1
|
Goldschmidt-Clermont PJ, Khan A, Jimsheleishvili G, Graham P, Brooks A, Silvera R, Goldschmidt AJ, Pearse DD, Dietrich WD, Levi AD, Guest JD. Treating amyotrophic lateral sclerosis with allogeneic Schwann cell-derived exosomal vesicles: a case report. Neural Regen Res 2025; 20:1207-1216. [PMID: 38922880 PMCID: PMC11438342 DOI: 10.4103/nrr.nrr-d-23-01815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 01/05/2024] [Accepted: 02/24/2024] [Indexed: 06/28/2024] Open
Abstract
Schwann cells are essential for the maintenance and function of motor neurons, axonal networks, and the neuromuscular junction. In amyotrophic lateral sclerosis, where motor neuron function is progressively lost, Schwann cell function may also be impaired. Recently, important signaling and potential trophic activities of Schwann cell-derived exosomal vesicles have been reported. This case report describes the treatment of a patient with advanced amyotrophic lateral sclerosis using serial intravenous infusions of allogeneic Schwann cell-derived exosomal vesicles, marking, to our knowledge, the first instance of such treatment. An 81-year-old male patient presented with a 1.5-year history of rapidly progressive amyotrophic lateral sclerosis. After initial diagnosis, the patient underwent a combination of generic riluzole, sodium phenylbutyrate for the treatment of amyotrophic lateral sclerosis, and taurursodiol. The patient volunteered to participate in an FDA-approved single-patient expanded access treatment and received weekly intravenous infusions of allogeneic Schwann cell-derived exosomal vesicles to potentially restore impaired Schwann cell and motor neuron function. We confirmed that cultured Schwann cells obtained from the amyotrophic lateral sclerosis patient via sural nerve biopsy appeared impaired (senescent) and that exposure of the patient's Schwann cells to allogeneic Schwann cell-derived exosomal vesicles, cultured expanded from a cadaver donor improved their growth capacity in vitro. After a period of observation lasting 10 weeks, during which amyotrophic lateral sclerosis Functional Rating Scale-Revised and pulmonary function were regularly monitored, the patient received weekly consecutive infusions of 1.54 × 10 12 (×2), and then consecutive infusions of 7.5 × 10 12 (×6) allogeneic Schwann cell-derived exosomal vesicles diluted in 40 mL of Dulbecco's phosphate-buffered saline. None of the infusions were associated with adverse events such as infusion reactions (allergic or otherwise) or changes in vital signs. Clinical lab serum neurofilament and cytokine levels measured prior to each infusion varied somewhat without a clear trend. A more sensitive in-house assay suggested possible inflammasome activation during the disease course. A trend for clinical stabilization was observed during the infusion period. Our study provides a novel approach to address impaired Schwann cells and possibly motor neuron function in patients with amyotrophic lateral sclerosis using allogeneic Schwann cell-derived exosomal vesicles. Initial findings suggest that this approach is safe.
Collapse
Affiliation(s)
| | - Aisha Khan
- Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - George Jimsheleishvili
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Patricia Graham
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Adriana Brooks
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Risset Silvera
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Damien D. Pearse
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D. Levi
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - James D. Guest
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
2
|
Simoes FA, Christoforidou E, Cassel R, Dupuis L, Hafezparast M. Severe dynein dysfunction in cholinergic neurons exacerbates ALS-like phenotypes in a new mouse model. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167540. [PMID: 39428001 DOI: 10.1016/j.bbadis.2024.167540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/12/2024] [Accepted: 10/06/2024] [Indexed: 10/22/2024]
Abstract
Cytoplasmic dynein 1, a motor protein essential for retrograde axonal transport, is increasingly implicated in the pathogenesis of neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS). In this study, we developed a novel mouse model that combines the Legs at odd angles (Loa, F580Y) point mutation in the dynein heavy chain with a cholinergic neuron-specific knockout of the dynein heavy chain. This model, for the first time, allows us to investigate the impact of Loa allele exclusivity in these neurons into adulthood. Our findings reveal that this selective increase in dynein dysfunction exacerbated the phenotypes observed in heterozygous Loa mice including pre-wean survival, reduced body weight and grip strength. Additionally, it induced ALS-like pathology in neuromuscular junctions (NMJs) not seen in heterozygous Loa mice. Notably, we also found a previously unobserved significant increase in neurons displaying TDP-43 puncta in both Loa mutants, suggesting early TDP-43 mislocalisation - a hallmark of ALS. The novel model also exhibited a concurrent rise in p62 puncta that did not co-localise with TDP-43, indicating broader impairments in autophagic clearance mechanisms. Overall, this new model underscores the fact that dynein impairment alone can induce ALS-like pathology and provides a valuable platform to further explore the role of dynein in ALS.
Collapse
Affiliation(s)
- Fabio A Simoes
- Department of Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Eleni Christoforidou
- Department of Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | | | - Luc Dupuis
- University of Strasbourg, INSERM, UMR-S1329, Strasbourg, France
| | - Majid Hafezparast
- Department of Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom.
| |
Collapse
|
3
|
Hazell G, McCallion E, Ahlskog N, Sutton ER, Okoh M, Shaqoura EIH, Hoolachan JM, Scaife T, Iqbal S, Bhomra A, Kordala AJ, Scamps F, Raoul C, Wood MJA, Bowerman M. Exercise, disease state and sex influence the beneficial effects of Fn14-depletion on survival and muscle pathology in the SOD1 G93A amyotrophic lateral sclerosis (ALS) mouse model. Skelet Muscle 2024; 14:23. [PMID: 39396990 PMCID: PMC11472643 DOI: 10.1186/s13395-024-00356-0] [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: 07/05/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a devastating and incurable neurodegenerative disease. Accumulating evidence strongly suggests that intrinsic muscle defects exist and contribute to disease progression, including imbalances in whole-body metabolic homeostasis. We have previously reported that tumour necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) and fibroblast growth factor inducible 14 (Fn14) are significantly upregulated in skeletal muscle of the SOD1G93A ALS mouse model. While antagonising TWEAK did not impact survival, we did observe positive effects in skeletal muscle. Given that Fn14 has been proposed as the main effector of the TWEAK/Fn14 activity and that Fn14 can act independently from TWEAK in muscle, we suggest that manipulating Fn14 instead of TWEAK in the SOD1G93A ALS mice could lead to differential and potentially improved benefits. METHODS We thus investigated the contribution of Fn14 to disease phenotypes in the SOD1G93A ALS mice. To do so, Fn14 knockout mice (Fn14-/-) were crossed onto the SOD1G93A background to generate SOD1G93A;Fn14-/- mice. Investigations were performed on both unexercised and exercised (rotarod and/or grid test) animals (wild type (WT), Fn14-/-, SOD1G93A and SOD1G93A;Fn14-/-). RESULTS Here, we firstly confirm that the TWEAK/Fn14 pathway is dysregulated in skeletal muscle of SOD1G93A mice. We then show that Fn14-depleted SOD1G93A mice display increased lifespan, myofiber size, neuromuscular junction endplate area as well as altered expression of known molecular effectors of the TWEAK/Fn14 pathway, without an impact on motor function. Importantly, we also observe a complex interaction between exercise (rotarod and grid test), genotype, disease state and sex that influences the overall effects of Fn14 deletion on survival, expression of known molecular effectors of the TWEAK/Fn14 pathway, expression of myosin heavy chain isoforms and myofiber size. CONCLUSIONS Our study provides further insights on the different roles of the TWEAK/Fn14 pathway in pathological skeletal muscle and how they can be influenced by age, disease, sex and exercise. This is particularly relevant in the ALS field, where combinatorial therapies that include exercise regimens are currently being explored. As such, a better understanding and consideration of the interactions between treatments, muscle metabolism, sex and exercise will be of importance in future studies.
Collapse
Affiliation(s)
- Gareth Hazell
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Eve McCallion
- School of Medicine, Keele University, Staffordshire, UK
| | - Nina Ahlskog
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Emma R Sutton
- School of Medicine, Keele University, Staffordshire, UK
| | - Magnus Okoh
- School of Medicine, Keele University, Staffordshire, UK
| | | | | | - Taylor Scaife
- School of Life Sciences, Keele University, Staffordshire, UK
| | - Sara Iqbal
- School of Life Sciences, Keele University, Staffordshire, UK
| | - Amarjit Bhomra
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Anna J Kordala
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Cedric Raoul
- INM, Univ Montpellier, INSERM, Montpellier, France
- ALS Reference Center, Univ Montpellier, CHU Montpellier, Montpellier, France
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Melissa Bowerman
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
- School of Medicine, Keele University, Staffordshire, UK.
