101
|
Strickland JB, Davis-Anderson K, Micheva-Viteva S, Twary S, Iyer R, Harris JF, Solomon EA. Optimization of Application-Driven Development of In Vitro Neuromuscular Junction Models. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:1180-1191. [PMID: 35018825 PMCID: PMC9805869 DOI: 10.1089/ten.teb.2021.0204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Neuromuscular junctions (NMJs) are specialized synapses responsible for signal transduction between motor neurons (MNs) and skeletal muscle tissue. Malfunction at this site can result from developmental disorders, toxic environmental exposures, and neurodegenerative diseases leading to severe neurological dysfunction. Exploring these conditions in human or animal subjects is restricted by ethical concerns and confounding environmental factors. Therefore, in vitro NMJ models provide exciting opportunities for advancements in tissue engineering. In the last two decades, multiple NMJ prototypes and platforms have been reported, and each model system design is strongly tied to a specific application: exploring developmental physiology, disease modeling, or high-throughput screening. Directing the differentiation of stem cells into mature MNs and/or skeletal muscle for NMJ modeling has provided critical cues to recapitulate early-stage development. Patient-derived inducible pluripotent stem cells provide a personalized approach to investigating NMJ disease, especially when disease etiology cannot be resolved down to a specific gene mutation. Having reproducible NMJ culture replicates is useful for high-throughput screening to evaluate drug toxicity and determine the impact of environmental threat exposures. Cutting-edge bioengineering techniques have propelled this field forward with innovative microfabrication and design approaches allowing both two-dimensional and three-dimensional NMJ culture models. Many of these NMJ systems require further validation for broader application by regulatory agencies, pharmaceutical companies, and the general research community. In this summary, we present a comprehensive review on the current state-of-art research in NMJ models and discuss their ability to provide valuable insight into cell and tissue interactions. Impact statement In vitro neuromuscular junction (NMJ) models reveal the specialized mechanisms of communication between neurons and muscle tissue. This site can be disrupted by developmental disorders, toxic environmental exposures, or neurodegenerative diseases, which often lead to fatal outcomes and is therefore of critical importance to the medical community. Many bioengineering approaches for in vitro NMJ modeling have been designed to mimic development and disease; other approaches include in vitro NMJ models for high-throughput toxicology screening, providing a platform to limit or replace animal testing. This review describes various NMJ applications and the bioengineering advancements allowing for human NMJ characteristics to be more accurately recapitulated. While the extensive range of NMJ device structures has hindered standardization attempts, there is still a need to harmonize these devices for broader application and to continue advancing the field of NMJ modeling.
Collapse
Affiliation(s)
- Julie B. Strickland
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Katie Davis-Anderson
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | | - Scott Twary
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Rashi Iyer
- Information System and Modeling, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | | - Emilia A. Solomon
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA.,Address correspondence to: Emilia A. Solomon, PhD, Bioscience Division, Los Alamos National Laboratory, PO Box 1663 MS M888, Los Alamos, NM 87545, USA
| |
Collapse
|
102
|
Morotti M, Gaeta A, Limatola C, Catalano M, Di Castro MA, Grassi F. Early Developmental Changes of Muscle Acetylcholine Receptors Are Little Influenced by Dystrophin Absence in mdx Mouse. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111861. [PMID: 36430996 PMCID: PMC9696329 DOI: 10.3390/life12111861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Dystrophin is a cytoskeletal protein contributing to the organization of the neuromuscular junction. In Duchenne muscular dystrophy, due to dystrophin absence, the distribution of endplate acetylcholine receptors (AChRs) becomes disorganized. It is still debated whether this is due to the absence of dystrophin or to the repeated damage/regeneration cycles typical of dystrophic muscle. We addressed this controversy studying the endplate in the first 3 postnatal weeks, when muscle damage in dystrophic (mdx) mice is minimal. By synaptic and extra-synaptic patch-clamp recordings in acutely dissociated mdx and wt muscle fibers, we recorded unitary events due to openings of AChR-channels containing the γ and ε subunit. We also examined AChR distribution at the endplate by immunofluorescence assays. No differences between wt and mdx fibers were found in the γ/ε switch, nor in the AChR organization at the endplates up to 21 postnatal days. Conversely, we detected a delayed appearance and disappearance of patches with high channel opening frequency in mdx fibers. Our data emphasize that the innervation-dependent γ/ε switch and AChR organization in the endplate are not affected by the absence of dystrophin, while extra-synaptic AChR cluster formation and disassembly could be differentially regulated in mdx mice.
Collapse
Affiliation(s)
- Marta Morotti
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandro Gaeta
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - Cristina Limatola
- Laboratory Affiliated to Istituto Pasteur Italia, Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, 86077 Pozzilli, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Amalia Di Castro
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesca Grassi
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
103
|
A link between agrin signalling and Ca v3.2 at the neuromuscular junction in spinal muscular atrophy. Sci Rep 2022; 12:18960. [PMID: 36347955 PMCID: PMC9643518 DOI: 10.1038/s41598-022-23703-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
SMN protein deficiency causes motoneuron disease spinal muscular atrophy (SMA). SMN-based therapies improve patient motor symptoms to variable degrees. An early hallmark of SMA is the perturbation of the neuromuscular junction (NMJ), a synapse between a motoneuron and muscle cell. NMJ formation depends on acetylcholine receptor (AChR) clustering triggered by agrin and its co-receptors lipoprotein receptor-related protein 4 (LRP4) and transmembrane muscle-specific kinase (MuSK) signalling pathway. We have previously shown that flunarizine improves NMJs in SMA model mice, but the mechanisms remain elusive. We show here that flunarizine promotes AChR clustering in cell-autonomous, dose- and agrin-dependent manners in C2C12 myotubes. This is associated with an increase in protein levels of LRP4, integrin-beta-1 and alpha-dystroglycan, three agrin co-receptors. Furthermore, flunarizine enhances MuSK interaction with integrin-beta-1 and phosphotyrosines. Moreover, the drug acts on the expression and splicing of Agrn and Cacna1h genes in a muscle-specific manner. We reveal that the Cacna1h encoded protein Cav3.2 closely associates in vitro with the agrin co-receptor LRP4. In vivo, it is enriched nearby NMJs during neonatal development and the drug increases this immunolabelling in SMA muscles. Thus, flunarizine modulates key players of the NMJ and identifies Cav3.2 as a new protein involved in the NMJ biology.
Collapse
|
104
|
Xu J, Zhu J, Li Y, Yao Y, Xuan A, Li D, Yu T, Zhu D. Three-dimensional mapping reveals heterochronic development of the neuromuscular system in postnatal mouse skeletal muscles. Commun Biol 2022; 5:1200. [PMID: 36347940 PMCID: PMC9643545 DOI: 10.1038/s42003-022-04159-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
The development of the neuromuscular system, including muscle growth and intramuscular neural development, in addition to central nervous system maturation, determines motor ability improvement. Motor development occurs asynchronously from cephalic to caudal. However, whether the structural development of different muscles is heterochronic is unclear. Here, based on the characteristics of motor behavior in postnatal mice, we examined the 3D structural features of the neuromuscular system in different muscles by combining tissue clearing with optical imaging techniques. Quantitative analyses of the structural data and related mRNA expression revealed that there was continued myofiber hyperplasia of the forelimb and hindlimb muscles until around postnatal day 3 (P3) and P6, respectively, as well as continued axonal arborization and neuromuscular junction formation until around P3 and P9, respectively; feature alterations of the cervical muscle ended at birth. Such structural heterochrony of muscles in different body parts corresponds to their motor function. Structural data on the neuromuscular system of neonatal muscles provide a 3D perspective in the understanding of the structural status during motor development.
Collapse
Affiliation(s)
- Jianyi Xu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
- Optics Valley Laboratory, 430074, Wuhan, Hubei, China
| | - Jingtan Zhu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
- Optics Valley Laboratory, 430074, Wuhan, Hubei, China
| | - Yusha Li
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
- Optics Valley Laboratory, 430074, Wuhan, Hubei, China
| | - Yingtao Yao
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
- Optics Valley Laboratory, 430074, Wuhan, Hubei, China
| | - Ang Xuan
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
- Optics Valley Laboratory, 430074, Wuhan, Hubei, China
| | - Dongyu Li
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China
- Optics Valley Laboratory, 430074, Wuhan, Hubei, China
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China.
- Optics Valley Laboratory, 430074, Wuhan, Hubei, China.
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics - MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics - Advanced Biomedical Imaging Facility, Huazhong University of Science and Technology, 430074, Wuhan, Hubei, China.
- Optics Valley Laboratory, 430074, Wuhan, Hubei, China.
| |
Collapse
|
105
|
Volpe P, Bosutti A, Nori A, Filadi R, Gherardi G, Trautmann G, Furlan S, Massaria G, Sciancalepore M, Megighian A, Caccin P, Bernareggi A, Salanova M, Sacchetto R, Sandonà D, Pizzo P, Lorenzon P. Nerve-dependent distribution of subsynaptic type 1 inositol 1,4,5-trisphosphate receptor at the neuromuscular junction. J Gen Physiol 2022; 154:213498. [PMID: 36149386 PMCID: PMC9513380 DOI: 10.1085/jgp.202213128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 11/20/2022] Open
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) are enriched at postsynaptic membrane compartments of the neuromuscular junction (NMJ), surrounding the subsynaptic nuclei and close to nicotinic acetylcholine receptors (nAChRs) of the motor endplate. At the endplate level, it has been proposed that nerve-dependent electrical activity might trigger IP3-associated, local Ca2+ signals not only involved in excitation-transcription (ET) coupling but also crucial to the development and stabilization of the NMJ itself. The present study was undertaken to examine whether denervation affects the subsynaptic IP3R distribution in skeletal muscles and which are the underlying mechanisms. Fluorescence microscopy, carried out on in vivo denervated muscles (following sciatectomy) and in vitro denervated skeletal muscle fibers from flexor digitorum brevis (FDB), indicates that denervation causes a reduction in the subsynaptic IP3R1-stained region, and such a decrease appears to be determined by the lack of muscle electrical activity, as judged by partial reversal upon field electrical stimulation of in vitro denervated skeletal muscle fibers.
Collapse
Affiliation(s)
- Pompeo Volpe
- Department of Biomedical Sciences and Interdepartmental Research Center of Myology (cirMYO), University of Padova, Padova, Italy
- Correspondence to Pompeo Volpe:
| | | | - Alessandra Nori
- Department of Biomedical Sciences and Interdepartmental Research Center of Myology (cirMYO), University of Padova, Padova, Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences and Interdepartmental Research Center of Myology (cirMYO), University of Padova, Padova, Italy
- National Research Council, Neuroscience Institute, Padova, Italy
| | - Gaia Gherardi
- Department of Biomedical Sciences and Interdepartmental Research Center of Myology (cirMYO), University of Padova, Padova, Italy
| | - Gabor Trautmann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Integrative Neuroanatomy, Berlin, Germany
| | - Sandra Furlan
- National Research Council, Neuroscience Institute, Padova, Italy
| | | | | | - Aram Megighian
- Department of Biomedical Sciences and Interdepartmental Research Center of Myology (cirMYO), University of Padova, Padova, Italy
| | - Paola Caccin
- Department of Biomedical Sciences and Interdepartmental Research Center of Myology (cirMYO), University of Padova, Padova, Italy
| | | | - Michele Salanova
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Integrative Neuroanatomy, Berlin, Germany
- Neuromuscular Signaling, Center of Space Medicine Berlin, Berlin, Germany
| | - Roberta Sacchetto
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Dorianna Sandonà
- Department of Biomedical Sciences and Interdepartmental Research Center of Myology (cirMYO), University of Padova, Padova, Italy
| | - Paola Pizzo
- Department of Biomedical Sciences and Interdepartmental Research Center of Myology (cirMYO), University of Padova, Padova, Italy
- National Research Council, Neuroscience Institute, Padova, Italy
| | - Paola Lorenzon
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
106
|
Negro S, Pirazzini M, Rigoni M. Models and methods to study Schwann cells. J Anat 2022; 241:1235-1258. [PMID: 34988978 PMCID: PMC9558160 DOI: 10.1111/joa.13606] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
Schwann cells (SCs) are fundamental components of the peripheral nervous system (PNS) of all vertebrates and play essential roles in development, maintenance, function, and regeneration of peripheral nerves. There are distinct populations of SCs including: (1) myelinating SCs that ensheath axons by a specialized plasma membrane, called myelin, which enhances the conduction of electric impulses; (2) non-myelinating SCs, including Remak SCs, which wrap bundles of multiple axons of small caliber, and perysinaptic SCs (PSCs), associated with motor axon terminals at the neuromuscular junction (NMJ). All types of SCs contribute to PNS regeneration through striking morphological and functional changes in response to nerve injury, are affected in peripheral neuropathies and show abnormalities and a diminished plasticity during aging. Therefore, methodological approaches to study and manipulate SCs in physiological and pathophysiological conditions are crucial to expand the present knowledge on SC biology and to devise new therapeutic strategies to counteract neurodegenerative conditions and age-derived denervation. We present here an updated overview of traditional and emerging methodologies for the study of SCs for scientists approaching this research field.
