor function in a murine ALS model. Hum Mol Genet 2018; 27:2913-2926. [DOI: 10.1093/hmg/ddy204] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/17/2018] [Indexed: 02/06/2023] Open
Affiliation(s)
- David Gonzalez
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela L Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Lina M Correa
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe A Court
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Waldo Cerpa
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Brigitte van Zundert
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile
- Centro de Investigaciones Biomédicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
37
|
Lehmann S, Esch E, Hartmann P, Goswami A, Nikolin S, Weis J, Beyer C, Johann S. Expression profile of pattern recognition receptors in skeletal muscle of SOD1 (G93A) amyotrophic lateral sclerosis (ALS) mice and sporadic ALS patients. Neuropathol Appl Neurobiol 2018; 44:606-627. [PMID: 29575052 DOI: 10.1111/nan.12483] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 02/20/2018] [Indexed: 12/14/2022]
Abstract
AIMS Amyotrophic lateral sclerosis (ALS) is characterized by degeneration of motoneurons and progressive muscle wasting. Inflammatory processes, mediated by non-neuronal cells, such as glial cells, are known to contribute to disease progression. Inflammasomes consist of pattern recognition receptors (PRRs), apoptosis-associated speck-like protein (ASC) and caspase 1 and are essential for interleukin (IL) processing and a rapid immune response after tissue damage. Recently, we described inflammasome activation in the spinal cord of ALS patients and in SOD1(G93A) ALS mice. Since pathological changes in the skeletal muscle are early events in ALS, we hypothesized that PRRs might be abnormally expressed in muscle fibre degeneration. METHODS Western blot analysis, real-time PCR and immunohistochemistry were performed with muscle tissue from presymptomatic and early-symptomatic male SOD1(G93A) mice and with muscle biopsies of control and sporadic ALS (sALS) patients. Analysed PRRs include nucleotide-binding oligomerization domain-like (NOD-like) receptor protein 1 (NLRP1), NLR protein 3 (NLRP3), NLR family CARD domain-containing 4 (NLRC4) and absent in melanoma 2. Additionally, expression levels of ASC, caspase 1, interleukin 1 beta (IL1β) and interleukin 18 (IL18) were evaluated. RESULTS Expression of PRRs and ASC was detected in murine and human tissue. The PRR NLRC4, caspase 1 and IL1β were significantly elevated in denervated muscle of SOD1(G93A) mice and sALS patients. Furthermore, levels of caspase 1 and IL1β were already increased in presymptomatic animals. CONCLUSION Our findings suggest that increased inflammasome activation may be involved in skeletal muscle pathology in ALS. Furthermore, elevated levels of NLRC4, caspase 1 and IL1β reflect early changes in the skeletal muscle and may contribute to the denervation process.
Collapse
Affiliation(s)
- S Lehmann
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany.,Institute Molecular and Cellular Anatomy (MOCA), Medical Clinic RWTH Aachen University, Aachen, Germany
| | - E Esch
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - P Hartmann
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - A Goswami
- Institute of Neuropathology, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - S Nikolin
- Institute of Neuropathology, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - J Weis
- Institute of Neuropathology, Medical Clinic RWTH Aachen University, Aachen, Germany
| | - C Beyer
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany.,JARA - Translational Brain Medicine, Aachen, Germany
| | - S Johann
- Institute of Neuroanatomy, Medical Clinic RWTH Aachen University, Aachen, Germany.,Institute of Anatomy II, Medical Faculty Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
38
|
Current Methods for Skeletal Muscle Tissue Repair and Regeneration. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1984879. [PMID: 29850487 PMCID: PMC5926523 DOI: 10.1155/2018/1984879] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/28/2018] [Accepted: 03/11/2018] [Indexed: 12/11/2022]
Abstract
Skeletal muscle has the capacity of regeneration after injury. However, for large volumes of muscle loss, this regeneration needs interventional support. Consequently, muscle injury provides an ongoing reconstructive and regenerative challenge in clinical work. To promote muscle repair and regeneration, different strategies have been developed within the last century and especially during the last few decades, including surgical techniques, physical therapy, biomaterials, and muscular tissue engineering as well as cell therapy. Still, there is a great need to develop new methods and materials, which promote skeletal muscle repair and functional regeneration. In this review, we give a comprehensive overview over the epidemiology of muscle tissue loss, highlight current strategies in clinical treatment, and discuss novel methods for muscle regeneration and challenges for their future clinical translation.