- Wolfson Centre for Inherited Neuromuscular Disease, RJAH Orthopaedic Hospital, Oswestry, UK.
| |
Collapse
|
4
|
Lundt S, Ding S. Potential Therapeutic Interventions Targeting NAD + Metabolism for ALS. Cells 2024; 13:1509. [PMID: 39273079 PMCID: PMC11394323 DOI: 10.3390/cells13171509] [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: 08/04/2024] [Revised: 08/27/2024] [Accepted: 09/07/2024] [Indexed: 09/15/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting both upper and lower motor neurons. While there have been many potential factors implicated for ALS development, such as oxidative stress and mitochondrial dysfunction, no exact mechanism has been determined at this time. Nicotinamide adenine dinucleotide (NAD+) is one of the most abundant metabolites in mammalian cells and is crucial for a broad range of cellular functions from DNA repair to energy homeostasis. NAD+ can be synthesized from three different intracellular pathways, but it is the NAD+ salvage pathway that generates the largest proportion of NAD+. Impaired NAD+ homeostasis has been connected to aging and neurodegenerative disease-related dysfunctions. In ALS mice, NAD+ homeostasis is potentially disrupted prior to the appearance of physical symptoms and is significantly reduced in the nervous system at the end stage. Treatments targeting NAD+ metabolism, either by administering NAD+ precursor metabolites or small molecules that alter NAD+-dependent enzyme activity, have shown strong beneficial effects in ALS disease models. Here, we review the therapeutic interventions targeting NAD+ metabolism for ALS and their effects on the most prominent pathological aspects of ALS in animal and cell models.
Collapse
Affiliation(s)
- Samuel Lundt
- Dalton Cardiovascular Research Center (DCRC), Columbia, MO 65203, USA
| | - Shinghua Ding
- Dalton Cardiovascular Research Center (DCRC), Columbia, MO 65203, USA
- Department of Chemical and Biomedical Engineering (ChBME), University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
5
|
Vieira de Sá R, Sudria-Lopez E, Cañizares Luna M, Harschnitz O, van den Heuvel DMA, Kling S, Vonk D, Westeneng HJ, Karst H, Bloemenkamp L, Varderidou-Minasian S, Schlegel DK, Mars M, Broekhoven MH, van Kronenburg NCH, Adolfs Y, Vangoor VR, de Jongh R, Ljubikj T, Peeters L, Seeler S, Mocholi E, Basak O, Gordon D, Giuliani F, Verhoeff T, Korsten G, Calafat Pla T, Venø MT, Kjems J, Talbot K, van Es MA, Veldink JH, van den Berg LH, Zelina P, Pasterkamp RJ. ATAXIN-2 intermediate-length polyglutamine expansions elicit ALS-associated metabolic and immune phenotypes. Nat Commun 2024; 15:7484. [PMID: 39209824 PMCID: PMC11362472 DOI: 10.1038/s41467-024-51676-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Intermediate-length repeat expansions in ATAXIN-2 (ATXN2) are the strongest genetic risk factor for amyotrophic lateral sclerosis (ALS). At the molecular level, ATXN2 intermediate expansions enhance TDP-43 toxicity and pathology. However, whether this triggers ALS pathogenesis at the cellular and functional level remains unknown. Here, we combine patient-derived and mouse models to dissect the effects of ATXN2 intermediate expansions in an ALS background. iPSC-derived motor neurons from ATXN2-ALS patients show altered stress granules, neurite damage and abnormal electrophysiological properties compared to healthy control and other familial ALS mutations. In TDP-43Tg-ALS mice, ATXN2-Q33 causes reduced motor function, NMJ alterations, neuron degeneration and altered in vitro stress granule dynamics. Furthermore, gene expression changes related to mitochondrial function and inflammatory response are detected and confirmed at the cellular level in mice and human neuron and organoid models. Together, these results define pathogenic defects underlying ATXN2-ALS and provide a framework for future research into ATXN2-dependent pathogenesis and therapy.
Collapse
Affiliation(s)
- Renata Vieira de Sá
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Emma Sudria-Lopez
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Marta Cañizares Luna
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Oliver Harschnitz
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
- Human Technopole, Viale Rita Levi-Montalcini, 1, 20157, Milan, Italy
| | - Dianne M A van den Heuvel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Sandra Kling
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Danielle Vonk
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Henk-Jan Westeneng
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Henk Karst
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Lauri Bloemenkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Suzy Varderidou-Minasian
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Domino K Schlegel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Mayte Mars
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Mark H Broekhoven
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Nicky C H van Kronenburg
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Vamshidhar R Vangoor
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Rianne de Jongh
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Tijana Ljubikj
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Lianne Peeters
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Sabine Seeler
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Enric Mocholi
- Center for Molecuar Medicine, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Onur Basak
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - David Gordon
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
| | - Fabrizio Giuliani
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Tessa Verhoeff
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Giel Korsten
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Teresa Calafat Pla
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - Morten T Venø
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- Omiics ApS, Aarhus, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Kevin Talbot
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Dorothy Crowfoot Hodgkin Building, Oxford, UK
| | - Michael A van Es
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Pavol Zelina
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
6
|
Zhang J, Yang F, Li M, Zhu Y, Huang X. Quantitative evaluation of factors influencing the 3 Hz repetitive nerve stimulation test in patients with amyotrophic lateral sclerosis. Muscle Nerve 2024; 70:194-203. [PMID: 38775303 DOI: 10.1002/mus.28165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 05/01/2024] [Accepted: 05/05/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION/AIMS Previous studies have suggested that treatments targeting the neuromuscular junction (NMJ) may play a role in the treatment of amyotrophic lateral sclerosis (ALS). However, factors impacting repetitive nerve stimulation (RNS), a technique to evaluate NMJ function, have yet to be fully elucidated. We aimed to identify independent factors contributing to the decremental response of the accessory nerve and evaluated its value in ALS clinical practice. METHODS A total of 626 patients who were diagnosed with ALS and underwent 3 Hz RNS tests on the accessory nerve were enrolled. Data on their clinical and electrophysiological indicators were divided into a training set (collected from June 2016 to December 2022) and a test set (collected from January to August 2023). Stepwise regression was used in independent variable selection and model building. RESULTS Forty-two percent of patients had a decrement larger than 10% and 24% had a decrement larger than 15%. Onset age, sex, onset site, forced vital capacity (FVC) and motor unit potential (MUP) duration were independent factors contributing to the results of the RNS test. MUP duration had the greatest impact on decremental response, followed by FVC and onset age. The decremental response in females was larger than in males. Upper limb onset was found to contribute more to the decrement than lower limb or bulbar onset. DISCUSSION In patients with ALS, NMJ safety factor is reduced during re-innervation. Decremental response is affected by multiple factors, which needs to be considered in clinical trials targeting the NMJ in these patients.
Collapse
Affiliation(s)
- Jinghong Zhang
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Fei Yang
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mao Li
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yahui Zhu
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xusheng Huang
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Chen HH, Yeo HT, Huang YH, Tsai LK, Lai HJ, Tsao YP, Chen SL. AAV-NRIP gene therapy ameliorates motor neuron degeneration and muscle atrophy in ALS model mice. Skelet Muscle 2024; 14:17. [PMID: 39044305 PMCID: PMC11267858 DOI: 10.1186/s13395-024-00349-z] [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: 02/09/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is characterized by progressive motor neuron (MN) degeneration, leading to neuromuscular junction (NMJ) dismantling and severe muscle atrophy. The nuclear receptor interaction protein (NRIP) functions as a multifunctional protein. It directly interacts with calmodulin or α-actinin 2, serving as a calcium sensor for muscle contraction and maintaining sarcomere integrity. Additionally, NRIP binds with the acetylcholine receptor (AChR) for NMJ stabilization. Loss of NRIP in muscles results in progressive motor neuron degeneration with abnormal NMJ architecture, resembling ALS phenotypes. Therefore, we hypothesize that NRIP could be a therapeutic factor for ALS. METHODS We used SOD1 G93A mice, expressing human SOD1 with the ALS-linked G93A mutation, as an ALS model. An adeno-associated virus vector encoding the human NRIP gene (AAV-NRIP) was generated and injected into the muscles of SOD1 G93A mice at 60 days of age, before disease onset. Pathological and behavioral changes were measured to evaluate the therapeutic effects of AAV-NRIP on the disease progression of SOD1 G93A mice. RESULTS SOD1 G93A mice exhibited lower NRIP expression than wild-type mice in both the spinal cord and skeletal muscle tissues. Forced NRIP expression through AAV-NRIP intramuscular injection was observed in skeletal muscles and retrogradely transduced into the spinal cord. AAV-NRIP gene therapy enhanced movement distance and rearing frequencies in SOD1 G93A mice. Moreover, AAV-NRIP increased myofiber size and slow myosin expression, ameliorated NMJ degeneration and axon terminal denervation at NMJ, and increased the number of α-motor neurons (α-MNs) and compound muscle action potential (CMAP) in SOD1 G93A mice. CONCLUSIONS AAV-NRIP gene therapy ameliorates muscle atrophy, motor neuron degeneration, and axon terminal denervation at NMJ, leading to increased NMJ transmission and improved motor functions in SOD1 G93A mice. Collectively, AAV-NRIP could be a potential therapeutic drug for ALS.