Collapse
Affiliation(s)
- Samuele Negro
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
| | - Marco Pirazzini
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| | - Michela Rigoni
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| |
Collapse
|
107
|
Sarto F, Stashuk DW, Franchi MV, Monti E, Zampieri S, Valli G, Sirago G, Candia J, Hartnell LM, Paganini M, McPhee JS, De Vito G, Ferrucci L, Reggiani C, Narici MV. Effects of short-term unloading and active recovery on human motor unit properties, neuromuscular junction transmission and transcriptomic profile. J Physiol 2022; 600:4731-4751. [PMID: 36071599 PMCID: PMC9828768 DOI: 10.1113/jp283381] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/26/2022] [Indexed: 01/12/2023] Open
Abstract
Electrophysiological alterations of the neuromuscular junction (NMJ) and motor unit potential (MUP) with unloading are poorly studied. We aimed to investigate these aspects and the underlying molecular mechanisms with short-term unloading and active recovery (AR). Eleven healthy males underwent a 10-day unilateral lower limb suspension (ULLS) period, followed by 21-day AR based on resistance exercise. Quadriceps femoris (QF) cross-sectional area (CSA) and isometric maximum voluntary contraction (MVC) were evaluated. Intramuscular electromyographic recordings were obtained during 10% and 25% MVC isometric contractions from the vastus lateralis (VL). Biomarkers of NMJ molecular instability (serum c-terminal agrin fragment, CAF), axonal damage (neurofilament light chain) and denervation status were assessed from blood samples and VL biopsies. NMJ and ion channel transcriptomic profiles were investigated by RNA-sequencing. QF CSA and MVC decreased with ULLS. Increased CAF and altered NMJ transcriptome with unloading suggested the emergence of NMJ molecular instability, which was not associated with impaired NMJ transmission stability. Instead, increased MUP complexity and decreased motor unit firing rates were found after ULLS. Downregulation of ion channel gene expression was found together with increased neurofilament light chain concentration and partial denervation. The AR period restored most of these neuromuscular alterations. In conclusion, the human NMJ is destabilized at the molecular level but shows functional resilience to a 10-day unloading period at least at relatively low contraction intensities. However, MUP properties are altered by ULLS, possibly due to alterations in ion channel dynamics and initial axonal damage and denervation. These changes are fully reversed by 21 days of AR. KEY POINTS: We used integrative electrophysiological and molecular approaches to comprehensively investigate changes in neuromuscular integrity and function after a 10-day unilateral lower limb suspension (ULLS), followed by 21 days of active recovery in young healthy men, with a particular focus on neuromuscular junction (NMJ) and motor unit potential (MUP) properties alterations. After 10-day ULLS, we found significant NMJ molecular alterations in the absence of NMJ transmission stability impairment. These findings suggest that the human NMJ is functionally resilient against insults and stresses induced by short-term disuse at least at relatively low contraction intensities, at which low-threshold, slow-type motor units are recruited. Intramuscular electromyography analysis revealed that unloading caused increased MUP complexity and decreased motor unit firing rates, and these alterations could be related to the observed changes in skeletal muscle ion channel pool and initial and partial signs of fibre denervation and axonal damage. The active recovery period restored these neuromuscular changes.
Collapse
Affiliation(s)
- Fabio Sarto
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Daniel W. Stashuk
- Department of Systems Design EngineeringUniversity of WaterlooOntarioCanada
| | - Martino V. Franchi
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly,CIR‐MYO Myology CenterUniversity of PadovaPadovaItaly
| | - Elena Monti
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Sandra Zampieri
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly,CIR‐MYO Myology CenterUniversity of PadovaPadovaItaly,Department of SurgeryOncology, and GastroenterologyUniversity of PadovaPadovaItaly
| | - Giacomo Valli
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Giuseppe Sirago
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Julián Candia
- Longitudinal Studies SectionTranslational Gerontology BranchNational Institute of AgingNational Institutes of HealthBaltimoreMDUSA
| | - Lisa M. Hartnell
- Longitudinal Studies SectionTranslational Gerontology BranchNational Institute of AgingNational Institutes of HealthBaltimoreMDUSA
| | - Matteo Paganini
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Jamie S. McPhee
- Department of Sport and Exercise SciencesManchester Metropolitan University Institute of SportManchesterUK
| | - Giuseppe De Vito
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly,CIR‐MYO Myology CenterUniversity of PadovaPadovaItaly
| | - Luigi Ferrucci
- Longitudinal Studies SectionTranslational Gerontology BranchNational Institute of AgingNational Institutes of HealthBaltimoreMDUSA
| | - Carlo Reggiani
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly,Science and Research Center KoperInstitute for Kinesiology ResearchKoperSlovenia
| | - Marco V. Narici
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly,CIR‐MYO Myology CenterUniversity of PadovaPadovaItaly,Science and Research Center KoperInstitute for Kinesiology ResearchKoperSlovenia
| |
Collapse
|
108
|
Medina‐Moreno A, Henríquez JP. Maturation of a postsynaptic domain: Role of small Rho GTPases in organising nicotinic acetylcholine receptor aggregates at the vertebrate neuromuscular junction. J Anat 2022; 241:1148-1156. [PMID: 34342888 PMCID: PMC9558164 DOI: 10.1111/joa.13526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
The neuromuscular junction (NMJ) is the peripheral synapse formed between a motor axon and a skeletal muscle fibre that allows muscle contraction and the coordinated movement in many species. A main hallmark of the mature NMJ is the assembly of nicotinic acetylcholine receptor (nAChR) aggregates in the muscle postsynaptic domain, that distributes in perfect apposition to presynaptic motor terminals. To assemble its unique functional architecture, initial embryonic NMJs undergo an early postnatal maturation process characterised by the transformation of homogenous nAChR-containing plaques to elaborate and branched pretzel-like structures. In spite of a detailed morphological characterisation, the molecular mechanisms controlling the intracellular scaffolding that organises a postsynaptic domain at the mature NMJ have not been fully elucidated. In this review, we integrate evidence of key processes and molecules that have shed light on our current understanding of the NMJ maturation process. On the one hand, we consider in vitro studies revealing the potential role of podosome-like structures to define discrete low nAChR-containing regions to consolidate a plaque-to-pretzel transition at the NMJ. On the other hand, we focus on in vitro and in vivo evidence demonstrating that members of the Ras homologous (Rho) protein family of small GTPases (small Rho GTPases) play indispensable roles on NMJ maturation by regulating the stability of nAChR aggregates. We combine this evidence to propose that small Rho GTPases are key players in the assembly of podosome-like structures that drive the postsynaptic maturation of vertebrate NMJs.
Collapse
Affiliation(s)
- Angelymar Medina‐Moreno
- Laboratory of Neuromuscular Studies (NeSt Lab)Department of Cell BiologyFaculty of Biological SciencesCenter for Advanced Microscopy (CMA BioBio)Universidad de ConcepciónConcepciónChile
| | - Juan Pablo Henríquez
- Laboratory of Neuromuscular Studies (NeSt Lab)Department of Cell BiologyFaculty of Biological SciencesCenter for Advanced Microscopy (CMA BioBio)Universidad de ConcepciónConcepciónChile
| |
Collapse
|
109
|
Costamagna D, Casters V, Beltrà M, Sampaolesi M, Van Campenhout A, Ortibus E, Desloovere K, Duelen R. Autologous iPSC-Derived Human Neuromuscular Junction to Model the Pathophysiology of Hereditary Spastic Paraplegia. Cells 2022; 11:3351. [PMID: 36359747 PMCID: PMC9655384 DOI: 10.3390/cells11213351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 08/27/2023] Open
Abstract
Hereditary spastic paraplegia (HSP) is a heterogeneous group of genetic neurodegenerative disorders, characterized by progressive lower limb spasticity and weakness resulting from retrograde axonal degeneration of motor neurons (MNs). Here, we generated in vitro human neuromuscular junctions (NMJs) from five HSP patient-specific induced pluripotent stem cell (hiPSC) lines, by means of microfluidic strategy, to model disease-relevant neuropathologic processes. The strength of our NMJ model lies in the generation of lower MNs and myotubes from autologous hiPSC origin, maintaining the genetic background of the HSP patient donors in both cell types and in the cellular organization due to the microfluidic devices. Three patients characterized by a mutation in the SPG3a gene, encoding the ATLASTIN GTPase 1 protein, and two patients with a mutation in the SPG4 gene, encoding the SPASTIN protein, were included in this study. Differentiation of the HSP-derived lines gave rise to lower MNs that could recapitulate pathological hallmarks, such as axonal swellings with accumulation of Acetyl-α-TUBULIN and reduction of SPASTIN levels. Furthermore, NMJs from HSP-derived lines were lower in number and in contact point complexity, denoting an impaired NMJ profile, also confirmed by some alterations in genes encoding for proteins associated with microtubules and responsible for axonal transport. Considering the complexity of HSP, these patient-derived neuronal and skeletal muscle cell co-cultures offer unique tools to study the pathologic mechanisms and explore novel treatment options for rescuing axonal defects and diverse cellular processes, including membrane trafficking, intracellular motility and protein degradation in HSP.
Collapse
Affiliation(s)
- Domiziana Costamagna
- Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Research Group for Neurorehabilitation, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Valérie Casters
- Research Group for Neurorehabilitation, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Marc Beltrà
- Department of Clinical and Biological Sciences, University of Torino, 10125 Torino, Italy
| | - Maurilio Sampaolesi
- Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Anja Van Campenhout
- Locomotor and Neurological Disorder, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Orthopedic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Els Ortibus
- Locomotor and Neurological Disorder, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
- Department of Pediatric Neurology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Kaat Desloovere
- Research Group for Neurorehabilitation, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium
- Clinical Motion Analysis Laboratory, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Robin Duelen
- Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
110
|
Qi Z, Wang S, Xuan A, Gu X, Deng J, Huang C, Zhang L, Yin X. MiR-142a-3p: A novel ACh receptor transcriptional regulator in association with peripheral nerve injury. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:325-336. [PMID: 36381585 PMCID: PMC9633872 DOI: 10.1016/j.omtn.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 10/12/2022] [Indexed: 12/15/2022]
Abstract
Long-term denervation leads to the disintegration of nicotinic acetylcholine receptor (nAChR) located at the endplate structure, which translates to deficits in functional activation despite nerve repair. Because of a lack of effective measures to protect AChR expression, we explored the effect of alterations in muscular miR-142a-3p on nAChR. In this study, we constructed a model of miR-142a-3p knockdown by transfecting a miR-142a-3p inhibitor short hairpin RNA (shRNA) into C2C12 myotubes, and we injected this miR-142a-3p inhibitor shRNA into the tibialis anterior (TA) muscle in uninjured mice and in denervated mice by transecting the sciatic nerve. Our results showed that miR-142a-3p knockdown led to an increased number and area of AChR clusters in myotubes in vitro and larger neuromuscular endplates in adult mice. Furthermore, miR-142a-3p knockdown delayed the disintegration of motor endplates after denervation. Last, upon miR-142a-3p knockdown in uninjured and denervated mice, we observed an increase in the mRNA levels of five AChR subunits as well as mRNAs of genes implicated in AChR transcription and AChR clustering. Together, these results suggest that miR-142a-3p may be a potential target for therapeutic intervention to prevent motor endplate degradation following peripheral nerve injury.
Collapse
Affiliation(s)
- Zhidan Qi
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Shen Wang
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Ang Xuan
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China,MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Gu
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Jin Deng
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Chen Huang
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Lei Zhang
- Electron Microscopy Analysis Laboratory, Medical and Health Analysis Center, Peking University, Beijing, China,Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiaofeng Yin
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China,Pizhou People’s Hospital, Jiangsu, China,Corresponding author Xiaofeng Yin, Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China.
| |
Collapse
|
111
|
SMN controls neuromuscular junction integrity through U7 snRNP. Cell Rep 2022; 40:111393. [PMID: 36130491 PMCID: PMC9533342 DOI: 10.1016/j.celrep.2022.111393] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/18/2022] [Accepted: 08/30/2022] [Indexed: 01/26/2023] Open
Abstract
The neuromuscular junction (NMJ) is an essential synapse whose loss is a key hallmark of the neurodegenerative disease spinal muscular atrophy (SMA). Here, we show that activity of the SMA-determining SMN protein in the assembly of U7 small nuclear ribonucleoprotein (snRNP)—which functions in the 3′-end processing of replication-dependent histone mRNAs—is required for NMJ integrity. Co-expression of U7-specific Lsm10 and Lsm11 proteins selectively enhances U7 snRNP assembly, corrects histone mRNA processing defects, and rescues key structural and functional abnormalities of neuromuscular pathology in SMA mice—including NMJ denervation, decreased synaptic transmission, and skeletal muscle atrophy. Furthermore, U7 snRNP dysfunction drives selective loss of the synaptic organizing protein Agrin at NMJs innervating vulnerable muscles of SMA mice. These findings reveal a direct contribution of U7 snRNP dysfunction to neuromuscular pathology in SMA and suggest a role for histone gene regulation in maintaining functional synaptic connections between motor neurons and muscles. NMJ denervation is a hallmark of SMA. Through selective restoration of U7 snRNP biogenesis in SMA mice, Tisdale et al. reveal a role for SMN-mediated U7 snRNP assembly and histone mRNA processing in controlling NMJ integrity through Agrin expression, uncovering RNA-mediated disease mechanisms and linking U7 function to neuromuscular development.