Collapse
|
39
|
Miyoshi S, Tezuka T, Arimura S, Tomono T, Okada T, Yamanashi Y. DOK7 gene therapy enhances motor activity and life span in ALS model mice. EMBO Mol Med 2018; 9:880-889. [PMID: 28490573 PMCID: PMC5494517 DOI: 10.15252/emmm.201607298] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive, multifactorial motor neurodegenerative disease with severe muscle atrophy. The glutamate release inhibitor riluzole is the only medication approved by the FDA, and prolongs patient life span by a few months, testifying to a strong need for new treatment strategies. In ALS, motor neuron degeneration first becomes evident at the motor nerve terminals in neuromuscular junctions (NMJs), the cholinergic synapse between motor neuron and skeletal muscle; degeneration then progresses proximally, implicating the NMJ as a therapeutic target. We previously demonstrated that activation of muscle‐specific kinase MuSK by the cytoplasmic protein Dok‐7 is essential for NMJ formation, and forced expression of Dok‐7 in muscle activates MuSK and enlarges NMJs. Here, we show that therapeutic administration of an adeno‐associated virus vector encoding the human DOK7 gene suppressed motor nerve terminal degeneration at NMJs together with muscle atrophy in the SOD1‐G93A ALS mouse model. Ultimately, we show that DOK7 gene therapy enhanced motor activity and life span in ALS model mice.
Collapse
Affiliation(s)
- Sadanori Miyoshi
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tohru Tezuka
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sumimasa Arimura
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Taro Tomono
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan.,Graduate School of Comprehensive Human Sciences, Majors in Medical Sciences University of Tsukuba, Ibaraki, Japan
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Yuji Yamanashi
- Division of Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
40
|
Vitamin D and Neurological Diseases: An Endocrine View. Int J Mol Sci 2017; 18:ijms18112482. [PMID: 29160835 PMCID: PMC5713448 DOI: 10.3390/ijms18112482] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 11/18/2017] [Accepted: 11/20/2017] [Indexed: 02/07/2023] Open
Abstract
Vitamin D system comprises hormone precursors, active metabolites, carriers, enzymes, and receptors involved in genomic and non-genomic effects. In addition to classical bone-related effects, this system has also been shown to activate multiple molecular mediators and elicit many physiological functions. In vitro and in vivo studies have, in fact, increasingly focused on the "non-calcemic" actions of vitamin D, which are associated with the maintenance of glucose homeostasis, cardiovascular morbidity, autoimmunity, inflammation, and cancer. In parallel, growing evidence has recognized that a multimodal association links vitamin D system to brain development, functions and diseases. With vitamin D deficiency reaching epidemic proportions worldwide, there is now concern that optimal levels of vitamin D in the bloodstream are also necessary to preserve the neurological development and protect the adult brain. The aim of this review is to highlight the relationship between vitamin D and neurological diseases.
Collapse
|
41
|
Cappello V, Francolini M. Neuromuscular Junction Dismantling in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2017; 18:ijms18102092. [PMID: 28972545 PMCID: PMC5666774 DOI: 10.3390/ijms18102092] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/25/2017] [Accepted: 09/28/2017] [Indexed: 12/13/2022] Open
Abstract
Neuromuscular junction assembly and plasticity during embryonic, postnatal, and adult life are tightly regulated by the continuous cross-talk among motor nerve endings, muscle fibers, and glial cells. Altered communications among these components is thought to be responsible for the physiological age-related changes at this synapse and possibly for its destruction in pathological states. Neuromuscular junction dismantling plays a crucial role in the onset of Amyotrophic Lateral Sclerosis (ALS). ALS is characterized by the degeneration and death of motor neurons leading to skeletal muscle denervation, atrophy and, most often, death of the patient within five years from diagnosis. ALS is a non-cell autonomous disease as, besides motor neuron degeneration, glial cells, and possibly muscle fibers, play a role in its onset and progression. Here, we will review the recent literature regarding the mechanisms leading to neuromuscular junction disassembly and muscle denervation focusing on the role of the three players of this peripheral tripartite synapse.