Collapse
Affiliation(s)
- Hsin-Hsiung Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Hsin-Tung Yeo
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Yun-Hsin Huang
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Hsing-Jung Lai
- Department of Neurology, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Yeou-Ping Tsao
- Department of Ophthalmology, Mackay Memorial Hospital, Taipei, 104, Taiwan
| | - Show-Li Chen
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
8
|
Turano E, Virla F, Scambi I, Dabrowska S, Bankole O, Mariotti R. Adipose mesenchymal stem cells-derived extracellular vesicles exert their preferential action in damaged central sites of SOD1 mice rather than peripherally. Eur J Histochem 2024; 68:4040. [PMID: 38963135 PMCID: PMC11256976 DOI: 10.4081/ejh.2024.4040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/10/2024] [Indexed: 07/05/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder involving motor neuron (MN) loss in the motor cortex, brainstem and spinal cord leading to progressive paralysis and death. Due to the pathogenetic complexity, there are no effective therapies available. In this context the use of mesenchymal stem cells and their vesicular counterpart is an emerging therapeutic strategy to counteract neurodegeneration. The extracellular vesicles derived from adipose stem cells (ASC-EVs) recapitulate and ameliorate the neuroprotective effect of stem cells and, thanks to their small dimensions, makes their use suitable to develop novel therapeutic approaches for neurodegenerative diseases as ALS. Here we investigate a therapeutic regimen of ASC-EVs injection in SOD1(G93A) mice, the most widely used murine model of ALS. Repeated intranasal administrations of high doses of ASC-EVs were able to ameliorate motor performance of injected SOD1(G93A) mice at the early stage of the disease and produce a significant improvement at the end-stage in the lumbar MNs rescue. Moreover, ASC-EVs preserve the structure of neuromuscular junction without counteracting the muscle atrophy. The results indicate that the intranasal ASC-EVs administration acts in central nervous system sites rather than at peripheral level in SOD1(G93A) mice. These considerations allow us to identify future applications of ASC-EVs that involve different targets simultaneously to maximize the clinical and neuropathological outcomes in ALS in vivo models.
Collapse
Affiliation(s)
- Ermanna Turano
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona.
| | - Federica Virla
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona.
| | - Ilaria Scambi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona.
| | - Sylwia Dabrowska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw.
| | - Oluwamolakun Bankole
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Alberta.
| | - Raffaella Mariotti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona.
| |
Collapse
|
9
|
Lundt S, Zhang N, Polo-Parada L, Wang X, Ding S. Dietary NMN supplementation enhances motor and NMJ function in ALS. Exp Neurol 2024; 374:114698. [PMID: 38266764 DOI: 10.1016/j.expneurol.2024.114698] [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: 10/16/2023] [Revised: 01/15/2024] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease that causes the degeneration of motor neurons in the motor cortex and spinal cord. Patients with ALS experience muscle weakness and atrophy in the limbs which eventually leads to paralysis and death. NAD+ is critical for energy metabolism, such as glycolysis and oxidative phosphorylation, but is also involved in non-metabolic cellular reactions. In the current study, we determined whether the supplementation of nicotinamide mononucleotide (NMN), an NAD+ precursor, in the diet had beneficial impacts on disease progression using a SOD1G93A mouse model of ALS. We found that the ALS mice fed with an NMN-supplemented diet (ALS+NMN mice) had modestly extended lifespan and exhibited delayed motor dysfunction. Using electrophysiology, we studied the effect of NMN on synaptic transmission at neuromuscular junctions (NMJs) in symptomatic of ALS mice (18 weeks old). ALS+NMN mice had larger end-plate potential (EPP) amplitudes and maintained better responses than ALS mice, and also had restored EPP facilitation. While quantal content was not affected by NMN, miniature EPP (mEPP) amplitude and frequency were elevated in ALS+NMN mice. NMN supplementation in diet also improved NMJ morphology, innervation, mitochondrial structure, and reduced reactive astrogliosis in the ventral horn of the lumbar spinal cord. Overall, our results indicate that dietary consumption of NMN can slow motor impairment, enhance NMJ function and improve healthspan of ALS mice.
Collapse
Affiliation(s)
- Samuel Lundt
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO 65211, United States of America; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States of America
| | - Nannan Zhang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States of America
| | - Luis Polo-Parada
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States of America; Department of Medical, Physiology and Pharmacology, University of Missouri, Columbia, MO 65211, United States of America
| | - Xinglong Wang
- Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ 85721, United States of America
| | - Shinghua Ding
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO 65211, United States of America; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, United States of America; Department of Chemical and Biomedical Engineering, University of Missouri, Columbia, MO 65211, United States of America.
| |
Collapse
|
10
|
Cohen J, Mathew A, Dourvetakis KD, Sanchez-Guerrero E, Pangeni RP, Gurusamy N, Aenlle KK, Ravindran G, Twahir A, Isler D, Sosa-Garcia SR, Llizo A, Bested AC, Theoharides TC, Klimas NG, Kempuraj D. Recent Research Trends in Neuroinflammatory and Neurodegenerative Disorders. Cells 2024; 13:511. [PMID: 38534355 DOI: 10.3390/cells13060511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Neuroinflammatory and neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease (PD), traumatic brain injury (TBI) and Amyotrophic lateral sclerosis (ALS) are chronic major health disorders. The exact mechanism of the neuroimmune dysfunctions of these disease pathogeneses is currently not clearly understood. These disorders show dysregulated neuroimmune and inflammatory responses, including activation of neurons, glial cells, and neurovascular unit damage associated with excessive release of proinflammatory cytokines, chemokines, neurotoxic mediators, and infiltration of peripheral immune cells into the brain, as well as entry of inflammatory mediators through damaged neurovascular endothelial cells, blood-brain barrier and tight junction proteins. Activation of glial cells and immune cells leads to the release of many inflammatory and neurotoxic molecules that cause neuroinflammation and neurodegeneration. Gulf War Illness (GWI) and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are chronic disorders that are also associated with neuroimmune dysfunctions. Currently, there are no effective disease-modifying therapeutic options available for these diseases. Human induced pluripotent stem cell (iPSC)-derived neurons, astrocytes, microglia, endothelial cells and pericytes are currently used for many disease models for drug discovery. This review highlights certain recent trends in neuroinflammatory responses and iPSC-derived brain cell applications in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Jessica Cohen
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Annette Mathew
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Kirk D Dourvetakis
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Estella Sanchez-Guerrero
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Rajendra P Pangeni
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Narasimman Gurusamy
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Kristina K Aenlle
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA
| | - Geeta Ravindran
- Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Assma Twahir
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Dylan Isler
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Sara Rukmini Sosa-Garcia
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Axel Llizo
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Alison C Bested
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Theoharis C Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Nancy G Klimas
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| |
Collapse
|
11
|
Jackson WS, Bauer S, Kaczmarczyk L, Magadi SS. Selective Vulnerability to Neurodegenerative Disease: Insights from Cell Type-Specific Translatome Studies. BIOLOGY 2024; 13:67. [PMID: 38392286 PMCID: PMC10886597 DOI: 10.3390/biology13020067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 02/24/2024]
Abstract
Neurodegenerative diseases (NDs) manifest a wide variety of clinical symptoms depending on the affected brain regions. Gaining insights into why certain regions are resistant while others are susceptible is vital for advancing therapeutic strategies. While gene expression changes offer clues about disease responses across brain regions, the mixture of cell types therein obscures experimental results. In recent years, methods that analyze the transcriptomes of individual cells (e.g., single-cell RNA sequencing or scRNAseq) have been widely used and have provided invaluable insights into specific cell types. Concurrently, transgene-based techniques that dissect cell type-specific translatomes (CSTs) in model systems, like RiboTag and bacTRAP, offer unique advantages but have received less attention. This review juxtaposes the merits and drawbacks of both methodologies, focusing on the use of CSTs in understanding conditions like amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), Alzheimer's disease (AD), and specific prion diseases like fatal familial insomnia (FFI), genetic Creutzfeldt-Jakob disease (gCJD), and acquired prion disease. We conclude by discussing the emerging trends observed across multiple diseases and emerging methods.
Collapse
Affiliation(s)
- Walker S Jackson
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Susanne Bauer
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Lech Kaczmarczyk
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| | - Srivathsa S Magadi
- Wallenberg Center for Molecular Medicine, Linköping University, 581 85 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden
| |
Collapse
|
12
|
Chen D, Philippidou P, Brenha BDF, Schaffer AE, Miranda HC. Scalable, optically-responsive human neuromuscular junction model reveals convergent mechanisms of synaptic dysfunction in familial ALS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575304. [PMID: 38260655 PMCID: PMC10802619 DOI: 10.1101/2024.01.11.575304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Neuromuscular junctions (NMJs) are specialized synapses that mediate communication between motor neurons and skeletal muscles and are essential for movement. The degeneration of this system can lead to symptoms observed in neuromuscular and motor neuron diseases. Studying these synapses and their degeneration has proven challenging. Prior NMJ studies heavily relied upon the use of mouse, chick, or isolated primary human cells, which have demonstrated limited fidelity for disease modeling. To enable the study of NMJ dysfunction and model genetic diseases, we, and others, have developed methods to generate human NMJs from pluripotent stem cells (PSCs), embryonic stem cells, and induced pluripotent stem cells. However, published studies have highlighted technical limitations associated with these complex in vitro NMJ models. In this study, we developed a robust PSC-derived motor neuron and skeletal muscle co-culture method, and demonstrated its sensitivity in modeling motor neuron disease. Our method spontaneously and reproducibly forms human NMJs. We developed multiwell-multielectrode array (MEA) parameters to quantify the activity of PSC-derived skeletal muscles, as well as measured the electrophysiological activity of functional human PSC-derived NMJs. We further leveraged our method to morphologically and functionally assess NMJs from the familial amyotrophic lateral sclerosis (fALS) PSCs, C9orf72 hexanucleotide (G4C2)n repeat expansion (HRE), SOD1 A5V , and TDP43 G298S to define the reproducibility and sensitivity of our system. We observed a significant decrease in the numbers and activity of PSC-derived NMJs developed from the different ALS lines compared to their respective controls. Furthermore, we evaluated a therapeutic candidate undergoing clinical trials and observed a variant-dependent rescue of functionality of NMJs. Our newly developed method provides a platform for the systematic investigation of genetic causes of NMJ neurodegeneration and highlights the need for therapeutic avenues to consider patient genotype.