Collapse
|
112
|
Cahalan SD, Boehm I, Jones RA, Piercy RJ. Recognising the potential of large animals for modelling neuromuscular junction physiology and disease. J Anat 2022; 241:1120-1132. [PMID: 36056593 PMCID: PMC9558152 DOI: 10.1111/joa.13749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 12/28/2022] Open
Abstract
The aetiology and pathophysiology of many diseases of the motor unit remain poorly understood and the role of the neuromuscular junction (NMJ) in this group of disorders is particularly overlooked, especially in humans, when these diseases are comparatively rare. However, elucidating the development, function and degeneration of the NMJ is essential to uncover its contribution to neuromuscular disorders, and to explore potential therapeutic avenues to treat these devastating diseases. Until now, an understanding of the role of the NMJ in disease pathogenesis has been hindered by inherent differences between rodent and human NMJs: stark contrasts in body size and corresponding differences in associated axon length underpin some of the translational issues in animal models of neuromuscular disease. Comparative studies in large mammalian models, including examination of naturally occurring, highly prevalent animal diseases and evaluation of their treatment, might provide more relevant insights into the pathogenesis and therapy of equivalent human diseases. This review argues that large animal models offer great potential to enhance our understanding of the neuromuscular system in health and disease, and in particular, when dealing with diseases for which nerve length dependency might underly the pathogenesis.
Collapse
Affiliation(s)
- Stephen D Cahalan
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, University of London, London, UK
| | - Ines Boehm
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.,Biozentrum University of Basel, Basel, Switzerland
| | - Ross A Jones
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK.,Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Richard J Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, University of London, London, UK
| |
Collapse
|
113
|
Vemula SK, Kim SA, Muvavarirwa T, Bell JL, Whitman MC. Impaired Extraocular Muscle Innervation Is Present Before Eye Opening in a Mouse Model of Infantile Nystagmus Syndrome. Invest Ophthalmol Vis Sci 2022; 63:4. [PMID: 36083589 PMCID: PMC9469029 DOI: 10.1167/iovs.63.10.4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Purpose To determine if extraocular muscles (EOMs) from mice with nystagmus show abnormalities in myofiber composition and innervation, as seen in EOMs from human nystagmus patients, and to determine when in development those changes occur. Methods Balb/c albino mice were crossed to pigmented mice to generate heterozygous mice, which were mated to create experimental litters containing albinos and wild-type controls. Orbits were harvested from adult animals (12 weeks old); on postnatal day (P)0, P10, P14, and P21; and from 6-week-old animals. EOM sections were collected from the intraorbital portion of the muscles. Sections were immunostained for slow and fast myosin and for neuromuscular junctions (NMJs). The proportion of each myofiber subtype and the density and size of NMJs were quantified. Initial innervation patterns were assessed using whole-mount immunostaining of embryonic day (E)13.5 embryos expressing IslMN:GFP. Results Adult albino EOMs display an increased proportion of slow myofibers, larger slow myofibers, and a decreased density of NMJs—similar to human nystagmus patients. The percentage of NMJs on slow myofibers is also lower in albino animals. The initial innervation pattern of the incoming ocular motor neurons is normal in E13.5 albino embryos. Differences in the proportion of slow and fast myofiber subtypes are present as early as P14, and a lower percentage of NMJs on slow myofibers is present by P21. There is a lower density of NMJs on albino EOMs as early as P10, prior to eye opening. Conclusions Changes in NMJ development observed before eye opening indicate that nystagmus is not solely secondary to poor vision.
Collapse
Affiliation(s)
- Sampath Kumar Vemula
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Seoyoung A Kim
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Tapiwa Muvavarirwa
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Jessica L Bell
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Mary C Whitman
- Department of Ophthalmology, Boston Children's Hospital, Boston, Massachusetts, United States.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts, United States
| |
Collapse
|
114
|
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: 12] [Impact Index Per Article: 4.0] [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
|
115
|
Giakoumaki I, Pollock N, Aljuaid T, Sannicandro AJ, Alameddine M, Owen E, Myrtziou I, Ozanne SE, Kanakis I, Goljanek-Whysall K, Vasilaki A. Postnatal Protein Intake as a Determinant of Skeletal Muscle Structure and Function in Mice-A Pilot Study. Int J Mol Sci 2022; 23:8815. [PMID: 35955948 PMCID: PMC9369224 DOI: 10.3390/ijms23158815] [Citation(s) in RCA: 3] [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: 07/16/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 12/04/2022] Open
Abstract
Sarcopenia is characterised by an age-related decrease in the number of muscle fibres and additional weakening of the remaining fibres, resulting in a reduction in muscle mass and function. Many studies associate poor maternal nutrition during gestation and/or lactation with altered skeletal muscle homeostasis in the offspring and the development of sarcopenia. The aim of this study was to determine whether the musculoskeletal physiology in offspring born to mouse dams fed a low-protein diet during pregnancy was altered and whether any physiological changes could be modulated by the nutritional protein content in early postnatal stages. Thy1-YFP female mice were fed ad libitum on either a normal (20%) or a low-protein (5%) diet. Newborn pups were cross-fostered to different lactating dams (maintained on a 20% or 5% diet) to generate three groups analysed at weaning (21 days): Normal-to-Normal (NN), Normal-to-Low (NL) and Low-to-Normal (LN). Further offspring were maintained ad libitum on the same diet as during lactation until 12 weeks of age, creating another three groups (NNN, NLL, LNN). Mice on a low protein diet postnatally (NL, NLL) exhibited a significant reduction in body and muscle weight persisting up to 12 weeks, unlike mice on a low protein diet only prenatally (LN, LNN). Muscle fibre size was reduced in mice from the NL but not LN group, showing recovery at 12 weeks of age. Muscle force was reduced in NLL mice, concomitant with changes in the NMJ site and changes in atrophy-related and myosin genes. In addition, μCT scans of mouse tibiae at 12 weeks of age revealed changes in bone mass and morphology, resulting in a higher bone mass in the NLL group than the control NNN group. Finally, changes in the expression of miR-133 in the muscle of NLL mice suggest a regulatory role for this microRNA in muscle development in response to postnatal diet changes. Overall, this data shows that a low maternal protein diet and early postnatal life low-protein intake in mice can impact skeletal muscle physiology and function in early life while postnatal low protein diet favours bone integrity in adulthood.
Collapse
Affiliation(s)
- Ifigeneia Giakoumaki
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Natalie Pollock
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
- The MRC—Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool L7 8TX, UK
| | - Turki Aljuaid
- Department of Physiology, School of Medicine and REMEDI, CMNHS, NUI Galway, H91 TK33 Galway, Ireland
- Department of Biotechnology, College of Science, Taif University, Taif 21944, Saudi Arabia
| | - Anthony J. Sannicandro
- Department of Physiology, School of Medicine and REMEDI, CMNHS, NUI Galway, H91 TK33 Galway, Ireland
| | - Moussira Alameddine
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Euan Owen
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Ioanna Myrtziou
- Chester Medical School, University of Chester, Bache Hall, Countess View, Chester CH2 1BR, UK
| | - Susan E. Ozanne
- University of Cambridge MRC Metabolic Diseases Unit and Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke’s Hospital Cambridge, Cambridge CB2 0QQ, UK
| | - Ioannis Kanakis
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Chester Medical School, University of Chester, Bache Hall, Countess View, Chester CH2 1BR, UK
| | - Katarzyna Goljanek-Whysall
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
- The MRC—Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool L7 8TX, UK
- Department of Physiology, School of Medicine and REMEDI, CMNHS, NUI Galway, H91 TK33 Galway, Ireland
| | - Aphrodite Vasilaki
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, Faculty of Health & Life Sciences, University of Liverpool, Liverpool L7 8TX, UK
- The MRC—Versus Arthritis Centre for Integrated Research into Musculoskeletal Ageing (CIMA), University of Liverpool, Liverpool L7 8TX, UK
| |
Collapse
|
116
|
vanLieshout TL, Stouth DW, Hartel NG, Vasam G, Ng SY, Webb EK, Rebalka IA, Mikhail AI, Graham NA, Menzies KJ, Hawke TJ, Ljubicic V. The CARM1 transcriptome and arginine methylproteome mediate skeletal muscle integrative biology. Mol Metab 2022; 64:101555. [PMID: 35872306 PMCID: PMC9379683 DOI: 10.1016/j.molmet.2022.101555] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Coactivator-associated arginine methyltransferase 1 (CARM1) catalyzes the methylation of arginine residues on target proteins to regulate critical processes in health and disease. A mechanistic understanding of the role(s) of CARM1 in skeletal muscle biology is only gradually emerging. The purpose of this study was to elucidate the function of CARM1 in regulating the maintenance and plasticity of skeletal muscle. METHODS We used transcriptomic, methylproteomic, molecular, functional, and integrative physiological approaches to determine the specific impact of CARM1 in muscle homeostasis. RESULTS Our data defines the occurrence of arginine methylation in skeletal muscle and demonstrates that this mark occurs on par with phosphorylation and ubiquitination. CARM1 skeletal muscle-specific knockout (mKO) mice displayed altered transcriptomic and arginine methylproteomic signatures with molecular and functional outcomes confirming remodeled skeletal muscle contractile and neuromuscular junction characteristics, which presaged decreased exercise tolerance. Moreover, CARM1 regulates AMPK-PGC-1α signalling during acute conditions of activity-induced muscle plasticity. CONCLUSIONS This study uncovers the broad impact of CARM1 in the maintenance and remodelling of skeletal muscle biology.
Collapse
Affiliation(s)
| | - Derek W Stouth
- Department of Kinesiology, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Nicolas G Hartel
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA
| | - Goutham Vasam
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Sean Y Ng
- Department of Kinesiology, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Erin K Webb
- Department of Kinesiology, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Irena A Rebalka
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Andrew I Mikhail
- Department of Kinesiology, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Nicholas A Graham
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, 90089, USA
| | - Keir J Menzies
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, K1H 8M5, Canada; Ottawa Institute of Systems Biology and the Centre for Neuromuscular Disease, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Rd, K1H 8M5, Ottawa, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton, ON, L8S 4L8, Canada.
| |
Collapse
|
117
|
Hughes DC, Baehr LM, Waddell DS, Sharples AP, Bodine SC. Ubiquitin Ligases in Longevity and Aging Skeletal Muscle. Int J Mol Sci 2022; 23:7602. [PMID: 35886949 PMCID: PMC9315556 DOI: 10.3390/ijms23147602] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/07/2022] Open
Abstract
The development and prevalence of diseases associated with aging presents a global health burden on society. One hallmark of aging is the loss of proteostasis which is caused in part by alterations to the ubiquitin-proteasome system (UPS) and lysosome-autophagy system leading to impaired function and maintenance of mass in tissues such as skeletal muscle. In the instance of skeletal muscle, the impairment of function occurs early in the aging process and is dependent on proteostatic mechanisms. The UPS plays a pivotal role in degradation of misfolded and aggregated proteins. For the purpose of this review, we will discuss the role of the UPS system in the context of age-related loss of muscle mass and function. We highlight the significant role that E3 ubiquitin ligases play in the turnover of key components (e.g., mitochondria and neuromuscular junction) essential to skeletal muscle function and the influence of aging. In addition, we will briefly discuss the contribution of the UPS system to lifespan. By understanding the UPS system as part of the proteostasis network in age-related diseases and disorders such as sarcopenia, new discoveries can be made and new interventions can be developed which will preserve muscle function and maintain quality of life with advancing age.
Collapse
Affiliation(s)
- David C. Hughes
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (L.M.B.); (S.C.B.)
| | - Leslie M. Baehr
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (L.M.B.); (S.C.B.)
| | - David S. Waddell
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA;
| | - Adam P. Sharples
- Institute for Physical Performance, Norwegian School of Sport Sciences (NiH), 0863 Oslo, Norway;
| | - Sue C. Bodine
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (L.M.B.); (S.C.B.)
| |
Collapse
|
118
|
Könemann S, von Wyl M, Vom Berg C. Zebrafish Larvae Rapidly Recover from Locomotor Effects and Neuromuscular Alterations Induced by Cholinergic Insecticides. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:8449-8462. [PMID: 35575681 DOI: 10.1021/acs.est.2c00161] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Owing to the importance of acetylcholine as a neurotransmitter, many insecticides target the cholinergic system. Across phyla, cholinergic signaling is essential for many neuro-developmental processes including axonal pathfinding and synaptogenesis. Consequently, early-life exposure to such insecticides can disturb these processes, resulting in an impaired nervous system. One test frequently used to assess developmental neurotoxicity is the zebrafish light-dark transition test, which measures larval locomotion as a response to light changes. However, it is only poorly understood which structural alterations cause insecticide-induced locomotion defects and how persistent these alterations are. Therefore, this study aimed to link locomotion defects with effects on neuromuscular structures, including motorneurons, synapses, and muscles, and to investigate the longevity of the effects. The cholinergic insecticides diazinon and dimethoate (organophosphates), methomyl and pirimicarb (carbamates), and imidacloprid and thiacloprid (neonicotinoids) were used to induce hypoactivity. Our analyses revealed that some insecticides did not alter any of the structures assessed, while others affected axon branching (methomyl, imidacloprid) or muscle integrity (methomyl, thiacloprid). The majority of effects, even structural, were reversible within 24 to 72 h. Overall, we find that both neurodevelopmental and non-neurodevelopmental effects of different longevity can account for the reduced locomotion. These findings provide unprecedented insights into the underpinnings of insecticide-induced hypoactivity.