Collapse
Affiliation(s)
- Valentina Cappello
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia Piazza San Silvestro 12, 56127 Pisa, Italy.
| | - Maura Francolini
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano-Via Vanvitelli 32, 20129 Milano, Italy.
| |
Collapse
|
42
|
Hu K, Bounni F, Williams Z. Editorial. Advancement in brain-machine interfaces for patients with tetraplegia: neurosurgical perspective. Neurosurg Focus 2017; 43:E5. [PMID: 28669301 DOI: 10.3171/2017.5.focus17244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Kejia Hu
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and.,Department of Microsurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Firas Bounni
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Ziv Williams
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
43
|
Gonzalez D, Contreras O, Rebolledo DL, Espinoza JP, van Zundert B, Brandan E. ALS skeletal muscle shows enhanced TGF-β signaling, fibrosis and induction of fibro/adipogenic progenitor markers. PLoS One 2017; 12:e0177649. [PMID: 28520806 PMCID: PMC5433732 DOI: 10.1371/journal.pone.0177649] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 04/30/2017] [Indexed: 02/06/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease in which upper and lower motoneurons degenerate leading to muscle wasting, paralysis and eventually death from respiratory failure. Several studies indicate that skeletal muscle contributes to disease progression; however the molecular mechanisms remain elusive. Fibrosis is a common feature in skeletal muscle under chronic damage conditions such as those caused by muscular dystrophies or denervation. However, the exact mechanisms of fibrosis induction and the cellular bases of this pathological response are unknown. We show that extracellular matrix (ECM) components are augmented in skeletal muscles of symptomatic hSOD1G93A mice, a widely used murine model of ALS. These mice also show increased TGF-β1 mRNA levels, total Smad3 protein levels and p-Smad3 positive nuclei. Furthermore, platelet-derived growth factor receptor-α (PDGFRα), Tcf4 and α-smooth muscle actin (α-SMA) levels are augmented in the skeletal muscle of symptomatic hSOD1G93A mice. Additionally, the fibro/adipogenic progenitors (FAPs), which are the main producers of ECM constituents, are also increased in these pathogenic conditions. Therefore, FAPs and ECM components are more abundant in symptomatic stages of the disease than in pre-symptomatic stages. We present evidence that fibrosis observed in skeletal muscle of symptomatic hSOD1G93A mice is accompanied with an induction of TGF-β signaling, and also that FAPs might be involved in triggering a fibrotic response. Co-localization of p-Smad3 positive cells together with PDGFRα was observed in the interstitial cells of skeletal muscles from symptomatic hSOD1G93A mice. Finally, the targeting of pro-fibrotic factors such as TGF-β, CTGF/CCN2 and platelet-derived growth factor (PDGF) signaling pathway might be a suitable therapeutic approach to improve muscle function in several degenerative diseases.
Collapse
Affiliation(s)
- David Gonzalez
- Centro de Envejecimiento y Regeneración, CARE Chile UC y Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Osvaldo Contreras
- Centro de Envejecimiento y Regeneración, CARE Chile UC y Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela L. Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC y Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Pablo Espinoza
- Centro de Envejecimiento y Regeneración, CARE Chile UC y Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Brigitte van Zundert
- Centro de Investigaciones Biomédicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andres Bello, Santiago, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC y Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
44
|
Peggion C, Massimino ML, Biancotto G, Angeletti R, Reggiani C, Sorgato MC, Bertoli A, Stella R. Absolute quantification of myosin heavy chain isoforms by selected reaction monitoring can underscore skeletal muscle changes in a mouse model of amyotrophic lateral sclerosis. Anal Bioanal Chem 2017; 409:2143-2153. [PMID: 28078418 DOI: 10.1007/s00216-016-0160-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/05/2016] [Accepted: 12/15/2016] [Indexed: 01/08/2023]
Abstract
Skeletal muscle fibers contain different isoforms of myosin heavy chain (MyHC) that define distinctive contractile properties. In light of the muscle capacity to adapt MyHC expression to pathophysiological conditions, a rapid and quantitative assessment of MyHC isoforms in small muscle tissue quantities would represent a valuable diagnostic tool for (neuro)muscular diseases. As past protocols did not meet these requirements, in the present study we applied a targeted proteomic approach based on selected reaction monitoring that allowed the absolute quantification of slow and fast MyHC isoforms in different mouse skeletal muscles with high reproducibility. This mass-spectrometry-based method was validated also in a pathological specimen, by comparison of the MyHC expression profiles in different muscles from healthy mice and a genetic mouse model of amyotrophic lateral sclerosis (ALS) expressing the SOD1(G93A) mutant. This analysis showed that terminally ill ALS mice have a fast-to-slow shift in the fiber type composition of the tibialis anterior and gastrocnemius muscles, as previously reported. These results will likely open the way to accurate and rapid diagnoses of human (neuro)muscular diseases by the proposed method. Graphical Abstract Methods for myosin heavy chain (MyHC) quantification: a comparison of classical methods and selected reaction monitoring (SRM)-based mass spectrometry approaches.