Collapse
|
13
|
Miao Y, Xie L, Song J, Cai X, Yang J, Ma X, Chen S, Xie P. Unraveling the causes of sarcopenia: Roles of neuromuscular junction impairment and mitochondrial dysfunction. Physiol Rep 2024; 12:e15917. [PMID: 38225199 PMCID: PMC10789655 DOI: 10.14814/phy2.15917] [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: 10/20/2023] [Revised: 12/11/2023] [Accepted: 12/22/2023] [Indexed: 01/17/2024] Open
Abstract
Sarcopenia is a systemic skeletal muscle disease characterized by a decline in skeletal muscle mass and function. Originally defined as an age-associated condition, sarcopenia presently also encompasses muscular atrophy due to various pathological factors, such as intensive care unit-acquired weakness, inactivity, and malnutrition. The exact pathogenesis of sarcopenia is still unknown; herein, we review the pathological roles of the neuromuscular junction and mitochondria in this condition. Sarcopenia is caused by complex and interdependent pathophysiological mechanisms, including aging, neuromuscular junction impairment, mitochondrial dysfunction, insulin resistance, lipotoxicity, endocrine factors, oxidative stress, and inflammation. Among these, neuromuscular junction instability and mitochondrial dysfunction are particularly significant. Dysfunction in neuromuscular junction can lead to muscle weakness or paralysis. Mitochondria, which are plentiful in neurons and muscle fibers, play an important role in neuromuscular junction transmission. Therefore, impairments in both mitochondria and neuromuscular junction may be one of the key pathophysiological mechanisms leading to sarcopenia. Moreover, this article explores the structural and functional alterations in the neuromuscular junction and mitochondria in sarcopenia, suggesting that a deeper understanding of these changes could provide valuable insights for the prevention or treatment of sarcopenia.
Collapse
Affiliation(s)
- Yanmei Miao
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| | - Leiyu Xie
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| | - Jiamei Song
- Department of Nursing of Affiliated HospitalZunyi Medical UniversityZunyiChina
| | - Xing Cai
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| | - Jinghe Yang
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
- Department of The First Clinical CollegeZunyi Medical UniversityZunyiChina
| | - Xinglong Ma
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| | - Shaolin Chen
- Department of Nursing of Affiliated HospitalZunyi Medical UniversityZunyiChina
| | - Peng Xie
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi)Zunyi Medical UniversityZunyiChina
| |
Collapse
|
14
|
Kang Q, Jiang S, Min J, Hu F, Xu R. Parvalbumin interneurons dysfunction is potentially associated with FαMNs decrease and NRG1-ErbB4 signaling inhibition in spinal cord in amyotrophic lateral sclerosis. Aging (Albany NY) 2023; 15:15324-15339. [PMID: 38157256 PMCID: PMC10781496 DOI: 10.18632/aging.205351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE To investigate the alteration of PV interneurons in ALS mainly focusing its dynamic changes and its relationship with motor neurons and ErbB4 signaling. METHODS SOD1G93A mice were used as ALS model. ALS animals were divided into different groups according to birth age: symptomatic prophase (50~60 days), symptomatic phase (90~100 days), and symptomatic progression (130~140 days). Immunofluorescence was performed for measurement of PV-positive interneurons, MMP-9, ChAT, NeuN and ErbB4. RT-qPCR and western blot were used to determine the expression of PV and MMP-9. RESULTS PV expression was remarkably higher in the anterior horn of gray matter compared with posterior horn and area in the middle of gray matter in control mice. In ALS mice, PV, MMP-9 and ErbB4 levels were gradually decreased along with onset. PV, MMP-9 and ErbB4 levels in ALS mice were significantly down-regulated than control mice after onset, indicating the alteration of PV interneurons, FαMNs and ErbB4. SαMNs levels only decreased remarkably at symptomatic progression in ALS mice compared with control mice, while γMNs levels showed no significant change during whole period in all mice. MMP-9 and ErbB4 were positively correlated with PV. NRG1 treatment significantly enhanced the expression of ErBb4, PV and MMP-9 in ALS mice. CONCLUSION PV interneurons decrease is along with FαMNs and ErbB4 decrease in ALS mice.
Collapse
Affiliation(s)
- Qin Kang
- Department of Neurology, Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Neurology, Jiangxi Provincial People’s Hospital, First Affiliated Hospital of Nanchang Medical College, Clinical College of Nanchang Medical College, Nanchang 330006, Jiangxi, P.R. China
| | - Shishi Jiang
- Department of Neurology, Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Jun Min
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Fan Hu
- Department of Neurology, Jiangxi Provincial People’s Hospital, First Affiliated Hospital of Nanchang Medical College, Clinical College of Nanchang Medical College, Nanchang 330006, Jiangxi, P.R. China
| | - Renshi Xu
- Department of Neurology, Medical College of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Neurology, Jiangxi Provincial People’s Hospital, First Affiliated Hospital of Nanchang Medical College, Clinical College of Nanchang Medical College, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
15
|
Pelaez MC, Desmeules A, Gelon PA, Glasson B, Marcadet L, Rodgers A, Phaneuf D, Pozzi S, Dutchak PA, Julien JP, Sephton CF. Neuronal dysfunction caused by FUSR521G promotes ALS-associated phenotypes that are attenuated by NF-κB inhibition. Acta Neuropathol Commun 2023; 11:182. [PMID: 37974279 PMCID: PMC10652582 DOI: 10.1186/s40478-023-01671-1] [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: 08/04/2023] [Accepted: 10/09/2023] [Indexed: 11/19/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are related neurodegenerative diseases that belong to a common disease spectrum based on overlapping clinical, pathological and genetic evidence. Early pathological changes to the morphology and synapses of affected neuron populations in ALS/FTD suggest a common underlying mechanism of disease that requires further investigation. Fused in sarcoma (FUS) is a DNA/RNA-binding protein with known genetic and pathological links to ALS/FTD. Expression of ALS-linked FUS mutants in mice causes cognitive and motor defects, which correlate with loss of motor neuron dendritic branching and synapses, in addition to other pathological features of ALS/FTD. The role of ALS-linked FUS mutants in causing ALS/FTD-associated disease phenotypes is well established, but there are significant gaps in our understanding of the cell-autonomous role of FUS in promoting structural changes to motor neurons, and how these changes relate to disease progression. Here we generated a neuron-specific FUS-transgenic mouse model expressing the ALS-linked human FUSR521G variant, hFUSR521G/Syn1, to investigate the cell-autonomous role of FUSR521G in causing loss of dendritic branching and synapses of motor neurons, and to understand how these changes relate to ALS-associated phenotypes. Longitudinal analysis of mice revealed that cognitive impairments in juvenile hFUSR521G/Syn1 mice coincide with reduced dendritic branching of cortical motor neurons in the absence of motor impairments or changes in the neuromorphology of spinal motor neurons. Motor impairments and dendritic attrition of spinal motor neurons developed later in aged hFUSR521G/Syn1 mice, along with FUS cytoplasmic mislocalisation, mitochondrial abnormalities and glial activation. Neuroinflammation promotes neuronal dysfunction and drives disease progression in ALS/FTD. The therapeutic effects of inhibiting the pro-inflammatory nuclear factor kappa B (NF-κB) pathway with an analog of Withaferin A, IMS-088, were assessed in symptomatic hFUSR521G/Syn1 mice and were found to improve cognitive and motor function, increase dendritic branches and synapses of motor neurons, and attenuate other ALS/FTD-associated pathological features. Treatment of primary cortical neurons expressing FUSR521G with IMS-088 promoted the restoration of dendritic mitochondrial numbers and mitochondrial activity to wild-type levels, suggesting that inhibition of NF-κB permits the restoration of mitochondrial stasis in our models. Collectively, this work demonstrates that FUSR521G has a cell-autonomous role in causing early pathological changes to dendritic and synaptic structures of motor neurons, and that these changes precede motor defects and other well-known pathological features of ALS/FTD. Finally, these findings provide further support that modulation of the NF-κB pathway in ALS/FTD is an important therapeutic approach to attenuate disease.