Collapse
Affiliation(s)
- Sarah Könemann
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
- École Polytechnique Fédéral de Lausanne, EPFL, Route Cantonale, 1015 Lausanne, Switzerland
| | - Melissa von Wyl
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
- University of Zurich, UZH, Rämistrassse 71, 8006 Zurich, Switzerland
| | - Colette Vom Berg
- Department of Environmental Toxicology, Eawag, Überlandstrasse 133, 8600 Dübendorf, Switzerland
| |
Collapse
|
119
|
Boëx M, Cottin S, Halliez M, Bauché S, Buon C, Sans N, Montcouquiol M, Molgó J, Amar M, Ferry A, Lemaitre M, Rouche A, Langui D, Baskaran A, Fontaine B, Messéant J, Strochlic L. The cell polarity protein Vangl2 in the muscle shapes the neuromuscular synapse by binding to and regulating the tyrosine kinase MuSK. Sci Signal 2022; 15:eabg4982. [PMID: 35580169 DOI: 10.1126/scisignal.abg4982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The development of the neuromuscular junction (NMJ) requires dynamic trans-synaptic coordination orchestrated by secreted factors, including Wnt family morphogens. To investigate how these synaptic cues in NMJ development are transduced, particularly in the regulation of acetylcholine receptor (AChR) accumulation in the postsynaptic membrane, we explored the function of Van Gogh-like protein 2 (Vangl2), a core component of Wnt planar cell polarity signaling. We found that conditional, muscle-specific ablation of Vangl2 in mice reproduced the NMJ differentiation defects seen in mice with global Vangl2 deletion. These alterations persisted into adulthood and led to NMJ disassembly, impaired neurotransmission, and deficits in motor function. Vangl2 and the muscle-specific receptor tyrosine kinase MuSK were functionally associated in Wnt signaling in the muscle. Vangl2 bound to and promoted the signaling activity of MuSK in response to Wnt11. The loss of Vangl2 impaired RhoA activation in cultured mouse myotubes and caused dispersed, rather than clustered, organization of AChRs at the postsynaptic or muscle cell side of NMJs in vivo. Our results identify Vangl2 as a key player of the core complex of molecules shaping neuromuscular synapses and thus shed light on the molecular mechanisms underlying NMJ assembly.
Collapse
Affiliation(s)
- Myriam Boëx
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Steve Cottin
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Marius Halliez
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Stéphanie Bauché
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Céline Buon
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Nathalie Sans
- Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, UMR-S 1215, Bordeaux 33077, France.,Université Bordeaux, Neurocentre Magendie, Bordeaux, 33000, France
| | - Mireille Montcouquiol
- Institut National de la Santé et de la Recherche Médicale, Neurocentre Magendie, UMR-S 1215, Bordeaux 33077, France.,Université Bordeaux, Neurocentre Magendie, Bordeaux, 33000, France
| | - Jordi Molgó
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux énergies Alternatives, Institut des Sciences du Vivant Frédéric Joliot, Département Médicaments et Technologies pour la Santé, Equipe Mixte de Recherche CNRS 9004, Service d'Ingénierie Moléculaire pour la Santé, Gif-sur-Yvette 91191, France
| | - Muriel Amar
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux énergies Alternatives, Institut des Sciences du Vivant Frédéric Joliot, Département Médicaments et Technologies pour la Santé, Equipe Mixte de Recherche CNRS 9004, Service d'Ingénierie Moléculaire pour la Santé, Gif-sur-Yvette 91191, France
| | - Arnaud Ferry
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Mégane Lemaitre
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Phénotypage du Petit Animal, Paris 75013, France
| | - Andrée Rouche
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Dominique Langui
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut du Cerveau et de la Moelle, Plate-forme d'Imagerie Cellulaire Pitié-Salpêtrière, Paris 75013, France
| | - Asha Baskaran
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut du Cerveau et de la Moelle, Plate-forme d'Imagerie Cellulaire Pitié-Salpêtrière, Paris 75013, France
| | - Bertrand Fontaine
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France.,Assistance Publique-Hôpitaux de Paris (AP-HP) Service de Neuro-Myologie, Hôpital Universitaire Pitié-Salpêtrière, Paris 75013, France
| | - Julien Messéant
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Laure Strochlic
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| |
Collapse
|
120
|
Raffa P, Easler M, Urciuolo A. Three-dimensional in vitro models of neuromuscular tissue. Neural Regen Res 2022; 17:759-766. [PMID: 34472462 PMCID: PMC8530117 DOI: 10.4103/1673-5374.322447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/08/2021] [Accepted: 05/18/2021] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle is a dynamic tissue in which homeostasis and function are guaranteed by a very defined three-dimensional organization of myofibers in respect to other non-muscular components, including the extracellular matrix and the nervous network. In particular, communication between myofibers and the nervous system is essential for the overall correct development and function of the skeletal muscle. A wide range of chronic, acute and genetic-based human pathologies that lead to the alteration of muscle function are associated with modified preservation of the fine interaction between motor neurons and myofibers at the neuromuscular junction. Recent advancements in the development of in vitro models for human skeletal muscle have shown that three-dimensionality and integration of multiple cell types are both key parameters required to unveil pathophysiological relevant phenotypes. Here, we describe recent achievement reached in skeletal muscle modeling which used biomaterials for the generation of three-dimensional constructs of myotubes integrated with motor neurons.
Collapse
Affiliation(s)
- Paolo Raffa
- Institute of Pediatric Research IRP, Padova, Italy
| | - Maria Easler
- Institute of Pediatric Research IRP, Padova, Italy
| | - Anna Urciuolo
- Institute of Pediatric Research IRP, Padova, Italy
- Molecular Medicine Department, University of Padova, Padova, Italy
| |
Collapse
|
121
|
Jones EJ, Chiou S, Atherton PJ, Phillips BE, Piasecki M. Ageing and exercise-induced motor unit remodelling. J Physiol 2022; 600:1839-1849. [PMID: 35278221 PMCID: PMC9314090 DOI: 10.1113/jp281726] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/14/2022] [Indexed: 11/08/2022] Open
Abstract
A motor unit (MU) comprises the neuron cell body, its corresponding axon and each of the muscle fibres it innervates. Many studies highlight age-related reductions in the number of MUs, yet the ability of a MU to undergo remodelling and to expand to rescue denervated muscle fibres is also a defining feature of MU plasticity. Remodelling of MUs involves two coordinated processes: (i) axonal sprouting and new branching growth from adjacent surviving neurons, and (ii) the formation of key structures around the neuromuscular junction to resume muscle-nerve communication. These processes rely on neurotrophins and coordinated signalling in muscle-nerve interactions. To date, several neurotrophins have attracted focus in animal models, including brain-derived neurotrophic factor and insulin-like growth factors I and II. Exercise in older age has demonstrated benefits in multiple physiological systems including skeletal muscle, yet evidence suggests this may also extend to peripheral MU remodelling. There is, however, a lack of research in humans due to methodological limitations which are easily surmountable in animal models. To improve mechanistic insight of the effects of exercise on MU remodelling with advancing age, future research should focus on combining methodological approaches to explore the in vivo physiological function of the MU alongside alterations of the localised molecular environment.
Collapse
Affiliation(s)
- Eleanor J. Jones
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC–Versus Arthritis Centre of Excellence for Musculoskeletal Ageing ResearchNottingham NIHR Biomedical Research CentreSchool of MedicineUniversity of NottinghamNottinghamUK
| | - Shin‐Yi Chiou
- School of SportExercise, and Rehabilitation Sciences, MRC‐Versus Arthritis Centre for Musculoskeletal Ageing Research, Centre for Human Brain HealthUniversity of BirminghamBirminghamUK
| | - Philip J. Atherton
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC–Versus Arthritis Centre of Excellence for Musculoskeletal Ageing ResearchNottingham NIHR Biomedical Research CentreSchool of MedicineUniversity of NottinghamNottinghamUK
| | - Bethan E. Phillips
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC–Versus Arthritis Centre of Excellence for Musculoskeletal Ageing ResearchNottingham NIHR Biomedical Research CentreSchool of MedicineUniversity of NottinghamNottinghamUK
| | - Mathew Piasecki
- Centre of Metabolism, Ageing & Physiology (COMAP), MRC–Versus Arthritis Centre of Excellence for Musculoskeletal Ageing ResearchNottingham NIHR Biomedical Research CentreSchool of MedicineUniversity of NottinghamNottinghamUK
| |
Collapse
|
122
|
Deschenes MR, Flannery R, Hawbaker A, Patek L, Mifsud M. Adaptive Remodeling of the Neuromuscular Junction with Aging. Cells 2022; 11:cells11071150. [PMID: 35406714 PMCID: PMC8997609 DOI: 10.3390/cells11071150] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 02/06/2023] Open
Abstract
Aging is associated with gradual degeneration, in mass and function, of the neuromuscular system. This process, referred to as “sarcopenia”, is considered a disease by itself, and it has been linked to a number of other serious maladies such as type II diabetes, osteoporosis, arthritis, cardiovascular disease, and even dementia. While the molecular causes of sarcopenia remain to be fully elucidated, recent findings have implicated the neuromuscular junction (NMJ) as being an important locus in the development and progression of that malady. This synapse, which connects motor neurons to the muscle fibers that they innervate, has been found to degenerate with age, contributing both to senescent-related declines in muscle mass and function. The NMJ also shows plasticity in response to a number of neuromuscular diseases such as amyotrophic lateral sclerosis (ALS) and Lambert-Eaton myasthenic syndrome (LEMS). Here, the structural and functional degradation of the NMJ associated with aging and disease is described, along with the measures that might be taken to effectively mitigate, if not fully prevent, that degeneration.
Collapse
|
123
|
Lavin KM, Coen PM, Baptista LC, Bell MB, Drummer D, Harper SA, Lixandrão ME, McAdam JS, O’Bryan SM, Ramos S, Roberts LM, Vega RB, Goodpaster BH, Bamman MM, Buford TW. State of Knowledge on Molecular Adaptations to Exercise in Humans: Historical Perspectives and Future Directions. Compr Physiol 2022; 12:3193-3279. [PMID: 35578962 PMCID: PMC9186317 DOI: 10.1002/cphy.c200033] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
For centuries, regular exercise has been acknowledged as a potent stimulus to promote, maintain, and restore healthy functioning of nearly every physiological system of the human body. With advancing understanding of the complexity of human physiology, continually evolving methodological possibilities, and an increasingly dire public health situation, the study of exercise as a preventative or therapeutic treatment has never been more interdisciplinary, or more impactful. During the early stages of the NIH Common Fund Molecular Transducers of Physical Activity Consortium (MoTrPAC) Initiative, the field is well-positioned to build substantially upon the existing understanding of the mechanisms underlying benefits associated with exercise. Thus, we present a comprehensive body of the knowledge detailing the current literature basis surrounding the molecular adaptations to exercise in humans to provide a view of the state of the field at this critical juncture, as well as a resource for scientists bringing external expertise to the field of exercise physiology. In reviewing current literature related to molecular and cellular processes underlying exercise-induced benefits and adaptations, we also draw attention to existing knowledge gaps warranting continued research effort. © 2021 American Physiological Society. Compr Physiol 12:3193-3279, 2022.
Collapse
Affiliation(s)
- Kaleen M. Lavin
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Human Health, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, Florida, USA
| | - Paul M. Coen
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Liliana C. Baptista
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Margaret B. Bell
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Devin Drummer
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sara A. Harper
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manoel E. Lixandrão
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jeremy S. McAdam
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samia M. O’Bryan
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sofhia Ramos
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Lisa M. Roberts
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rick B. Vega
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Bret H. Goodpaster
- Translational Research Institute for Metabolism and Diabetes, Advent Health, Orlando, Florida, USA
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Marcas M. Bamman
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Center for Human Health, Resilience, and Performance, Institute for Human and Machine Cognition, Pensacola, Florida, USA
| | - Thomas W. Buford
- Center for Exercise Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
124
|
Lei L, Fan Z, Su S, Xu M, Chen H, Zhu W, Luan Q, Da Y. Involvement of Ocular Muscles in Patients With Myasthenia Gravis With Nonocular Onset. J Neuroophthalmol 2022; 42:e260-e266. [PMID: 34369469 DOI: 10.1097/wno.0000000000001325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Myasthenia gravis (MG) is an autoimmune disorder involving neuromuscular junctions and more than half of MG patients manifested with extraocular muscle weakness initially. In the remained patients, ocular weakness may occur later in the course of the disease. However, little data are available about ocular involvement in such patients. Therefore, the study aims to investigate ocular weakness in MG patients with nonocular onset and evaluate the associated factors influencing it. METHODS In our monocentric retrospective study, 54 adult-onset patients with MG with nonocular onset were included and were followed up for at least 2 years from the onset. The primary outcome was the occurrence of ptosis, diplopia, or both. Kaplan-Meier analysis was performed to estimate the time to the ocular weakness, and log-rank tests were used to analyze the association between clinical characteristics and ocular weakness. Multivariate Cox proportional hazards regression models were used to identify factors associated with ocular involvement. RESULTS A total of 47 (87.0%) patients developed ocular weakness during the study period. The median time to ocular weakness was 6.0 months. Time to the ocular involvement was earlier in patients with bulbar onset (P = 0.007), whereas patients receiving pyridostigmine monotherapy and immunomodulatory therapy had a longer median time of ocular weakness (P < 0.0001). No significant difference was noted between ocular weakness and age of onset, gender, and thymoma. The Cox analysis showed that bulbar onset was a risk factor of ocular weakness (adjusted hazard ratio [HR] 2.65, 95% confidence interval [CI] 1.41-4.99), whereas pyridostigmine monotherapy (adjusted HR 0.28, 95% CI 0.13-0.60) and immunotherapy (adjusted HR 0.09, 95% CI 0.04-0.22) were protective factors. CONCLUSIONS Eighty-seven percent of patients with MG with nonocular onset developed ocular weakness. Bulbar onset was an independent risk factor for ocular involvement, whereas pyridostigmine and immunotherapy were protective factors.