Collapse
Affiliation(s)
- Caterina Peggion
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58/B, 35131, Padua, PD, Italy
| | - Maria Lina Massimino
- CNR Neuroscience Institute, University of Padua, Via Ugo Bassi 58/B, 35131, Padua, PD, Italy
| | - Giancarlo Biancotto
- Department of Chemistry, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università, 10, 35020, Legnaro, PD, Italy
| | - Roberto Angeletti
- Department of Chemistry, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università, 10, 35020, Legnaro, PD, Italy
| | - Carlo Reggiani
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58/B, 35131, Padua, PD, Italy
| | - Maria Catia Sorgato
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58/B, 35131, Padua, PD, Italy.,CNR Neuroscience Institute, University of Padua, Via Ugo Bassi 58/B, 35131, Padua, PD, Italy
| | - Alessandro Bertoli
- Department of Biomedical Sciences, University of Padua, Via Ugo Bassi 58/B, 35131, Padua, PD, Italy.
| | - Roberto Stella
- Department of Chemistry, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università, 10, 35020, Legnaro, PD, Italy.
| |
Collapse
|
45
|
Greising SM, Dearth CL, Corona BT. Regenerative and Rehabilitative Medicine: A Necessary Synergy for Functional Recovery from Volumetric Muscle Loss Injury. Cells Tissues Organs 2016; 202:237-249. [PMID: 27825146 DOI: 10.1159/000444673] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2016] [Indexed: 01/04/2023] Open
Abstract
Volumetric muscle loss (VML) is a complex and heterogeneous problem due to significant traumatic or surgical loss of skeletal muscle tissue. The consequences of VML are substantial functional deficits in joint range of motion and skeletal muscle strength, resulting in life-long dysfunction and disability. Traditional physical medicine and rehabilitation paradigms do not address the magnitude of force loss due to VML and related musculoskeletal comorbidities. Recent advancements in regenerative medicine have set forth encouraging and emerging therapeutic options for VML injuries. There is significant potential that combined rehabilitative and regenerative therapies can restore limb and muscle function following VML injury in a synergistic manner. This review presents the current state of the VML field, spanning clinical and preclinical literature, with particular focus on rehabilitation and regenerative medicine in addition to their synergy. Moving forward, multidisciplinary collaboration between clinical and research fields is encouraged in order to continue to improve the treatment of VML injuries and specifically address the encompassing physiology, pathology, and specific needs of this patient population. This is a work of the US Government and is not subject to copyright protection in the USA. Foreign copyrights may apply. Published by S. Karger AG, Basel.
Collapse
|
46
|
A Novel Iron Chelator-Radical Scavenger Ameliorates Motor Dysfunction and Improves Life Span and Mitochondrial Biogenesis in SOD1G93A ALS Mice. Neurotox Res 2016; 31:230-244. [DOI: 10.1007/s12640-016-9677-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/05/2016] [Accepted: 10/13/2016] [Indexed: 12/12/2022]
|
47
|
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) or motor neuron disease is a rapidly progressive neurodegenerative disorder. The primary involvement is of motor neurons in the brain, spinal cord and peripherally. There is secondary weakness of muscles and primary involvement of other brain regions, especially involving cognition. SOURCES OF DATA Peer-reviewed journal articles and reviews. PubMed.gov AREAS OF AGREEMENT The pathogenesis of ALS remains largely unknown. There are a wide range of potential mechanisms related to neurodegeneration. An increasing number of genetic factors are recognized. AREAS OF CONTROVERSY There remains controversy, or lack of knowledge, in explaining how cellular events manifest as the complex human disease. There is controversy as to how well cellular and animal models of disease relate to the human disease. GROWING POINTS Large-scale international collaborative genetic epidemiological studies are replacing local studies. Therapies related to pathogenesis remain elusive, with the greatest advances to date relating to provision of care (including multidisciplinary management) and supportive care (nutrition and respiratory support). AREAS TIMELY FOR DEVELOPING RESEARCH The identification of C9orf72 hexanucleotide repeats as the most frequent genetic background to ALS, and the association with frontotemporal dementia, gives the potential of a genetic background against which to study other risk factors, triggers and pathogenic mechanisms, and to develop potential therapies.