Collapse
Affiliation(s)
- Mari Carmen Pelaez
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Antoine Desmeules
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Pauline A Gelon
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Bastien Glasson
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Laetitia Marcadet
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Alicia Rodgers
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Daniel Phaneuf
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Silvia Pozzi
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Paul A Dutchak
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Jean-Pierre Julien
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Chantelle F Sephton
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada.
| |
Collapse
|
16
|
Huang YT, Crick HR, Chaytow H, van der Hoorn D, Alhindi A, Jones RA, Hector RD, Cobb SR, Gillingwater TH. Long-term muscle-specific overexpression of DOK7 in mice using AAV9-tMCK-DOK7. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:617-628. [PMID: 37637210 PMCID: PMC10457688 DOI: 10.1016/j.omtn.2023.07.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023]
Abstract
Neuromuscular junction (NMJ) dysfunction underlies several diseases, including congenital myasthenic syndromes (CMSs) and motor neuron disease (MND). Molecular pathways governing NMJ stability are therefore of interest from both biological and therapeutic perspectives. Muscle-specific kinase (MuSK) is necessary for the formation and maintenance of post-synaptic elements of the NMJ, and downstream of tyrosine kinases 7 (DOK7) is crucial for activation of the MuSK pathway. Overexpression of DOK7 using AAV9 has been shown to ameliorate neuromuscular pathology in pre-clinical disease models of CMS and MND. However, long-term consequences of DOK7 expression have been sparsely investigated and targeted overexpression of DOK7 in skeletal muscle yet to be established. Here, we developed and characterized a novel AAV9-DOK7 facilitating forced expression of DOK7 under a skeletal muscle-specific promoter. AAV9-tMCK-DOK7 was systemically delivered to newborn mice that were monitored over 6 months. DOK7 overexpression was restricted to skeletal muscles. Body weight, blood biochemistry, and histopathological assessments were unaffected by AAV9-tMCK-DOK7 treatment. In contrast, forced expression of DOK7 resulted in enlargement of both the pre- and post-synaptic components of the NMJ, without causing denervation. We conclude that muscle-specific DOK7 overexpression can be achieved in a safe manner, with the capacity to target NMJs in vivo.
Collapse
Affiliation(s)
- Yu-Ting Huang
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | - Hannah R. Crick
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | - Helena Chaytow
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | - Dinja van der Hoorn
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | - Abrar Alhindi
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
- Faculty of Medicine, Department of Anatomy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ross A. Jones
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| | | | | | - Thomas H. Gillingwater
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh EH16 4SB, UK
| |
Collapse
|
17
|
Alhindi A, Shand M, Smith HL, Leite AS, Huang YT, van der Hoorn D, Ridgway Z, Faller KME, Jones RA, Gillingwater TH, Chaytow H. Neuromuscular junction denervation and terminal Schwann cell loss in the hTDP-43 overexpression mouse model of amyotrophic lateral sclerosis. Neuropathol Appl Neurobiol 2023; 49:e12925. [PMID: 37465879 DOI: 10.1111/nan.12925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 07/20/2023]
Abstract
AIMS Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with complex aetiology. Despite evidence of neuromuscular junction (NMJ) denervation and 'dying-back' pathology in models of SOD1-dependent ALS, evidence in other genetic forms of ALS is limited by a lack of suitable animal models. TDP-43, a key mediator protein in ALS, is overexpressed in neurons in Thy1-hTDP-43WT mice. We therefore aimed to comprehensively analyse NMJ pathology in this model of ALS. METHODS Expression of TDP-43 was assessed via western blotting. Immunohistochemistry techniques, alongside NMJ-morph quantification, were used to analyse motor neuron number, NMJ denervation status and terminal Schwann cell morphology. RESULTS We present a time course of progressive, region-specific motor neuron pathology in Thy1-hTDP-43WT mice. Thy1-driven hTDP-43 expression increased steadily, correlating with developing hindlimb motor weakness and associated motor neuron loss in the spinal cord with a median survival of 21 days. Pronounced NMJ denervation was observed in hindlimb muscles, mild denervation in cranial muscles but no evidence of denervation in either forelimb or trunk muscles. NMJ pathology was restricted to motor nerve terminals, with denervation following the same time course as motor neuron loss. Terminal Schwann cells were lost from NMJs in hindlimb muscles, directly correlating with denervation status. CONCLUSIONS Thy1-hTDP-43WT mice represent a severe model of ALS, with NMJ pathology/denervation of distal muscles and motor neuron loss, as observed in ALS patients. This model therefore provides an ideal platform to investigate mechanisms of dying-back pathology, as well as NMJ-targeting disease-modifying therapies in ALS.
Collapse
Affiliation(s)
- Abrar Alhindi
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
- Faculty of Medicine, Department of Anatomy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Megan Shand
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
| | - Hannah L Smith
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
| | - Ana S Leite
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
- School of Medicine, UNESP-São Paulo State University, Botucatu, Sao Paulo, Brazil
| | - Yu-Ting Huang
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
| | - Dinja van der Hoorn
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
| | - Zara Ridgway
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
| | - Kiterie M E Faller
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Ross A Jones
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
| | - Helena Chaytow
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, Edinburgh, UK
| |
Collapse
|
18
|
Kandhavivorn W, Glaß H, Herrmannsdörfer T, Böckers TM, Uhlarz M, Gronemann J, Funk RHW, Pietzsch J, Pal A, Hermann A. Restoring Axonal Organelle Motility and Regeneration in Cultured FUS-ALS Motoneurons through Magnetic Field Stimulation Suggests an Alternative Therapeutic Approach. Cells 2023; 12:1502. [PMID: 37296623 PMCID: PMC10252208 DOI: 10.3390/cells12111502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating motoneuron disease characterized by sustained loss of neuromuscular junctions, degenerating corticospinal motoneurons and rapidly progressing muscle paralysis. Motoneurons have unique features, essentially a highly polarized, lengthy architecture of axons, posing a considerable challenge for maintaining long-range trafficking routes for organelles, cargo, mRNA and secretion with a high energy effort to serve crucial neuronal functions. Impaired intracellular pathways implicated in ALS pathology comprise RNA metabolism, cytoplasmic protein aggregation, cytoskeletal integrity for organelle trafficking and maintenance of mitochondrial morphology and function, cumulatively leading to neurodegeneration. Current drug treatments only have marginal effects on survival, thereby calling for alternative ALS therapies. Exposure to magnetic fields, e.g., transcranial magnetic stimulations (TMS) on the central nervous system (CNS), has been broadly explored over the past 20 years to investigate and improve physical and mental activities through stimulated excitability as well as neuronal plasticity. However, studies of magnetic treatments on the peripheral nervous system are still scarce. Thus, we investigated the therapeutic potential of low frequency alternating current magnetic fields on cultured spinal motoneurons derived from induced pluripotent stem cells of FUS-ALS patients and healthy persons. We report a remarkable restoration induced by magnetic stimulation on axonal trafficking of mitochondria and lysosomes and axonal regenerative sprouting after axotomy in FUS-ALS in vitro without obvious harmful effects on diseased and healthy neurons. These beneficial effects seem to derive from improved microtubule integrity. Thus, our study suggests the therapeutic potential of magnetic stimulations in ALS, which awaits further exploration and validation in future long-term in vivo studies.
Collapse
Affiliation(s)
- Wonphorn Kandhavivorn
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
- Institute of Anatomy, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Hannes Glaß
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, D-01307 Dresden, Germany;
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, D-18147 Rostock, Germany
| | - Thomas Herrmannsdörfer
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
| | - Tobias M. Böckers
- Institute of Anatomy and Cell Biology, University of Ulm, D-89081 Ulm, Germany;
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Ulm, D-89081 Ulm, Germany
| | - Marc Uhlarz
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
| | - Jonas Gronemann
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
| | - Richard H. W. Funk
- Institute of Anatomy, Technische Universität Dresden, D-01307 Dresden, Germany
- Dresden International University, D-01067 Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, D-01069 Dresden, Germany
| | - Arun Pal
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, D-01307 Dresden, Germany;
| | - Andreas Hermann
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, D-01307 Dresden, Germany;
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, D-18147 Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, D-18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, D-18147 Rostock, Germany
| |
Collapse
|
19
|
Mukhamedyarov MA, Khabibrakhmanov AN, Khuzakhmetova VF, Giniatullin AR, Zakirjanova GF, Zhilyakov NV, Mukhutdinova KA, Samigullin DV, Grigoryev PN, Zakharov AV, Zefirov AL, Petrov AM. Early Alterations in Structural and Functional Properties in the Neuromuscular Junctions of Mutant FUS Mice. Int J Mol Sci 2023; 24:9022. [PMID: 37240370 PMCID: PMC10218837 DOI: 10.3390/ijms24109022] [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: 04/04/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is manifested as skeletal muscle denervation, loss of motor neurons and finally severe respiratory failure. Mutations of RNA-binding protein FUS are one of the common genetic reasons of ALS accompanied by a 'dying back' type of degeneration. Using fluorescent approaches and microelectrode recordings, the early structural and functional alterations in diaphragm neuromuscular junctions (NMJs) were studied in mutant FUS mice at the pre-onset stage. Lipid peroxidation and decreased staining with a lipid raft marker were found in the mutant mice. Despite the preservation of the end-plate structure, immunolabeling revealed an increase in levels of presynaptic proteins, SNAP-25 and synapsin 1. The latter can restrain Ca2+-dependent synaptic vesicle mobilization. Indeed, neurotransmitter release upon intense nerve stimulation and its recovery after tetanus and compensatory synaptic vesicle endocytosis were markedly depressed in FUS mice. There was a trend to attenuation of axonal [Ca2+]in increase upon nerve stimulation at 20 Hz. However, no changes in neurotransmitter release and the intraterminal Ca2+ transient in response to low frequency stimulation or in quantal content and the synchrony of neurotransmitter release at low levels of external Ca2+ were detected. At a later stage, shrinking and fragmentation of end plates together with a decrease in presynaptic protein expression and disturbance of the neurotransmitter release timing occurred. Overall, suppression of synaptic vesicle exo-endocytosis upon intense activity probably due to alterations in membrane properties, synapsin 1 levels and Ca2+ kinetics could be an early sign of nascent NMJ pathology, which leads to neuromuscular contact disorganization.