Collapse
Affiliation(s)
- Lin Lei
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
125
|
Xie T, Qiao X, Sun C, Chu B, Meng J, Chen C. GAPDH S-nitrosation contributes to age-related sarcopenia through mediating apoptosis. Nitric Oxide 2022; 120:1-8. [PMID: 34973445 DOI: 10.1016/j.niox.2021.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/08/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022]
Abstract
The age-related loss of muscle mass and muscle function known as sarcopenia is a major public health problem among older people. Recent research suggests that activation of apoptotic signaling is a critical aspect of the pathogenesis of age-related sarcopenia. However, little information exists in the literature about the apoptotic mechanism of sarcopenia in aging. Herein, we report that elevated glyceraldehyde-3-phosphate dehydrogenase (GAPDH) S-nitrosation and apoptosis occur in sarcopenia during natural aging and that translocation of S-nitrosated GAPDH to the nucleus and S-nitrosated GAPDH-mediated apoptosis contributed to sarcopenia. The levels and sites of GAPDH S-nitrosation in muscle tissues of young, adult and old mice were studied with a quantitative S-nitrosation proteomic analysis approach. GAPDH S-nitrosation increased with aging, and the GAPDH modification sites Cys150, Cys154 and Cys245 were identified. The upregulated S-nitrosation of GAPDH relies on inducible nitric oxide synthase (iNOS) rather than enzymes involved in denitrosylation. Treatment with the iNOS inhibitor 1400W or mutation of GAPDH S-nitrosation sites alleviated apoptosis of C2C12 cells, further demonstrating that GAPDH S-nitrosation in aging contributes to sarcopenia. Taken together, these findings reveal a new cellular mechanism underlying age-related sarcopenia, and the demonstration of muscle loss mediated by iNOS-induced GAPDH S-nitrosation suggests a potential therapeutic strategy for sarcopenia.
Collapse
Affiliation(s)
- Ting Xie
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, 646000, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chuanxin Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Boyu Chu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiao Meng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chang Chen
- School of Basic Medical Sciences of Southwest Medical University, Luzhou, 646000, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
126
|
Modulation of the cholinergic pathway induced by skin secretion of Phyllomedusa iheringii Boulenger, 1885 in a vertebrate model. Toxicon 2022; 210:115-122. [PMID: 35202645 DOI: 10.1016/j.toxicon.2022.02.017] [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: 11/22/2021] [Revised: 02/10/2022] [Accepted: 02/19/2022] [Indexed: 11/24/2022]
Abstract
Amphibians represent one of the main natural sources of bioactive molecules of interest to biotechnological research. The Phyllomedusidae family has several species occurring in Brazil and some studies demonstrate the biological potential of poisons of these species, however many still need to be characterized. Phyllomedusa iheringii is endemic in Brazilian and Uruguayan Pampa Biome and has little data in the literature regarding the action of its poison on experimental organisms. Thus, the present work evaluates the biological activity of P. iheringii secretion on the central and peripheral nervous system of a vertebrate model. The skin secretions of P. iheringii (SSPI) were collected through manual compression and electrical stimulation of the animal's bodies. The resulting content was used in neurobiological tests searching for modulatory effects on the main pathways involved in the neurotoxicity mechanism of vertebrates. SSPI affected the contraction force of the chick biventer cervicis muscle (Gallus gallus domesticus) at some concentrations used (5, 10, and 12 μg/mL). In slices from the cerebral cortex of G. gallus domesticus an increase in cell viability was observed after treatment with SSPI (10 μg/mL) and a neuroprotective effect when treated simultaneously with hydrogen peroxide (H2O2), Neostigmine (NEO) and Trichlorfon (TRI). The cholinergic pathway is possibly the main pathway modulated by SSPI since assays with the cerebral cortex and biventer cervicis muscle demonstrated the increased activity of the enzyme acetylcholinesterase (AChE) (SSPI 10 μg/mL and 12 μg/mL, respectively). SSPI (10 μg/mL) also prevented the modulation of NEO and TRI, two recognized anticholinesterase agents, in AChE activity in slices of the cerebral cortex. Therefore, our results have demonstrated the unpublished biotechnological potential of P. iheringii over the vertebrate model and its modulation on the nervous system, with apparent action on the cholinergic pathway.
Collapse
|
127
|
Anderson JE. Key concepts in muscle regeneration: muscle "cellular ecology" integrates a gestalt of cellular cross-talk, motility, and activity to remodel structure and restore function. Eur J Appl Physiol 2022; 122:273-300. [PMID: 34928395 PMCID: PMC8685813 DOI: 10.1007/s00421-021-04865-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/10/2021] [Indexed: 12/21/2022]
Abstract
This review identifies some key concepts of muscle regeneration, viewed from perspectives of classical and modern research. Early insights noted the pattern and sequence of regeneration across species was similar, regardless of the type of injury, and differed from epimorphic limb regeneration. While potential benefits of exercise for tissue repair was debated, regeneration was not presumed to deliver functional restoration, especially after ischemia-reperfusion injury; muscle could develop fibrosis and ectopic bone and fat. Standard protocols and tools were identified as necessary for tracking injury and outcomes. Current concepts vastly extend early insights. Myogenic regeneration occurs within the environment of muscle tissue. Intercellular cross-talk generates an interactive system of cellular networks that with the extracellular matrix and local, regional, and systemic influences, forms the larger gestalt of the satellite cell niche. Regenerative potential and adaptive plasticity are overlain by epigenetically regionalized responsiveness and contributions by myogenic, endothelial, and fibroadipogenic progenitors and inflammatory and metabolic processes. Muscle architecture is a living portrait of functional regulatory hierarchies, while cellular dynamics, physical activity, and muscle-tendon-bone biomechanics arbitrate regeneration. The scope of ongoing research-from molecules and exosomes to morphology and physiology-reveals compelling new concepts in muscle regeneration that will guide future discoveries for use in application to fitness, rehabilitation, and disease prevention and treatment.
Collapse
Affiliation(s)
- Judy E Anderson
- Department of Biological Sciences, Faculty of Science, University of Manitoba, 50 Sifton Road, Winnipeg, MB, R3T 2N2, Canada.
| |
Collapse
|
128
|
Latrotoxin-Induced Neuromuscular Junction Degeneration Reveals Urocortin 2 as a Critical Contributor to Motor Axon Terminal Regeneration. Int J Mol Sci 2022; 23:ijms23031186. [PMID: 35163106 PMCID: PMC8835473 DOI: 10.3390/ijms23031186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/24/2022] Open
Abstract
We used α-Latrotoxin (α-LTx), the main neurotoxic component of the black widow spider venom, which causes degeneration of the neuromuscular junction (NMJ) followed by a rapid and complete regeneration, as a molecular tool to identify by RNA transcriptomics factors contributing to the structural and functional recovery of the NMJ. We found that Urocortin 2 (UCN2), a neuropeptide involved in the stress response, is rapidly expressed at the NMJ after acute damage and that inhibition of CRHR2, the specific receptor of UCN2, delays neuromuscular transmission rescue. Experiments in neuronal cultures show that CRHR2 localises at the axonal tips of growing spinal motor neurons and that its expression inversely correlates with synaptic maturation. Moreover, exogenous UCN2 enhances the growth of axonal sprouts in cultured neurons in a CRHR2-dependent manner, pointing to a role of the UCN2-CRHR2 axis in the regulation of axonal growth and synaptogenesis. Consistently, exogenous administration of UCN2 strongly accelerates the regrowth of motor axon terminals degenerated by α-LTx, thereby contributing to the functional recovery of neuromuscular transmission after damage. Taken together, our results posit a novel role for UCN2 and CRHR2 as a signalling axis involved in NMJ regeneration.
Collapse
|
129
|
Graham ZA. Mini-review: Local and downstream responses to the neuromuscular junction: Potential roles for integrins, connexins and ephrins in altering muscle characteristics and function. Neurosci Lett 2022; 768:136359. [PMID: 34813913 DOI: 10.1016/j.neulet.2021.136359] [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: 12/30/2020] [Revised: 09/08/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Abstract
Skeletal muscle develops in a manner directly related to its innervating motor neuron. The formation of the neuromuscular junction (NMJ) is a well-described process that is coordinated to allow for efficient communication between the central nervous system and muscle for muscle contraction and movement. Some of the major mediators of NMJ formation, like muscle-specific kinase, agrin and laminin, have been thoroughly described but there are other important proteins that have an integral role in muscle health that have also been associated with proper NMJ integrity and fiber health and function. This mini-review focuses on integrins, connexin hemichannels and ephrins and their relationship with the NMJin regulating muscle health.
Collapse
Affiliation(s)
- Zachary A Graham
- Birmingham VA Medical Center, Birmingham, AL, United States; Department of Cell, Developmental and Integrative Biology, University of Alabama-Birmingham, Birmingham, AL, United States.
| |
Collapse
|
130
|
Mankowski RT, Laitano O, Darden D, Kelly L, Munley J, Loftus TJ, Mohr AM, Efron PA, Thomas RM. Sepsis-Induced Myopathy and Gut Microbiome Dysbiosis: Mechanistic Links and Therapeutic Targets. Shock 2022; 57:15-23. [PMID: 34726875 PMCID: PMC9373856 DOI: 10.1097/shk.0000000000001843] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
ABSTRACT Sepsis is currently defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection. The skeletal muscle system is among the host organ systems compromised by sepsis. The resulting neuromuscular dysfunction and impaired regenerative capacity defines sepsis-induced myopathy and manifests as atrophy, loss of strength, and hindered regeneration after injury. These outcomes delay recovery from critical illness and confer increased vulnerability to morbidity and mortality. The mechanisms underlying sepsis-induced myopathy, including the potential contribution of peripheral organs, remain largely unexplored. The gut microbiome is an immunological and homeostatic entity that interacts with and controls end-organ function, including the skeletal muscle system. Sepsis induces alterations in the gut microbiota composition, which is globally termed a state of "dysbiosis" for the host compared to baseline microbiota composition. In this review, we critically evaluate existing evidence and potential mechanisms linking sepsis-induced myopathy with gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Robert T. Mankowski
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL
| | - Orlando Laitano
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL
| | - Dijoia Darden
- Department of Surgery, University of Florida, Gainesville, FL
| | - Lauren Kelly
- Department of Surgery, University of Florida, Gainesville, FL
| | - Jennifer Munley
- Department of Surgery, University of Florida, Gainesville, FL
| | - Tyler J. Loftus
- Department of Surgery, University of Florida, Gainesville, FL
| | - Alicia M. Mohr
- Department of Surgery, University of Florida, Gainesville, FL
| | - Philip A. Efron
- Department of Surgery, University of Florida, Gainesville, FL
| | - Ryan M. Thomas
- Department of Surgery, University of Florida, Gainesville, FL
- Department of Molecular Genetics and Microbiology; University of Florida College of Medicine; Gainesville, FL
- Section of General Surgery, North Florida/South Georgia Veterans Health System; Gainesville, FL
| |
Collapse
|
131
|
Massopust R, Juros D, Shapiro D, Lopes M, Haldar SM, Taetzsch T, Valdez G. KLF15 overexpression in myocytes fails to ameliorate ALS-related pathology or extend the lifespan of SOD1G93A mice. Neurobiol Dis 2022; 162:105583. [PMID: 34902552 PMCID: PMC8750438 DOI: 10.1016/j.nbd.2021.105583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/19/2021] [Accepted: 12/10/2021] [Indexed: 01/22/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a currently incurable disease that causes progressive motor neuron loss, paralysis and death. Skeletal muscle pathology occurs early during the course of ALS. It is characterized by impaired mitochondrial biogenesis, metabolic dysfunction and deterioration of the neuromuscular junction (NMJ), the synapse through which motor neurons communicate with muscles. Therefore, a better understanding of the molecules that underlie this pathology may lead to therapies that slow motor neuron loss and delay ALS progression. Kruppel Like Factor 15 (KLF15) has been identified as a transcription factor that activates alternative metabolic pathways and NMJ maintenance factors, including Fibroblast Growth Factor Binding Protein 1 (FGFBP1), in skeletal myocytes. In this capacity, KLF15 has been shown to play a protective role in Duchenne muscular dystrophy (DMD) and spinal muscular atrophy (SMA), however its role in ALS has not been evaluated. Here, we examined whether muscle-specific KLF15 overexpression promotes the health of skeletal muscles and NMJs in the SOD1G93A ALS mouse model. We show that muscle-specific KLF15 overexpression did not elicit a significant beneficial effect on skeletal muscle atrophy, NMJ health, motor function, or survival in SOD1G93A ALS mice. Our findings suggest that, unlike in mouse models of DMD and SMA, KLF15 overexpression has a minimal impact on ALS disease progression in SOD1G93A mice.