Collapse
Affiliation(s)
- Sarah Morgan
- Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Richard W Orrell
- Department of Clinical Neuroscience, UCL Institute of Neurology, Rowland Hill Street, London NW3 2PF, UK
| |
Collapse
|
48
|
Beqollari D, Romberg CF, Dobrowolny G, Martini M, Voss AA, Musarò A, Bannister RA. Progressive impairment of CaV1.1 function in the skeletal muscle of mice expressing a mutant type 1 Cu/Zn superoxide dismutase (G93A) linked to amyotrophic lateral sclerosis. Skelet Muscle 2016; 6:24. [PMID: 27340545 PMCID: PMC4918102 DOI: 10.1186/s13395-016-0094-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 06/03/2016] [Indexed: 11/24/2022] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disorder that is typically fatal within 3–5 years of diagnosis. While motoneuron death is the defining characteristic of ALS, the events that underlie its pathology are not restricted to the nervous system. In this regard, ALS muscle atrophies and weakens significantly before presentation of neurological symptoms. Since the skeletal muscle L-type Ca2+ channel (CaV1.1) is a key regulator of both mass and force, we investigated whether CaV1.1 function is impaired in the muscle of two distinct mouse models carrying an ALS-linked mutation. Methods We recorded L-type currents, charge movements, and myoplasmic Ca2+ transients from dissociated flexor digitorum brevis (FDB) fibers to assess CaV1.1 function in two mouse models expressing a type 1 Cu/Zn superoxide dismutase mutant (SOD1G93A). Results In FDB fibers obtained from “symptomatic” global SOD1G93A mice, we observed a substantial reduction of SR Ca2+ release in response to depolarization relative to fibers harvested from age-matched control mice. L-type current and charge movement were both reduced by ~40 % in symptomatic SOD1G93A fibers when compared to control fibers. Ca2+ transients were not significantly reduced in similar experiments performed with FDB fibers obtained from “early-symptomatic” SOD1G93A mice, but L-type current and charge movement were decreased (~30 and ~20 %, respectively). Reductions in SR Ca2+ release (~35 %), L-type current (~20 %), and charge movement (~15 %) were also observed in fibers obtained from another model where SOD1G93A expression was restricted to skeletal muscle. Conclusions We report reductions in EC coupling, L-type current density, and charge movement in FDB fibers obtained from symptomatic global SOD1G93A mice. Experiments performed with FDB fibers obtained from early-symptomatic SOD1G93A and skeletal muscle autonomous MLC/SOD1G93A mice support the idea that events occurring locally in the skeletal muscle contribute to the impairment of CaV1.1 function in ALS muscle independently of innervation status. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0094-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Donald Beqollari
- Department of Medicine-Cardiology Division, University of Colorado School of Medicine, 12700 East 19th Avenue, B-139, Aurora, CO 80045 USA
| | - Christin F Romberg
- Department of Medicine-Cardiology Division, University of Colorado School of Medicine, 12700 East 19th Avenue, B-139, Aurora, CO 80045 USA
| | - Gabriella Dobrowolny
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, La Sapienza University, Via A. Scarpa, 14, 00161 Rome, Italy ; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Martina Martini
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, La Sapienza University, Via A. Scarpa, 14, 00161 Rome, Italy ; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Andrew A Voss
- Department of Biological Sciences, College of Science and Mathematics, Wright State University, 235A Biological Sciences, 3640 Colonel Glenn Highway, Dayton, OH 45435 USA
| | - Antonio Musarò
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, La Sapienza University, Via A. Scarpa, 14, 00161 Rome, Italy ; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Roger A Bannister
- Department of Medicine-Cardiology Division, University of Colorado School of Medicine, 12700 East 19th Avenue, B-139, Aurora, CO 80045 USA
| |
Collapse
|
49
|