Collapse
Affiliation(s)
- Marat A. Mukhamedyarov
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova St., Kazan 420012, Russia; (M.A.M.)
| | - Aydar N. Khabibrakhmanov
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova St., Kazan 420012, Russia; (M.A.M.)
| | - Venera F. Khuzakhmetova
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center ‘‘Kazan Scientific Center of RAS”, 2/31 Lobachevsky St., P.O. Box 30, Kazan 420111, Russia (N.V.Z.)
| | - Arthur R. Giniatullin
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova St., Kazan 420012, Russia; (M.A.M.)
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center ‘‘Kazan Scientific Center of RAS”, 2/31 Lobachevsky St., P.O. Box 30, Kazan 420111, Russia (N.V.Z.)
| | - Guzalia F. Zakirjanova
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova St., Kazan 420012, Russia; (M.A.M.)
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center ‘‘Kazan Scientific Center of RAS”, 2/31 Lobachevsky St., P.O. Box 30, Kazan 420111, Russia (N.V.Z.)
| | - Nikita V. Zhilyakov
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center ‘‘Kazan Scientific Center of RAS”, 2/31 Lobachevsky St., P.O. Box 30, Kazan 420111, Russia (N.V.Z.)
| | - Kamilla A. Mukhutdinova
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova St., Kazan 420012, Russia; (M.A.M.)
| | - Dmitry V. Samigullin
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center ‘‘Kazan Scientific Center of RAS”, 2/31 Lobachevsky St., P.O. Box 30, Kazan 420111, Russia (N.V.Z.)
- Department of Radiophotonics and Microwave Technologies, Kazan National Research Technical University, 10 K. Marx St., Kazan 420111, Russia
| | - Pavel N. Grigoryev
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova St., Kazan 420012, Russia; (M.A.M.)
| | - Andrey V. Zakharov
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova St., Kazan 420012, Russia; (M.A.M.)
- Laboratory of Neurobiology, Kazan Federal University, Kazan 420008, Russia
| | - Andrey L. Zefirov
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova St., Kazan 420012, Russia; (M.A.M.)
| | - Alexey M. Petrov
- Department of Normal Physiology, Kazan State Medial University, 49 Butlerova St., Kazan 420012, Russia; (M.A.M.)
- Kazan Institute of Biochemistry and Biophysics, Federal Research Center ‘‘Kazan Scientific Center of RAS”, 2/31 Lobachevsky St., P.O. Box 30, Kazan 420111, Russia (N.V.Z.)
| |
Collapse
|
20
|
McIntosh J, Mekrouda I, Dashti M, Giuraniuc CV, Banks RW, Miles GB, Bewick GS. Development of abnormalities at the neuromuscular junction in the SOD1-G93A mouse model of ALS: dysfunction then disruption of postsynaptic structure precede overt motor symptoms. Front Mol Neurosci 2023; 16:1169075. [PMID: 37273905 PMCID: PMC10237339 DOI: 10.3389/fnmol.2023.1169075] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/12/2023] [Indexed: 06/06/2023] Open
Abstract
Introduction The ultimate deficit in amyotrophic lateral sclerosis (ALS) is neuromuscular junction (NMJ) loss, producing permanent paralysis, ultimately in respiratory muscles. However, understanding the functional and structural deficits at NMJs prior to this loss is crucial for therapeutic strategy design. Should early interventions focus on reversing denervation, or supporting largely intact NMJs that are functionally impaired? We therefore determined when functional and structural deficits appeared in diaphragmatic NMJs relative to the onset of hindlimb tremor (the first overt motor symptoms) in vivo in the SOD1-G93A mouse model of ALS. Materials and methods We employed electrophysiological recording of NMJ postsynaptic potentials for spontaneous and nerve stimulation-evoked responses. This was correlated with fluorescent imaging microscopy of the postsynaptic acetylcholine receptor (AChR) distribution throughout the postnatal developmental timecourse from 2 weeks to early symptomatic ages. Results Significant reduction in the amplitudes of spontaneous miniature endplate potentials (mEPPs) and evoked EPPs emerged only at early symptomatic ages (in our colony, 18-22 weeks). Reductions in mEPP frequency, number of vesicles per EPP, and EPP rise time were seen earlier, at 16weeks, but this reversed by early symptomatic ages. However, the earliest and most striking impairment was an inability to maintain EPP amplitude during a 20 Hz stimulus train, which appeared 6 weeks before overt in vivo motor symptoms. Despite this, fluorescent α-bungarotoxin labelling revealed no systematic, progressive changes in 11 comprehensive NMJ morphological parameters (area, shape, compactness, number of acetylcholine receptor, AChR, regions, etc.) with disease progression. Rather, while NMJs were largely normally-shaped, from 16 weeks there was a progressive and substantial disruption in AChR concentration and distribution within the NMJ footprint. Discussion Thus, NMJ functional deficits appear at least 6 weeks before motor symptoms in vivo, while structural deficits occur 4 weeks later, and predominantly within NMJs. These data suggest initial therapies focused on rectifying suboptimal NMJ function could produce effective relief of symptoms of weakness.
Collapse
Affiliation(s)
- Jayne McIntosh
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Imane Mekrouda
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Maryam Dashti
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | | - Robert W. Banks
- Department of Biosciences, Durham University, Durham, United Kingdom
| | - Gareth B. Miles
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, United Kingdom
| | - Guy S. Bewick
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
21
|
In vivo imaging of axonal transport in peripheral nerves of rodent forelimbs. Neuronal Signal 2023; 7:NS20220098. [PMID: 36743438 PMCID: PMC9867938 DOI: 10.1042/ns20220098] [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: 12/01/2022] [Revised: 12/23/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023] Open
Abstract
Axonal transport is the essential process by which neurons actively traffic a variety of cargoes between the cell soma and axon terminals. Accordingly, dysfunctional axonal transport is linked to many nervous system conditions. Therefore, being able to image and quantify this dynamic process in live neurons of animal disease models is beneficial for understanding neuropathology and testing new therapies at the preclinical level. As such, intravital approaches have been developed to assess cargo movement in the hindlimb sciatic nerves of live, anaesthetised mice. Here, we describe an adapted method for in vivo imaging of axonal transport in intact median and ulnar nerves of the rodent forelimb. Injection of a fluorescently labelled and non-toxic fragment of tetanus neurotoxin (HCT) into the mouse forepaw permits the identification of signalling endosomes in intact axons of median and ulnar nerves. Through immunofluorescent analysis of forelimb lumbrical muscles and median/ulnar nerves, we confirmed that HCT is taken up at motor nerve terminals and predominantly locates to motor axons. We then showed that the baseline trafficking of signalling endosomes is similar between the median/ulnar nerves and the sciatic nerve in adult wild-type mice. Importantly, this adapted method can be readily tailored for assessment of additional cargoes, such as mitochondria. By measuring transport in forelimb and hindlimb nerves, comparative anatomical and functional analyses can be performed in rodent disease models to aid our understanding of peripheral nerve disease pathogenesis and response to injury.