Collapse
Affiliation(s)
- Ryan Massopust
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Devin Juros
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Dillon Shapiro
- Molecular Biology, Cell Biology, & Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Mikayla Lopes
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Saptarsi M. Haldar
- Gladstone Institutes, San Francisco, California, USA,Department of Medicine, Cardiology Division, UCSF School of Medicine, San Francisco, California, USA,Current address: Amgen Research, South San Francisco, California, USA
| | - Thomas Taetzsch
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA,Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, USA,Department of Neurology, Warren Alpert Medical School of Brown University, Providence, United States
| |
Collapse
|
132
|
Chien P, Xi H, Pyle AD. Recapitulating human myogenesis ex vivo using human pluripotent stem cells. Exp Cell Res 2021; 411:112990. [PMID: 34973262 DOI: 10.1016/j.yexcr.2021.112990] [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: 05/03/2021] [Revised: 12/15/2021] [Accepted: 12/22/2021] [Indexed: 11/25/2022]
Abstract
Human pluripotent stem cells (hPSCs) provide a human model for developmental myogenesis, disease modeling and development of therapeutics. Differentiation of hPSCs into muscle stem cells has the potential to provide a cell-based therapy for many skeletal muscle wasting diseases. This review describes the current state of hPSCs towards recapitulating human myogenesis ex vivo, considerations of stem cell and progenitor cell state as well as function for future use of hPSC-derived muscle cells in regenerative medicine.
Collapse
Affiliation(s)
- Peggie Chien
- Department of Microbiology, Immunology and Molecular Genetics, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - Haibin Xi
- Department of Microbiology, Immunology and Molecular Genetics, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA
| | - April D Pyle
- Department of Microbiology, Immunology and Molecular Genetics, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
133
|
Pinto C, Pérez V, Mella J, Albistur M, Caprile T, Bronfman FC, Henríquez JP. Transport and Secretion of the Wnt3 Ligand by Motor Neuron-like Cells and Developing Motor Neurons. Biomolecules 2021; 11:biom11121898. [PMID: 34944540 PMCID: PMC8699186 DOI: 10.3390/biom11121898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
The vertebrate neuromuscular junction (NMJ) is formed by a presynaptic motor nerve terminal and a postsynaptic muscle specialization. Cumulative evidence reveals that Wnt ligands secreted by the nerve terminal control crucial steps of NMJ synaptogenesis. For instance, the Wnt3 ligand is expressed by motor neurons at the time of NMJ formation and induces postsynaptic differentiation in recently formed muscle fibers. However, the behavior of presynaptic-derived Wnt ligands at the vertebrate NMJ has not been deeply analyzed. Here, we conducted overexpression experiments to study the expression, distribution, secretion, and function of Wnt3 by transfection of the motor neuron-like NSC-34 cell line and by in ovo electroporation of chick motor neurons. Our findings reveal that Wnt3 is transported along motor axons in vivo following a vesicular-like pattern and reaches the NMJ area. In vitro, we found that endogenous Wnt3 expression increases as the differentiation of NSC-34 cells proceeds. Although NSC-34 cells overexpressing Wnt3 do not modify their morphological differentiation towards a neuronal phenotype, they effectively induce acetylcholine receptor clustering on co-cultured myotubes. These findings support the notion that presynaptic Wnt3 is transported and secreted by motor neurons to induce postsynaptic differentiation in nascent NMJs.
Collapse
Affiliation(s)
- Cristina Pinto
- Neuromuscular Studies Laboratory (NeSt Lab), CMA Bio-Bio, Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Universidad de Concepción, Concepción 4070112, Chile; (C.P.); (V.P.); (J.M.); (M.A.)
| | - Viviana Pérez
- Neuromuscular Studies Laboratory (NeSt Lab), CMA Bio-Bio, Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Universidad de Concepción, Concepción 4070112, Chile; (C.P.); (V.P.); (J.M.); (M.A.)
| | - Jessica Mella
- Neuromuscular Studies Laboratory (NeSt Lab), CMA Bio-Bio, Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Universidad de Concepción, Concepción 4070112, Chile; (C.P.); (V.P.); (J.M.); (M.A.)
| | - Miguel Albistur
- Neuromuscular Studies Laboratory (NeSt Lab), CMA Bio-Bio, Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Universidad de Concepción, Concepción 4070112, Chile; (C.P.); (V.P.); (J.M.); (M.A.)
| | - Teresa Caprile
- Axon Guidance Laboratory, Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Universidad de Concepción, Concepción 4070112, Chile;
| | - Francisca C. Bronfman
- Institute of Biomedical Sciences (ICB), Faculty of Medicine and Faculty of Life Science, Universidad Andres Bello, Santiago 8320000, Chile;
- CARE Biomedical Research Center, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
| | - Juan Pablo Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), CMA Bio-Bio, Group for the Study of Developmental Processes (GDeP), Department of Cell Biology, Universidad de Concepción, Concepción 4070112, Chile; (C.P.); (V.P.); (J.M.); (M.A.)
- Correspondence: ; Tel.: +56-41-220-3492
| |
Collapse
|
134
|
Fralish Z, Lotz EM, Chavez T, Khodabukus A, Bursac N. Neuromuscular Development and Disease: Learning From in vitro and in vivo Models. Front Cell Dev Biol 2021; 9:764732. [PMID: 34778273 PMCID: PMC8579029 DOI: 10.3389/fcell.2021.764732] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized cholinergic synaptic interface between a motor neuron and a skeletal muscle fiber that translates presynaptic electrical impulses into motor function. NMJ formation and maintenance require tightly regulated signaling and cellular communication among motor neurons, myogenic cells, and Schwann cells. Neuromuscular diseases (NMDs) can result in loss of NMJ function and motor input leading to paralysis or even death. Although small animal models have been instrumental in advancing our understanding of the NMJ structure and function, the complexities of studying this multi-tissue system in vivo and poor clinical outcomes of candidate therapies developed in small animal models has driven the need for in vitro models of functional human NMJ to complement animal studies. In this review, we discuss prevailing models of NMDs and highlight the current progress and ongoing challenges in developing human iPSC-derived (hiPSC) 3D cell culture models of functional NMJs. We first review in vivo development of motor neurons, skeletal muscle, Schwann cells, and the NMJ alongside current methods for directing the differentiation of relevant cell types from hiPSCs. We further compare the efficacy of modeling NMDs in animals and human cell culture systems in the context of five NMDs: amyotrophic lateral sclerosis, myasthenia gravis, Duchenne muscular dystrophy, myotonic dystrophy, and Pompe disease. Finally, we discuss further work necessary for hiPSC-derived NMJ models to function as effective personalized NMD platforms.
Collapse
Affiliation(s)
| | | | | | | | - Nenad Bursac
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
135
|
Verdile V, Guizzo G, Ferrante G, Paronetto MP. RNA Targeting in Inherited Neuromuscular Disorders: Novel Therapeutic Strategies to Counteract Mis-Splicing. Cells 2021; 10:2850. [PMID: 34831073 PMCID: PMC8616048 DOI: 10.3390/cells10112850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/08/2021] [Accepted: 10/18/2021] [Indexed: 01/14/2023] Open
Abstract
Neuromuscular disorders represent multifaceted abnormal conditions, with little or no cure, leading to patient deaths from complete muscle wasting and atrophy. Despite strong efforts in the past decades, development of effective treatments is still urgently needed. Advent of next-generation sequencing technologies has allowed identification of novel genes and mutations associated with neuromuscular pathologies, highlighting splicing defects as essential players. Deciphering the significance and relative contributions of defective RNA metabolism will be instrumental to address and counteract these malignancies. We review here recent progress on the role played by alternative splicing in ensuring functional neuromuscular junctions (NMJs), and its involvement in the pathogenesis of NMJ-related neuromuscular disorders, with particular emphasis on congenital myasthenic syndromes and muscular dystrophies. We will also discuss novel strategies based on oligonucleotides designed to bind their cognate sequences in the RNA or targeting intermediary of mRNA metabolism. These efforts resulted in several chemical classes of RNA molecules that have recently proven to be clinically effective, more potent and better tolerated than previous strategies.
Collapse
Affiliation(s)
- Veronica Verdile
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143 Rome, Italy; (V.V.); (G.G.); (G.F.)
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 6, 00135 Rome, Italy
| | - Gloria Guizzo
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143 Rome, Italy; (V.V.); (G.G.); (G.F.)
| | - Gabriele Ferrante
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143 Rome, Italy; (V.V.); (G.G.); (G.F.)
| | - Maria Paola Paronetto
- Laboratory of Molecular and Cellular Neurobiology, Fondazione Santa Lucia, CERC, 00143 Rome, Italy; (V.V.); (G.G.); (G.F.)
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 6, 00135 Rome, Italy
| |
Collapse
|
136
|
Wu M, Liu CZ, Barrall EA, Rissman RA, Joiner WJ. Unbalanced Regulation of α7 nAChRs by Ly6h and NACHO Contributes to Neurotoxicity in Alzheimer's Disease. J Neurosci 2021; 41:8461-8474. [PMID: 34446574 PMCID: PMC8513707 DOI: 10.1523/jneurosci.0494-21.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/24/2022] Open
Abstract
α7 nicotinic acetylcholine receptors (nAChRs) are widely expressed in the brain where they promote fast cholinergic synaptic transmission and serve important neuromodulatory functions. However, their high permeability to Ca2+ also predisposes them to contribute to disease states. Here, using transfected HEK-tsa cells and primary cultured hippocampal neurons from male and female rats, we demonstrate that two proteins called Ly6h and NACHO compete for access to α7 subunits, operating together but in opposition to maintain α7 assembly and activity within a narrow range that is optimal for neuronal function and viability. Using mixed gender human temporal cortex and cultured hippocampal neurons from rats we further show that this balance is perturbed during Alzheimer's disease (AD) because of amyloid β (Aβ)-driven reduction in Ly6h, with severe reduction leading to increased phosphorylated tau and α7-mediated neurotoxicity. Ly6h release into human CSF is also correlated with AD severity. Thus, Ly6h links cholinergic signaling, Aβ and phosphorylated tau and may serve as a novel marker for AD progression.SIGNIFICANCE STATEMENT One of the earliest and most persistent hypotheses regarding Alzheimer's disease (AD) attributes cognitive impairment to loss of cholinergic signaling. More recently, interest has focused on crucial roles for amyloid β (Aβ) and phosphorylated tau in Alzheimer's pathogenesis. Here, we demonstrate that these elements are linked by Ly6h and its counterpart, NACHO, functioning in opposition to maintain assembly of nicotinic acetylcholine receptors (nAChRs) within the physiological range. Our data suggests that Aβ shifts the balance away from Ly6h and toward NACHO, resulting in increased assembly of Ca2+-permeable nAChRs and thus a conversion of basal cholinergic to neurotoxic signaling.
Collapse
Affiliation(s)
- Meilin Wu
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| | - Clifford Z Liu
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| | - Erika A Barrall
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| | - Robert A Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, California 92093
- Alzheimer's Disease Research Center, University of California San Diego, La Jolla, California 92093
| | - William J Joiner
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
- Center for Circadian Biology, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
137
|
Gromova A, La Spada AR. Harmony Lost: Cell-Cell Communication at the Neuromuscular Junction in Motor Neuron Disease. Trends Neurosci 2021; 43:709-724. [PMID: 32846148 DOI: 10.1016/j.tins.2020.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/05/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022]
Abstract
The neuromuscular junction (NMJ) is a specialized synapse that is the point of connection between motor neurons and skeletal muscle. Although developmental studies have established the importance of cell-cell communication at the NMJ for the integrity and full functionality of this synapse, the contribution of this structure as a primary driver in motor neuron disease pathogenesis remains uncertain. Here, we consider the biology of the NMJ and review emerging lines of investigation that are highlighting the importance of cell-cell interaction at the NMJ in spinal muscular atrophy (SMA), X-linked spinal and bulbar muscular atrophy (SBMA), and amyotrophic lateral sclerosis (ALS). Ongoing research may reveal NMJ targets and pathways whose therapeutic modulation will help slow the progression of motor neuron disease, offering a novel treatment paradigm for ALS, SBMA, SMA, and related disorders.
Collapse
Affiliation(s)
- Anastasia Gromova
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA; Department of Pathology and Laboratory Medicine and Department of Neurology, University of California Irvine, Irvine, CA 92697, USA
| | - Albert R La Spada
- Department of Pathology and Laboratory Medicine and Department of Neurology, University of California Irvine, Irvine, CA 92697, USA; Department of Neurology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
138
|
Pimentel Neto J, Rocha LC, Dos Santos Jacob C, Klein Barbosa G, Ciena AP. Postsynaptic cleft density changes with combined exercise protocols in an experimental model of muscular hypertrophy. Eur J Histochem 2021; 65. [PMID: 34346666 PMCID: PMC8404527 DOI: 10.4081/ejh.2021.3274] [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: 05/15/2021] [Accepted: 07/14/2021] [Indexed: 11/22/2022] Open
Abstract
The vertical ladder-based protocols contribute to the NMJ junction's adaptations, and when combined with and without load, can be potentiated. The present study aimed to investigate postsynaptic regions of the biceps brachii muscle in adult male Wistar rats submitted to different vertical ladder-based protocols (Sedentary - S; Climbing - C; Climbing with Load - LC and Combined Climbing - CC). The protocols (C, LC, CC) were performed in 24 sessions, 3 x/week, for 8 weeks. The myofibrillar ATPase analysis showed an increase in cross-sectional area (CSA) of the muscle fibers Type I in all trained Groups; Type II in C and LC and reduction in CC; Type IIx higher in all trained Groups. In the postsynaptic cleft, the stained area presents smaller in Groups C, LC, and CC; the total area showed smaller than LC and higher in C and CC. The stained and total perimeter, and dispersion showed a reduction in C, LC, and CC, higher maximum diameter in Groups C and CC, and decreased in LC. Regarding the postsynaptic cleft distribution, the stained area presented a decrease in all trained Groups. The integrated density presented higher principally in CC. The NMJ count showed an increase in all trained Groups. We concluded that the vertical ladder-based protocols combined contributed to the postsynaptic region adaptations.