von Grabowiecki Y, Abreu P, Blanchard O, Palamiuc L, Benosman S, Mériaux S, Devignot V, Gross I, Mellitzer G, Gonzalez de Aguilar JL, Gaiddon C. Transcriptional activator TAp63 is upregulated in muscular atrophy during ALS and induces the pro-atrophic ubiquitin ligase Trim63. eLife 2016; 5. [PMID: 26919175 PMCID: PMC4786414 DOI: 10.7554/elife.10528] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/08/2016] [Indexed: 12/14/2022] Open
Abstract
Mechanisms of muscle atrophy are complex and their understanding might help finding therapeutic solutions for pathologies such as amyotrophic lateral sclerosis (ALS). We meta-analyzed transcriptomic experiments of muscles of ALS patients and mouse models, uncovering a p53 deregulation as common denominator. We then characterized the induction of several p53 family members (p53, p63, p73) and a correlation between the levels of p53 family target genes and the severity of muscle atrophy in ALS patients and mice. In particular, we observed increased p63 protein levels in the fibers of atrophic muscles via denervation-dependent and -independent mechanisms. At a functional level, we demonstrated that TAp63 and p53 transactivate the promoter and increased the expression of Trim63 (MuRF1), an effector of muscle atrophy. Altogether, these results suggest a novel function for p63 as a contributor to muscular atrophic processes via the regulation of multiple genes, including the muscle atrophy gene Trim63.
Collapse
Affiliation(s)
- Yannick von Grabowiecki
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Paula Abreu
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Orphee Blanchard
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Lavinia Palamiuc
- Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France.,Sanford Burnham Medical Research Institute, San Diego, United States
| | - Samir Benosman
- Sanford Burnham Medical Research Institute, San Diego, United States
| | - Sophie Mériaux
- Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France.,Sanford Burnham Medical Research Institute, San Diego, United States
| | - Véronique Devignot
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Isabelle Gross
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - Georg Mellitzer
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| | - José L Gonzalez de Aguilar
- Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France.,Institut national de la santé et de la recherche médicale, Laboratoire SMN, Strasbourg, France
| | - Christian Gaiddon
- UMR_S 1113, Molecular mechanisms of stress response and pathologies, Institut national de la santé et de la recherche médicale, Strasbourg, France.,Fédération de Recherche Translationnelle, Strasbourg University, Strasbourg, France
| |
Collapse
|
50
|
Chen KS, Sakowski SA, Feldman EL. Intraspinal stem cell transplantation for amyotrophic lateral sclerosis. Ann Neurol 2016; 79:342-53. [PMID: 26696091 DOI: 10.1002/ana.24584] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 12/18/2015] [Accepted: 12/18/2015] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder in which the loss of upper and lower motor neurons produces progressive weakness and eventually death. In the decades since the approval of riluzole, the only US Food and Drug Administration-approved medication to moderately slow progression of ALS, no new therapeutics have arisen to alter the course of the disease. This is partly due to our incomplete understanding of the complex pathogenesis of motor neuron degeneration. Stem cells have emerged as an attractive option in treating ALS, because they come armed with equally complex cellular machinery and may modulate the local microenvironment in many ways to rescue diseased motor neurons. Various stem cell types are being evaluated in preclinical and early clinical applications; here, we review the preclinical strategies and advances supporting the recent clinical translation of neural progenitor cell therapy for ALS. Specifically, we focus on the use of spinal cord neural progenitor cells and the pipeline starting from preclinical studies to the designs of phase I and IIa clinical trials involving direct intraspinal transplantation in humans.
Collapse
Affiliation(s)
- Kevin S Chen
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI
| | - Stacey A Sakowski
- A. Alfred Taubman Medical Research Institute, University of Michigan, Ann Arbor, MI
| | - Eva L Feldman
- A. Alfred Taubman Medical Research Institute and Department of Neurology, University of Michigan, Ann Arbor, MI
| |
Collapse
|