Collapse
|
22
|
Negro S, Lauria F, Stazi M, Tebaldi T, D’Este G, Pirazzini M, Megighian A, Lessi F, Mazzanti CM, Sales G, Romualdi C, Fillo S, Lista F, Sleigh JN, Tosolini AP, Schiavo G, Viero G, Rigoni M. Hydrogen peroxide induced by nerve injury promotes axon regeneration via connective tissue growth factor. Acta Neuropathol Commun 2022; 10:189. [PMID: 36567321 PMCID: PMC9791753 DOI: 10.1186/s40478-022-01495-5] [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: 08/26/2022] [Accepted: 12/12/2022] [Indexed: 12/26/2022] Open
Abstract
Regeneration of the neuromuscular junction (NMJ) leverages on extensive exchange of factors released from motor axon terminals (MATs), muscle fibers and perisynaptic Schwann cells (PSCs), among which hydrogen peroxide (H2O2) is a major pro-regenerative signal. To identify critical determinants of NMJ remodeling in response to injury, we performed temporal transcriptional profiling of NMJs from 2 month-old mice during MAT degeneration/regeneration, and cross-referenced the differentially expressed genes with those elicited by H2O2 in SCs. We identified an enrichment in extracellular matrix (ECM) transcripts, including Connective Tissue Growth Factor (Ctgf), which is usually expressed during development. We discovered that Ctgf levels are increased in a Yes-associated protein (YAP)-dependent fashion in response to rapid, local H2O2 signaling generated by stressed mitochondria in the injured sciatic nerve, a finding highlighting the importance of signals triggered by mechanical force to motor nerve repair. Through sequestration of Ctgf or inactivation of H2O2, we delayed the recovery of neuromuscular function by impairing SC migration and, in turn, axon-oriented re-growth. These data indicate that H2O2 and its downstream effector Ctgf are pro-regenerative factors that enable axonal growth, and reveal a striking ECM remodeling process during nerve regeneration upon local H2O2 signaling. Our study identifies key transcriptomic changes at the regenerating NMJ, providing a rich source of pro-regenerative factors with potential for alleviating the consequences of peripheral nerve injuries.
Collapse
Affiliation(s)
- Samuele Negro
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy ,grid.5608.b0000 0004 1757 3470U.O.C. Clinica Neurologica, Azienda Ospedale, University of Padua, 35128 Padua, Italy
| | - Fabio Lauria
- grid.419463.d0000 0004 1756 3731Institute of Biophysics, CNR Unit at Trento, 38123 Povo, Italy
| | - Marco Stazi
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Toma Tebaldi
- grid.11696.390000 0004 1937 0351Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Povo, Italy ,grid.47100.320000000419368710Section of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Giorgia D’Este
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Marco Pirazzini
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy ,grid.5608.b0000 0004 1757 3470Myology Center (CIR-Myo), University of Padua, 35129 Padua, Italy
| | - Aram Megighian
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy ,grid.5608.b0000 0004 1757 3470Padua Neuroscience Center, University of Padua, 35131 Padua, Italy
| | - Francesca Lessi
- Laboratory of Genomics, Pisa Science Foundation, 56017 San Giuliano Terme, Italy
| | - Chiara M. Mazzanti
- Laboratory of Genomics, Pisa Science Foundation, 56017 San Giuliano Terme, Italy
| | - Gabriele Sales
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padua, 35131 Padua, Italy
| | - Chiara Romualdi
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padua, 35131 Padua, Italy
| | - Silvia Fillo
- grid.470599.60000 0004 1760 920XCenter of Medical and Veterinary Research of the Ministry of Defence, 00184 Rome, Italy
| | - Florigio Lista
- grid.470599.60000 0004 1760 920XCenter of Medical and Veterinary Research of the Ministry of Defence, 00184 Rome, Italy
| | - James N. Sleigh
- grid.83440.3b0000000121901201Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UK Dementia Research Institute, University College London, London, WC1E 6BT UK
| | - Andrew P. Tosolini
- grid.83440.3b0000000121901201Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG UK
| | - Giampietro Schiavo
- grid.83440.3b0000000121901201Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UK Dementia Research Institute, University College London, London, WC1E 6BT UK
| | - Gabriella Viero
- grid.419463.d0000 0004 1756 3731Institute of Biophysics, CNR Unit at Trento, 38123 Povo, Italy
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, 35131, Padua, Italy. .,Myology Center (CIR-Myo), University of Padua, 35129, Padua, Italy.
| |
Collapse
|
23
|
Sleigh JN. Editorial: Peripheral nerve anatomy in health and disease. J Anat 2022; 241:1083-1088. [PMID: 36226698 PMCID: PMC9558158 DOI: 10.1111/joa.13746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/27/2022] Open
Affiliation(s)
- James N. Sleigh
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research InstituteUniversity College LondonLondonUK
| |
Collapse
|
24
|
Gelon PA, Dutchak PA, Sephton CF. Synaptic dysfunction in ALS and FTD: anatomical and molecular changes provide insights into mechanisms of disease. Front Mol Neurosci 2022; 15:1000183. [PMID: 36263379 PMCID: PMC9575515 DOI: 10.3389/fnmol.2022.1000183] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022] Open
Abstract
Synaptic loss is a pathological feature of all neurodegenerative diseases including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). ALS is a disease of the cortical and spinal motor neurons resulting in fatal paralysis due to denervation of muscles. FTD is a form of dementia that primarily affects brain regions controlling cognition, language and behavior. Once classified as two distinct diseases, ALS and FTD are now considered as part of a common disease spectrum based on overlapping clinical, pathological and genetic evidence. At the cellular level, aggregation of common proteins and overlapping gene susceptibilities are shared in both ALS and FTD. Despite the convergence of these two fields of research, the underlying disease mechanisms remain elusive. However, recent discovers from ALS and FTD patient studies and models of ALS/FTD strongly suggests that synaptic dysfunction is an early event in the disease process and a unifying hallmark of these diseases. This review provides a summary of the reported anatomical and cellular changes that occur in cortical and spinal motor neurons in ALS and FTD tissues and models of disease. We also highlight studies that identify changes in the proteome and transcriptome of ALS and FTD models and provide a conceptual overview of the processes that contribute to synaptic dysfunction in these diseases. Due to space limitations and the vast number of publications in the ALS and FTD fields, many articles have not been discussed in this review. As such, this review focuses on the three most common shared mutations in ALS and FTD, the hexanucleuotide repeat expansion within intron 1 of chromosome 9 open reading frame 72 (C9ORF72), transactive response DNA binding protein 43 (TARDBP or TDP-43) and fused in sarcoma (FUS), with the intention of highlighting common pathways that promote synaptic dysfunction in the ALS-FTD disease spectrum.
Collapse
|
25
|
Bai L, Tu WY, Xiao Y, Zhang K, Shen C. Motoneurons innervation determines the distinct gene expressions in multinucleated myofibers. Cell Biosci 2022; 12:140. [PMID: 36042463 PMCID: PMC9429338 DOI: 10.1186/s13578-022-00876-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 08/09/2022] [Indexed: 11/22/2022] Open
Abstract
Background Neuromuscular junctions (NMJs) are peripheral synapses connecting motoneurons and skeletal myofibers. At the postsynaptic side in myofibers, acetylcholine receptor (AChR) proteins are clustered by the neuronal agrin signal. Meanwhile, several nuclei in each myofiber are specially enriched around the NMJ for postsynaptic gene transcription. It remains mysterious that how gene expressions in these synaptic nuclei are systematically regulated, especially by motoneurons. Results We found that synaptic nuclei have a distinctive chromatin structure and gene expression profiling. Synaptic nuclei are formed during NMJ development and maintained by motoneuron innervation. Transcriptome analysis revealed that motoneuron innervation determines the distinct expression patterns in the synaptic region and non-synaptic region in each multinucleated myofiber, probably through epigenetic regulation. Myonuclei in synaptic and non-synaptic regions have different responses to denervation. Weighted gene co-expression network analysis revealed that the histone lysine demethylases Kdm1a is a negative regulator of synaptic gene expression. Inhibition of Kdm1a promotes AChR expression but impairs motor functions. Conclusion These results demonstrate that motoneurons innervation determines the distinct gene expressions in multinucleated myofibers. Thus, dysregulation of nerve-controlled chromatin structure and muscle gene expression might cause muscle weakness and atrophy in motoneuron degenerative disorders. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00876-6.
Collapse
|
26
|
Tosolini AP, Sleigh JN, Surana S, Rhymes ER, Cahalan SD, Schiavo G. BDNF-dependent modulation of axonal transport is selectively impaired in ALS. Acta Neuropathol Commun 2022; 10:121. [PMID: 35996201 PMCID: PMC9396851 DOI: 10.1186/s40478-022-01418-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 02/08/2023] Open
Abstract
Axonal transport ensures long-range delivery of essential cargoes between proximal and distal compartments, and is needed for neuronal development, function, and survival. Deficits in axonal transport have been detected at pre-symptomatic stages in the SOD1G93A and TDP-43M337V mouse models of amyotrophic lateral sclerosis (ALS), suggesting that impairments in this critical process are fundamental for disease pathogenesis. Strikingly, in ALS, fast motor neurons (FMNs) degenerate first whereas slow motor neurons (SMNs) are more resistant, and this is a currently unexplained phenomenon. The main aim of this investigation was to determine the effects of brain-derived neurotrophic factor (BDNF) on in vivo axonal transport in different α-motor neuron (MN) subtypes in wild-type (WT) and SOD1G93A mice. We report that despite displaying similar basal transport speeds, stimulation of wild-type MNs with BDNF enhances in vivo trafficking of signalling endosomes specifically in FMNs. This BDNF-mediated enhancement of transport was also observed in primary ventral horn neuronal cultures. However, FMNs display selective impairment of axonal transport in vivo in symptomatic SOD1G93A mice, and are refractory to BDNF stimulation, a phenotype that was also observed in primary embryonic SOD1G93A neurons. Furthermore, symptomatic SOD1G93A mice display upregulation of the classical non-pro-survival truncated TrkB and p75NTR receptors in muscles, sciatic nerves, and Schwann cells. Altogether, these data indicate that cell- and non-cell autonomous BDNF signalling is impaired in SOD1G93A MNs, thus identifying a new key deficit in ALS.