Collapse
Affiliation(s)
- Jurandyr Pimentel Neto
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro-SP.
| | - Lara Caetano Rocha
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro-SP.
| | - Carolina Dos Santos Jacob
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro-SP.
| | - Gabriela Klein Barbosa
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro-SP.
| | - Adriano Polican Ciena
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences (IB), São Paulo State University (UNESP), Rio Claro-SP.
| |
Collapse
|
139
|
Presynaptic Paraneoplastic Disorders of the Neuromuscular Junction: An Update. Brain Sci 2021; 11:brainsci11081035. [PMID: 34439654 PMCID: PMC8392118 DOI: 10.3390/brainsci11081035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 01/17/2023] Open
Abstract
The neuromuscular junction (NMJ) is the target of a variety of immune-mediated disorders, usually classified as presynaptic and postsynaptic, according to the site of the antigenic target and consequently of the neuromuscular transmission alteration. Although less common than the classical autoimmune postsynaptic myasthenia gravis, presynaptic disorders are important to recognize due to the frequent association with cancer. Lambert Eaton myasthenic syndrome is due to a presynaptic failure to release acetylcholine, caused by antibodies to the presynaptic voltage-gated calcium channels. Acquired neuromyotonia is a condition characterized by nerve hyperexcitability often due to the presence of antibodies against proteins associated with voltage-gated potassium channels. This review will focus on the recent developments in the autoimmune presynaptic disorders of the NMJ.
Collapse
|
140
|
Gras S, Blasco A, Mòdol-Caballero G, Tarabal O, Casanovas A, Piedrafita L, Barranco A, Das T, Rueda R, Pereira SL, Navarro X, Esquerda JE, Calderó J. Beneficial effects of dietary supplementation with green tea catechins and cocoa flavanols on aging-related regressive changes in the mouse neuromuscular system. Aging (Albany NY) 2021; 13:18051-18093. [PMID: 34319911 PMCID: PMC8351677 DOI: 10.18632/aging.203336] [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: 04/29/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022]
Abstract
Besides skeletal muscle wasting, sarcopenia entails morphological and molecular changes in distinct components of the neuromuscular system, including spinal cord motoneurons (MNs) and neuromuscular junctions (NMJs); moreover, noticeable microgliosis has also been observed around aged MNs. Here we examined the impact of two flavonoid-enriched diets containing either green tea extract (GTE) catechins or cocoa flavanols on age-associated regressive changes in the neuromuscular system of C57BL/6J mice. Compared to control mice, GTE- and cocoa-supplementation significantly improved the survival rate of mice, reduced the proportion of fibers with lipofuscin aggregates and central nuclei, and increased the density of satellite cells in skeletal muscles. Additionally, both supplements significantly augmented the number of innervated NMJs and their degree of maturity compared to controls. GTE, but not cocoa, prominently increased the density of VAChT and VGluT2 afferent synapses on MNs, which were lost in control aged spinal cords; conversely, cocoa, but not GTE, significantly augmented the proportion of VGluT1 afferent synapses on aged MNs. Moreover, GTE, but not cocoa, reduced aging-associated microgliosis and increased the proportion of neuroprotective microglial phenotypes. Our data indicate that certain plant flavonoids may be beneficial in the nutritional management of age-related deterioration of the neuromuscular system.
Collapse
Affiliation(s)
- Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Guillem Mòdol-Caballero
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Alejandro Barranco
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
| | - Tapas Das
- Abbott Nutrition, Research and Development, Columbus, OH 43215, USA
| | - Ricardo Rueda
- Abbott Nutrition, Research and Development, Granada, Spain
| | | | - Xavier Navarro
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Spain
| | - Josep E. Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| |
Collapse
|
141
|
Zhang K, Bai L, Xu W, Shen C. Human neuromuscular junction three-dimensional organoid models and the insight in motor disorders. J Mol Cell Biol 2021; 13:767-773. [PMID: 34270721 PMCID: PMC8782584 DOI: 10.1093/jmcb/mjab046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 12/02/2022] Open
Abstract
The neuromuscular junction (NMJ), a peripheral synaptic connection between motoneurons and skeletal muscle fibers, controls movement. Dysregulation of NMJs has been implicated in various motor disorders. Because of their large size and easy accessibility, NMJs have been extensively investigated in the neuroscience field and have greatly contributed to our understanding of the fundamental principles of synapses in the central nervous system. Researchers have tried multiple ways to develop models to recreate NMJs. Rapid progress in the research and development of tissue-like organoids has made it possible to produce human NMJ three-dimensional (3D) models in vitro, providing an additional powerful strategy to study NMJs. Here, we introduce the most recent advances of human embryonic stem cell- or induced pluripotent stem cell-derived organoids to model 3D NMJs.
Collapse
Affiliation(s)
- Kejing Zhang
- Department of Neurobiology, The First Affiliated Hospital, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Lei Bai
- Department of Neurobiology, The First Affiliated Hospital, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Wentao Xu
- Department of Neurobiology, The First Affiliated Hospital, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Chengyong Shen
- Department of Neurobiology, The First Affiliated Hospital, Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou 310020, China
| |
Collapse
|
142
|
Marcolin G, Franchi MV, Monti E, Pizzichemi M, Sarto F, Sirago G, Paoli A, Maggio M, Zampieri S, Narici M. Active older dancers have lower C-terminal Agrin fragment concentration, better balance and gait performance than sedentary peers. Exp Gerontol 2021; 153:111469. [PMID: 34246731 DOI: 10.1016/j.exger.2021.111469] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022]
Abstract
Motor neuron degeneration, denervation, neuromuscular junction (NMJ) fragmentation and loss of motor units (MUs), play a key-role in the development of sarcopenia. The aim of the present study was to investigate the beneficial effects of regular practice of dancing in physically active elders on concentration of C-terminal Agrin fragment (CAF), a marker of NMJ instability, muscle mass, strength, and physical performance in a group of 16 recreationally active older dancers (AOD; 70.1 ± 3.4 yr) compared to 15 age-matched sedentary peers (OS; 70.9 ± 6.2 yr). Circulating concentration of CAF was measured in serum, while morphology of the vastus lateralis and multifidus muscles was assessed by ultrasound imaging. In addition, the participants underwent two functional performance tests, the Timed Up and Go (TUG) and the 10-meter walk test (10-MWT), a lower and upper limb isometric strength test, a static and a dynamic balance test. Although no statistically significant differences were detected for both muscle morphology and isometric strength, higher CAF concentration (20%, p < 0.01) was found in OS. AOD showed a better performance in TUG (22%, p < 0.001), 10-MWT (17%, p < 0.001) and dynamic balance (25%, p < 0.01) than OS. Notably, CAF concentration correlated with dynamic balance performance (r = 0.3711, p < 0.05). Our results provide evidence that the regular practice of dancing in older age, together with non-structured light aerobic physical activities, is associated to lower CAF concentration and improved walking and balance performance. Our findings also suggest that NMJ instability, as indicated by elevated CAF serum concentration, seems to precede the loss of muscle size and alterations in muscle architecture normally associated with sarcopenia.
Collapse
Affiliation(s)
- Giuseppe Marcolin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Martino V Franchi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Elena Monti
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Fabio Sarto
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giuseppe Sirago
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Antonio Paoli
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marcello Maggio
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Sandra Zampieri
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, Italy
| | - Marco Narici
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Myology Center (CIR-Myo), Department of Biomedical Sciences, University of Padova, Italy.
| |
Collapse
|
143
|
Gustafsson T, Ulfhake B. Sarcopenia: What Is the Origin of This Aging-Induced Disorder? Front Genet 2021; 12:688526. [PMID: 34276788 PMCID: PMC8285098 DOI: 10.3389/fgene.2021.688526] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/10/2021] [Indexed: 01/03/2023] Open
Abstract
We here review the loss of muscle function and mass (sarcopenia) in the framework of human healthspan and lifespan, and mechanisms involved in aging. The rapidly changing composition of the human population will impact the incidence and the prevalence of aging-induced disorders such as sarcopenia and, henceforth, efforts to narrow the gap between healthspan and lifespan should have top priority. There are substantial knowledge gaps in our understanding of aging. Heritability is estimated to account for only 25% of lifespan length. However, as we push the expected lifespan at birth toward those that we consider long-lived, the genetics of aging may become increasingly important. Linkage studies of genetic polymorphisms to both the susceptibility and aggressiveness of sarcopenia are still missing. Such information is needed to shed light on the large variability in clinical outcomes between individuals and why some respond to interventions while others do not. We here make a case for the concept that sarcopenia has a neurogenic origin and that in manifest sarcopenia, nerve and myofibers enter into a vicious cycle that will escalate the disease progression. We point to gaps in knowledge, for example the crosstalk between the motor axon, terminal Schwann cell, and myofiber in the denervation processes that leads to a loss of motor units and muscle weakness. Further, we argue that the operational definition of sarcopenia should be complemented with dynamic metrics that, along with validated biomarkers, may facilitate early preclinical diagnosis of individuals vulnerable to develop advanced sarcopenia. We argue that preventive measures are likely to be more effective to counter act aging-induced disorders than efforts to treat manifest clinical conditions. To achieve compliance with a prescription of preventive measures that may be life-long, we need to identify reliable predictors to design rational and convincing interventions.
Collapse
Affiliation(s)
- Thomas Gustafsson
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Brun Ulfhake
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
144
|
Kravtsova VV, Krivoi II. Molecular and Functional Heterogeneity of Na,K-ATPase in the Skeletal Muscle. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
145
|
Oury J, Zhang W, Leloup N, Koide A, Corrado AD, Ketavarapu G, Hattori T, Koide S, Burden SJ. Mechanism of disease and therapeutic rescue of Dok7 congenital myasthenia. Nature 2021; 595:404-408. [PMID: 34163073 PMCID: PMC8277574 DOI: 10.1038/s41586-021-03672-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/25/2021] [Indexed: 11/09/2022]
Abstract
Congenital myasthenia (CM) is a devastating neuromuscular disease, and mutations in DOK7, an adaptor protein that is crucial for forming and maintaining neuromuscular synapses, are a major cause of CM1,2. The most common disease-causing mutation (DOK71124_1127 dup) truncates DOK7 and leads to the loss of two tyrosine residues that are phosphorylated and recruit CRK proteins, which are important for anchoring acetylcholine receptors at synapses. Here we describe a mouse model of this common form of CM (Dok7CM mice) and a mouse with point mutations in the two tyrosine residues (Dok72YF). We show that Dok7CM mice had severe deficits in neuromuscular synapse formation that caused neonatal lethality. Unexpectedly, these deficits were due to a severe deficiency in phosphorylation and activation of muscle-specific kinase (MUSK) rather than a deficiency in DOK7 tyrosine phosphorylation. We developed agonist antibodies against MUSK and show that these antibodies restored neuromuscular synapse formation and prevented neonatal lethality and late-onset disease in Dok7CM mice. These findings identify an unexpected cause for disease and a potential therapy for both DOK7 CM and other forms of CM caused by mutations in AGRIN, LRP4 or MUSK, and illustrate the potential of targeted therapy to rescue congenital lethality.
Collapse
Affiliation(s)
- Julien Oury
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Wei Zhang
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Nadia Leloup
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Akiko Koide
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
- Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | | | | | - Takamitsu Hattori
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Shohei Koide
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.
| | - Steven J Burden
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
146
|
Lutz AK, Pfaender S, Incearap B, Ioannidis V, Ottonelli I, Föhr KJ, Cammerer J, Zoller M, Higelin J, Giona F, Stetter M, Stoecker N, Alami NO, Schön M, Orth M, Liebau S, Barbi G, Grabrucker AM, Delorme R, Fauler M, Mayer B, Jesse S, Roselli F, Ludolph AC, Bourgeron T, Verpelli C, Demestre M, Boeckers TM. Autism-associated SHANK3 mutations impair maturation of neuromuscular junctions and striated muscles. Sci Transl Med 2021; 12:12/547/eaaz3267. [PMID: 32522805 DOI: 10.1126/scitranslmed.aaz3267] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/09/2019] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Heterozygous mutations of the gene encoding the postsynaptic protein SHANK3 are associated with syndromic forms of autism spectrum disorders (ASDs). One of the earliest clinical symptoms in SHANK3-associated ASD is neonatal skeletal muscle hypotonia. This symptom can be critical for the early diagnosis of affected children; however, the mechanism mediating hypotonia in ASD is not completely understood. Here, we used a combination of patient-derived human induced pluripotent stem cells (hiPSCs), Shank3Δ11(-/-) mice, and Phelan-McDermid syndrome (PMDS) muscle biopsies from patients of different ages to analyze the role of SHANK3 on motor unit development. Our results suggest that the hypotonia in SHANK3 deficiency might be caused by dysfunctions in all elements of the voluntary motor system: motoneurons, neuromuscular junctions (NMJs), and striated muscles. We found that SHANK3 localizes in Z-discs in the skeletal muscle sarcomere and co-immunoprecipitates with α-ACTININ. SHANK3 deficiency lead to shortened Z-discs and severe impairment of acetylcholine receptor clustering in hiPSC-derived myotubes and in muscle from Shank3Δ11(-/-) mice and patients with PMDS, indicating a crucial role for SHANK3 in the maturation of NMJs and striated muscle. Functional motor defects in Shank3Δ11(-/-) mice could be rescued with the troponin activator Tirasemtiv that sensitizes muscle fibers to calcium. Our observations give insight into the function of SHANK3 besides the central nervous system and imply potential treatment strategies for SHANK3-associated ASD.
Collapse
Affiliation(s)
- Anne-Kathrin Lutz
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Stefanie Pfaender
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Berra Incearap
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Valentin Ioannidis
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Ilaria Ottonelli
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Karl J Föhr
- Department of Anesthesiology, Ulm University Hospital, 89081 Ulm, Germany
| | - Judith Cammerer
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Marvin Zoller
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Julia Higelin
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Federica Giona
- CNR Neuroscience Institute, University of Milan, 20129 Milan, Italy.,BIOMETRA University of Milan, 20129 Milan, Italy
| | - Maximilian Stetter
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | - Nicole Stoecker
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | | | - Michael Schön
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany
| | | | - Stefan Liebau
- Institute of Neuroanatomy and Developmental Biology, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Gotthold Barbi
- Institute for Human Genetics, Ulm University Hospital, 89081 Ulm, Germany
| | - Andreas M Grabrucker
- Cellular Neurobiology and Neuro-Nanotechnology Lab, Department of Biological Sciences, University of Limerick, V94PH61 Limerick, Ireland.,Bernal Institute, University of Limerick, V94T9PX Limerick, Ireland.,Health Research Institute (HRI), University of Limerick, V94T9PX Limerick, Ireland
| | - Richard Delorme
- Child and Adolescent Psychiatry Department, APHP, Robert-Debré Hospital, 750197 Paris, France
| | - Michael Fauler
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, 89075 Ulm, Germany
| | | | | | | | - Thomas Bourgeron
- Génétique Humaine et Fonctions Cognitives, Université Paris Diderot, Institut Pasteur, 75015 Paris, France
| | - Chiara Verpelli
- CNR Neuroscience Institute, University of Milan, 20129 Milan, Italy.,BIOMETRA University of Milan, 20129 Milan, Italy
| | - Maria Demestre
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany.
| | - Tobias M Boeckers
- Institute for Anatomy and Cell Biology, Ulm University, 89081 Ulm, Germany. .,DZNE, Ulm Site, 89081 Ulm, Germany
| |
Collapse
|
147
|
NMJ-Analyser identifies subtle early changes in mouse models of neuromuscular disease. Sci Rep 2021; 11:12251. [PMID: 34112844 PMCID: PMC8192785 DOI: 10.1038/s41598-021-91094-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
The neuromuscular junction (NMJ) is the peripheral synapse formed between a motor neuron axon terminal and a muscle fibre. NMJs are thought to be the primary site of peripheral pathology in many neuromuscular diseases, but innervation/denervation status is often assessed qualitatively with poor systematic criteria across studies, and separately from 3D morphological structure. Here, we describe the development of ‘NMJ-Analyser’, to comprehensively screen the morphology of NMJs and their corresponding innervation status automatically. NMJ-Analyser generates 29 biologically relevant features to quantitatively define healthy and aberrant neuromuscular synapses and applies machine learning to diagnose NMJ degeneration. We validated this framework in longitudinal analyses of wildtype mice, as well as in four different neuromuscular disease models: three for amyotrophic lateral sclerosis (ALS) and one for peripheral neuropathy. We showed that structural changes at the NMJ initially occur in the nerve terminal of mutant TDP43 and FUS ALS models. Using a machine learning algorithm, healthy and aberrant neuromuscular synapses are identified with 95% accuracy, with 88% sensitivity and 97% specificity. Our results validate NMJ-Analyser as a robust platform for systematic and structural screening of NMJs, and pave the way for transferrable, and cross-comparison and high-throughput studies in neuromuscular diseases.
Collapse
|
148
|
Jing H, Chen P, Hui T, Yu Z, Zhou J, Fei E, Wang S, Ren D, Lai X, Li B. Synapse-specific Lrp4 mRNA enrichment requires Lrp4/MuSK signaling, muscle activity and Wnt non-canonical pathway. Cell Biosci 2021; 11:105. [PMID: 34090516 PMCID: PMC8180081 DOI: 10.1186/s13578-021-00619-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 05/25/2021] [Indexed: 11/10/2022] Open
Abstract
Background The neuromuscular junction (NMJ) is a peripheral synapse critical to muscle contraction. Like acetylcholine receptors (AChRs), many essential proteins of NMJ are extremely concentrated at the postjunctional membrane. However, the mechanisms of synapse-specific concentration are not well understood; furthermore, it is unclear whether signaling molecules critical to NMJ formation and maintenance are also locally transcribed. Results We studied the β-gal activity encoded by a lacZ cassette driven by the promoter of the Lrp4 gene. As reported for Lrp4 mRNA, β-gal was in the central region in embryonic muscles and at the NMJ after its formation. However, β-gal was no longer in the central areas of muscle fibers in Lrp4 or MuSK mutant mice, indicating a requirement of Lrp4/MuSK signaling. This phenotype could be rescued by transgenic expression of LRP4 with a transmembrane domain but not soluble ECD in Lrp4 mutant mice. β-gal and AChR clusters were distributed in a broader region in lacZ/ECD than that of heterozygous lacZ/+ mice, indicating an important role of the transmembrane domain in Lrp4 signaling. Synaptic β-gal activity became diffused after denervation or treatment with µ-conotoxin, despite its mRNA was increased, indicating synaptic Lrp4 mRNA enrichment requires muscle activity. β-gal was also diffused in aged mice but became re-concentrated after muscle stimulation. Finally, Lrp4 mRNA was increased in C2C12 myotubes by Wnt ligands in a manner that could be inhibited by RKI-1447, an inhibitor of ROCK in Wnt non-canonical signaling. Injecting RKI-1447 into muscles of adult mice diminished Lrp4 synaptic expression. Conclusions This study demonstrates that synapse-specific enrichment of Lrp4 mRNA requires a coordinated interaction between Lrp4/MuSK signaling, muscle activity, and Wnt non-canonical signaling. Thus, the study provides a new mechanism for Lrp4 mRNA enrichment. It also provides a potential target for the treatment of NMJ aging and other NMJ-related diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00619-z.
Collapse
Affiliation(s)
- Hongyang Jing
- School of Life Science, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Peng Chen
- School of Life Science, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Tiankun Hui
- School of Life Science, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Zheng Yu
- School of Life Science, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Jin Zhou
- School of Life Science, Nanchang University, Nanchang, 330031, China.,Human Aging Research Institute, Nanchang University, Nanchang, 330031, China
| | - Erkang Fei
- School of Life Science, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Shunqi Wang
- School of Life Science, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Dongyan Ren
- School of Life Science, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Xinsheng Lai
- School of Life Science, Nanchang University, Nanchang, 330031, China. .,Institute of Life Science, Nanchang University, Nanchang, 330031, China.
| | - Baoming Li
- School of Life Science, Nanchang University, Nanchang, 330031, China. .,Institute of Life Science, Nanchang University, Nanchang, 330031, China. .,Department of Psychology and Institute of Brain Science, School of Education, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|
149
|
Rimington RP, Fleming JW, Capel AJ, Wheeler PC, Lewis MP. Bioengineered model of the human motor unit with physiologically functional neuromuscular junctions. Sci Rep 2021; 11:11695. [PMID: 34083648 PMCID: PMC8175425 DOI: 10.1038/s41598-021-91203-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/24/2021] [Indexed: 02/04/2023] Open
Abstract
Investigations of the human neuromuscular junction (NMJ) have predominately utilised experimental animals, model organisms, or monolayer cell cultures that fail to represent the physiological complexity of the synapse. Consequently, there remains a paucity of data regarding the development of the human NMJ and a lack of systems that enable investigation of the motor unit. This work addresses this need, providing the methodologies to bioengineer 3D models of the human motor unit. Spheroid culture of iPSC derived motor neuron progenitors augmented the transcription of OLIG2, ISLET1 and SMI32 motor neuron mRNAs ~ 400, ~ 150 and ~ 200-fold respectively compared to monolayer equivalents. Axon projections of adhered spheroids exceeded 1000 μm in monolayer, with transcription of SMI32 and VACHT mRNAs further enhanced by addition to 3D extracellular matrices in a type I collagen concentration dependent manner. Bioengineered skeletal muscles produced functional tetanic and twitch profiles, demonstrated increased acetylcholine receptor (AChR) clustering and transcription of MUSK and LRP4 mRNAs, indicating enhanced organisation of the post-synaptic membrane. The number of motor neuron spheroids, or motor pool, required to functionally innervate 3D muscle tissues was then determined, generating functional human NMJs that evidence pre- and post-synaptic membrane and motor nerve axon co-localisation. Spontaneous firing was significantly elevated in 3D motor units, confirmed to be driven by the motor nerve via antagonistic inhibition of the AChR. Functional analysis outlined decreased time to peak twitch and half relaxation times, indicating enhanced physiology of excitation contraction coupling in innervated motor units. Our findings provide the methods to maximise the maturity of both iPSC motor neurons and primary human skeletal muscle, utilising cell type specific extracellular matrices and developmental timelines to bioengineer the human motor unit for the study of neuromuscular junction physiology.
Collapse
Affiliation(s)
- Rowan P. Rimington
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| | - Jacob W. Fleming
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| | - Andrew J. Capel
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| | - Patrick C. Wheeler
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| | - Mark P. Lewis
- grid.6571.50000 0004 1936 8542National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU Leicestershire UK
| |
Collapse
|
150
|
Balch MHH, Harris H, Chugh D, Gnyawali S, Rink C, Nimjee SM, Arnold WD. Ischemic stroke-induced polyaxonal innervation at the neuromuscular junction is attenuated by robot-assisted mechanical therapy. Exp Neurol 2021; 343:113767. [PMID: 34044000 DOI: 10.1016/j.expneurol.2021.113767] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/30/2021] [Accepted: 05/22/2021] [Indexed: 01/12/2023]
Abstract
Ischemic stroke is a leading cause of disability world-wide. Mounting evidence supports neuromuscular pathology following stroke, yet mechanisms of dysfunction and therapeutic action remain undefined. The objectives of our study were to investigate neuromuscular pathophysiology following ischemic stroke and to evaluate the therapeutic effect of Robot-Assisted Mechanical massage Therapy (RAMT) on neuromuscular junction (NMJ) morphology. Using an ischemic stroke model in male rats, we demonstrated longitudinal losses of muscle contractility and electrophysiological estimates of motor unit number in paretic hindlimb muscles within 21 days of stroke. Histological characterization demonstrated striking pre- and postsynaptic alterations at the NMJ. Stroke prompted enlargement of motor axon terminals, acetylcholine receptor (AChR) area, and motor endplate size. Paretic muscle AChRs were also more homogenously distributed across motor endplates, exhibiting fewer clusters and less fragmentation. Most interestingly, NMJs in paretic muscle exhibited increased frequency of polyaxonal innervation. This finding of increased polyaxonal innervation in stroke-affected skeletal muscle suggests that reduction of motor unit number following stroke may be a spurious artifact due to overlapping of motor units rather than losses. Furthermore, we tested the effects of RAMT - which we recently showed to improve motor function and protect against subacute myokine disturbance - and found significant attenuation of stroke-induced NMJ alterations. RAMT not only normalized the post-stroke presentation of polyaxonal innervation but also mitigated postsynaptic expansion. These findings confirm complex neuromuscular pathophysiology after stroke, provide mechanistic direction for ongoing research, and inform development of future therapeutic strategies. SIGNIFICANCE: Ischemic stroke is a leading contributor to chronic disability, and there is growing evidence that neuromuscular pathology may contribute to the impact of stroke on physical function. Following ischemic stroke in a rat model, there are progressive declines of motor unit number estimates and muscle contractility. These changes are paralleled by striking pre- and postsynaptic maladaptive changes at the neuromuscular junction, including polyaxonal innervation. When administered to paretic hindlimb muscle, Robot-Assisted Mechanical massage Therapy - previously shown to improve motor function and protect against subacute myokine disturbance - prevents stroke-induced neuromuscular junction alterations. These novel observations provide insight into the neuromuscular response to cerebral ischemia, identify peripheral mechanisms of functional disability, and present a therapeutic rehabilitation strategy with clinical relevance.
Collapse
Affiliation(s)
- Maria H H Balch
- Department of Biomedical Education and Anatomy, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Hallie Harris
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Deepti Chugh
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Surya Gnyawali
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Cameron Rink
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Shahid M Nimjee
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - W David Arnold
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physical Medicine and Rehabilitation, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|