Collapse
Affiliation(s)
- Andrew P Tosolini
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK.
| | - James N Sleigh
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Sunaina Surana
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Elena R Rhymes
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK
| | - Stephen D Cahalan
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, University of London, London, NW1 0TU, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK.
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
27
|
Chakravorty A, Sharma A, Sheeba V, Manjithaya R. Glutamatergic Synapse Dysfunction in Drosophila Neuromuscular Junctions Can Be Rescued by Proteostasis Modulation. Front Mol Neurosci 2022; 15:842772. [PMID: 35909443 PMCID: PMC9337869 DOI: 10.3389/fnmol.2022.842772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the nervous system, and the Drosophila glutamatergic neuromuscular junctions (NMJs) offer a tractable platform to understand excitatory synapse biology both in health and disease. Synaptopathies are neurodegenerative diseases that are associated with synaptic dysfunction and often display compromised proteostasis. One such rare, progressive neurodegenerative condition, Spinocerebellar Ataxia Type 3 (SCA3) or Machado-Joseph Disease (MJD), is characterized by cerebellar ataxia, Parkinsonism, and degeneration of motor neuron synapses. While the polyQ repeat mutant protein ataxin-3 is implicated in MJD, it is unclear how it leads to impaired synaptic function. In this study, we indicated that a Drosophila model of MJD recapitulates characteristics of neurodegenerative disorders marked by motor neuron dysfunction. Expression of 78 polyQ repeats of mutant ataxin-3 protein in Drosophila motor neurons resulted in behavioral defects, such as impaired locomotion in both larval and adult stages. Furthermore, defects in eclosion and lifespan were observed in adult flies. Detailed characterization of larval glutamatergic neuromuscular junctions (NMJs) revealed defects in morphological features along with compromised NMJ functioning. Autophagy, one of the key proteostasis pathways, is known to be impaired in the case of several synaptopathies. Our study reveals that overexpression of the autophagy-related protein Atg8a rescued behavioral defects. Thus, we present a model for glutamatergic synapse dysfunction that recapitulates synaptic and behavioral deficits and show that it is an amenable system for carrying out genetic and chemical biology screens to identify potential therapeutic targets for synaptopathies.
Collapse
Affiliation(s)
- Anushka Chakravorty
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Ankit Sharma
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Vasu Sheeba
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
- *Correspondence: Vasu Sheeba
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
- Ravi Manjithaya
| |
Collapse
|
28
|
Melatonin Induces Autophagy in Amyotrophic Lateral Sclerosis Mice via Upregulation of SIRT1. Mol Neurobiol 2022; 59:4747-4760. [PMID: 35606613 DOI: 10.1007/s12035-022-02875-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 05/12/2022] [Indexed: 12/29/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is the neurodegenerative disease that leads to the motor dysfunction damaged by both upper and lower motor neurons. The etiology and pathogenesis of ALS hasn't completely been understood yet up to now, the current study suggests that autophagy plays an important role in the development of ALS. Meanwhile, melatonin is found to inhibit the progression of ALS. To this end, this study aimed to investigate the potential relation between melatonin and autophagy in ALS. The in vivo model of ALS was established to investigate the effects of melatonin in ALS. The mRNA expressions were performed to detect by RT-qPCR, and the protein levels were tested by western blot and immunofluorescence histochemistry staining. The inflammatory cytokine was applied to detect by ELISA. The results showed that melatonin dose-dependently reversed the ALS-induced survival time shortened, weight loss and rotating rod latency decrease. The expressions of both SIRT1 and Beclin-1 as well as the ratio of LC3II/LC3I were significantly upregulated in the ALS mice, while melatonin reversed the upregulation of both SIRT1 and Beclin-1 expression and LC3II/LC3I ratio in a dose-dependent manner. In contrast, melatonin dose-dependently significantly restored the ALS-induced downregulation of p62. Furthermore, SIRT1 silencing notably reduced the effect of melatonin on Beclin-1, LC3II/LC3I, and p62. Melatonin induced autophagy in the ALS mice via the upregulation of SIRT1. Thus, melatonin might act as a new agent for the treatment of ALS.
Collapse
|
29
|
Arslanbaeva L, Bisaglia M. Activation of the Nrf2 Pathway as a Therapeutic Strategy for ALS Treatment. Molecules 2022; 27:1471. [PMID: 35268572 PMCID: PMC8911691 DOI: 10.3390/molecules27051471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022] Open
Abstract
Amyotrophic lateral sclerosis is a progressive and fatal disease that causes motoneurons degeneration and functional impairment of voluntary muscles, with limited and poorly efficient therapies. Alterations in the Nrf2-ARE pathway are associated with ALS pathology and result in aberrant oxidative stress, making the stimulation of the Nrf2-mediated antioxidant response a promising therapeutic strategy in ALS to reduce oxidative stress. In this review, we first introduce the involvement of the Nrf2 pathway in the pathogenesis of ALS and the role played by astrocytes in modulating such a protective pathway. We then describe the currently developed activators of Nrf2, used in both preclinical animal models and clinical studies, taking into consideration their potentialities as well as the possible limitations associated with their use.
Collapse
Affiliation(s)
| | - Marco Bisaglia
- Department of Biology, University of Padua, 35131 Padua, Italy
- Center Study for Neurodegeneration (CESNE), University of Padua, 35131 Padua, Italy
| |
Collapse
|
30
|
Poulin-Brière A, Rezaei E, Pozzi S. Antibody-Based Therapeutic Interventions for Amyotrophic Lateral Sclerosis: A Systematic Literature Review. Front Neurosci 2021; 15:790114. [PMID: 34912191 PMCID: PMC8667723 DOI: 10.3389/fnins.2021.790114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a mid-life onset neurodegenerative disease that manifests its symptomatology with motor impairments and cognitive deficits overlapping with Frontotemporal Lobar Degeneration (FTLD). The etiology of ALS remains elusive, with various mechanisms and cellular targets implicated, and no treatment can reverse or stop the progression of the pathology. Therapeutic interventions based on passive immunization are gaining attention for neurodegenerative diseases, and FDA recently approved the first antibody-based approach for Alzheimer's disease. The present systematic review of the literature aims to highlight the efforts made over the past years at developing antibody-based strategies to cure ALS. Thirty-one original research papers have been selected where the therapeutic efficacy of antibodies were investigated and described in patients and animal models of ALS. Antibody-based interventions analyzed, target both extracellular molecules implicated in the pathology and intracellular pathogenic proteins known to drive the disease, such as SOD1, TDP-43 or C9ORF72 repeats expansions. The potentials and limitations of these therapeutic interventions have been described and discussed in the present review.
Collapse
Affiliation(s)
| | - Edris Rezaei
- Department of Psychiatry and Neuroscience, Laval University, Quebec, QC, Canada
| | - Silvia Pozzi
- Department of Psychiatry and Neuroscience, Laval University, Quebec, QC, Canada
- Cellular and Molecular Neuroscience Division, CERVO Brain Research Centre, Quebec, QC, Canada
| |
Collapse
|
31
|
Hörner SJ, Couturier N, Bruch R, Koch P, Hafner M, Rudolf R. hiPSC-Derived Schwann Cells Influence Myogenic Differentiation in Neuromuscular Cocultures. Cells 2021; 10:cells10123292. [PMID: 34943800 PMCID: PMC8699767 DOI: 10.3390/cells10123292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 12/13/2022] Open
Abstract
Motoneurons, skeletal muscle fibers, and Schwann cells form synapses, termed neuromuscular junctions (NMJs). These control voluntary body movement and are affected in numerous neuromuscular diseases. Therefore, a variety of NMJ in vitro models have been explored to enable mechanistic and pharmacological studies. So far, selective integration of Schwann cells in these models has been hampered, due to technical limitations. Here we present robust protocols for derivation of Schwann cells from human induced pluripotent stem cells (hiPSC) and their coculture with hiPSC-derived motoneurons and C2C12 muscle cells. Upon differentiation with tuned BMP signaling, Schwann cells expressed marker proteins, S100b, Gap43, vimentin, and myelin protein zero. Furthermore, they displayed typical spindle-shaped morphologies with long processes, which often aligned with motoneuron axons. Inclusion of Schwann cells in coculture experiments with hiPSC-derived motoneurons and C2C12 myoblasts enhanced myotube growth and affected size and number of acetylcholine receptor plaques on myotubes. Altogether, these data argue for the availability of a consistent differentiation protocol for Schwann cells and their amenability for functional integration into neuromuscular in vitro models, fostering future studies of neuromuscular mechanisms and disease.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
| | - Roman Bruch
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
| | - Philipp Koch
- Central Institute of Mental Health, Medical Faculty Mannheim of Heidelberg University, 68159 Mannheim, Germany;
- Hector Institute for Translational Brain Research (HITBR gGmbH), 68159 Mannheim, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, 68163 Mannheim, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, 68163 Mannheim, Germany
- Correspondence:
| |
Collapse
|