151
|
Xing G, Jing H, Yu Z, Chen P, Wang H, Xiong WC, Mei L. Membraneless condensates by Rapsn phase separation as a platform for neuromuscular junction formation. Neuron 2021; 109:1963-1978.e5. [PMID: 34033754 DOI: 10.1016/j.neuron.2021.04.021] [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: 01/05/2021] [Revised: 03/27/2021] [Accepted: 04/22/2021] [Indexed: 12/29/2022]
Abstract
Our daily life depends on muscle contraction, a process that is controlled by the neuromuscular junction (NMJ). However, the mechanisms of NMJ assembly remain unclear. Here we show that Rapsn, a protein critical for NMJ formation, undergoes liquid-liquid phase separation (LLPS) and condensates into liquid-like assemblies. Such assemblies can recruit acetylcholine receptors (AChRs), cytoskeletal proteins, and signaling proteins for postsynaptic differentiation. Rapsn LLPS requires multivalent binding of tetratricopeptide repeats (TPRs) and is increased by Musk signaling. The capacity of Rapsn to condensate and co-condensate with interaction proteins is compromised by mutations of congenital myasthenic syndromes (CMSs). NMJ formation is impaired in mutant mice carrying a CMS-associated, LLPS-deficient mutation. These results reveal a critical role of Rapsn LLPS in forming a synaptic semi-membraneless compartment for NMJ formation.
Collapse
Affiliation(s)
- Guanglin Xing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Hongyang Jing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zheng Yu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Peng Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA.
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA; Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH 44106, USA.
| |
Collapse
|
152
|
Pratt J, De Vito G, Narici M, Segurado R, Pessanha L, Dolan J, Conroy J, Boreham C. Plasma C-Terminal Agrin Fragment as an Early Biomarker for Sarcopenia: Results from the Genofit Study. J Gerontol A Biol Sci Med Sci 2021; 76:2090-2096. [PMID: 33993303 PMCID: PMC8599080 DOI: 10.1093/gerona/glab139] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Indexed: 12/21/2022] Open
Abstract
Barriers associated with direct muscle quantification have prevented a consistent implementation of therapeutic measures for sarcopenia. Recently, the relevance of circulating C-terminal agrin fragment (CAF) as an accessible screening method alternative for sarcopenia has gained credence. Accordingly, this study aimed to verify the pertinence of plasma CAF as a biomarker for sarcopenia. Three hundred healthy adults aged between 50 and 83 years took part in this study. Sarcopenia was diagnosed according to the European Working Group on Sarcopenia in Older People criteria. Body composition was assessed using dual-energy x-ray absorptiometry, while muscle strength was examined using hand dynamometry. Plasma CAF concentrations were determined using a commercially available ELISA kit. CAF concentrations were significantly associated with appendicular lean mass (ALM), but not grip strength (p = .028, p = .575, respectively). Plasma CAF concentrations were significantly elevated in sarcopenic individuals compared to nonsarcopenic (p < .001). Overall, individuals with low grip strength or low ALM displayed significantly higher CAF levels compared to healthy controls, after adjusting for age and body mass index (p = .027, p = .003, respectively). In males, those with low grip strength or low ALM had significantly elevated CAF levels (p = .039, p = .027, respectively), while in females, only those with low ALM had significantly raised CAF concentrations, compared to healthy controls (p = .035). Our findings illuminate the potential relevance of CAF as an accessible biomarker for skeletal muscle health. CAF determination may enhance clinical practice by facilitating more widespread treatment strategies for sarcopenia. Nevertheless, future research is needed to confirm the diagnostic pertinence of CAF concentrations in screening for sarcopenia.
Collapse
Affiliation(s)
- Jedd Pratt
- Institute for Sport and Health, University College Dublin, Dublin, Ireland.,Genuity Science, Dublin, Ireland
| | - Giuseppe De Vito
- Department of Biomedical Sciences, CIR-Myo Myology Centre, Neuromuscular Physiology Laboratory, University of Padova, Padua, Italy
| | - Marco Narici
- Department of Biomedical Sciences, CIR-Myo Myology Centre, Neuromuscular Physiology Laboratory, University of Padova, Padua, Italy
| | - Ricardo Segurado
- Centre for Support and Training in Analysis and Research, and School of Public Health, Physiotherapy and Sports Sciences, University College Dublin, Dublin, Ireland
| | - Ludmilla Pessanha
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | | | | | - Colin Boreham
- Institute for Sport and Health, University College Dublin, Dublin, Ireland
| |
Collapse
|
153
|
Hui T, Jing H, Zhou T, Chen P, Liu Z, Dong X, Yan M, Ren D, Zou S, Wang S, Fei E, Hong D, Lai X. Increasing LRP4 diminishes neuromuscular deficits in a mouse model of Duchenne muscular dystrophy. Hum Mol Genet 2021; 30:1579-1590. [PMID: 33987657 PMCID: PMC8369839 DOI: 10.1093/hmg/ddab135] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular disease characterized by progressive wasting of skeletal muscles. The neuromuscular junction (NMJ) is a synapse between motor neurons and skeletal muscle fibers, critical for the control of muscle contraction. The NMJ decline is observed in DMD patients, but the mechanism is unclear. LRP4 serves as a receptor for agrin, a proteoglycan secreted by motor neurons to induce NMJ, and plays a critical role in NMJ formation and maintenance. Interestingly, we found that protein levels of LRP4 were reduced both in muscles of the DMD patients and DMD model mdx mice. We explored whether increasing LRP4 is beneficial for DMD and crossed muscle-specific LRP4 transgenic mice with mdx mice (mdx; HSA-LRP4). The LRP4 transgene increased muscle strength, together with improved neuromuscular transmission in mdx mice. Furthermore, we found the LRP4 expression mitigated NMJ fragments and denervation in mdx mice. Mechanically, we showed that overexpression of LRP4 increased the activity of MuSK and expression of dystrophin-associated glycoprotein complex proteins in the mdx mice. Overall, our findings suggest that increasing LRP4 improves both function and structure of NMJ in the mdx mice and Agrin signaling might serve as a new therapeutic strategy in DMD.
Collapse
Affiliation(s)
- Tiankun Hui
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Hongyang Jing
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Tian Zhou
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Peng Chen
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Ziyang Liu
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Xia Dong
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China.,School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Min Yan
- Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China.,School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Dongyan Ren
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Suqi Zou
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Shunqi Wang
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Erkang Fei
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Daojun Hong
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xinsheng Lai
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
154
|
Saini J, Faroni A, Reid AJ, Mouly V, Butler-Browne G, Lightfoot AP, McPhee JS, Degens H, Al-Shanti N. Cross-talk between motor neurons and myotubes via endogenously secreted neural and muscular growth factors. Physiol Rep 2021; 9:e14791. [PMID: 33931983 PMCID: PMC8087923 DOI: 10.14814/phy2.14791] [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: 12/09/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023] Open
Abstract
Neuromuscular junction (NMJ) research is vital to advance the understanding of neuromuscular patho‐physiology and development of novel therapies for diseases associated with NM dysfunction. In vivo, the micro‐environment surrounding the NMJ has a significant impact on NMJ formation and maintenance via neurotrophic and differentiation factors that are secreted as a result of cross‐talk between muscle fibers and motor neurons. Recently we showed the formation of functional NMJs in vitro in a co‐culture of immortalized human myoblasts and motor neurons from rat‐embryo spinal‐cord explants, using a culture medium free from serum and neurotrophic or growth factors. The aim of this study was to assess how functional NMJs were established in this co‐culture devoid of exogenous neural growth factors. To investigate this, an ELISA‐based microarray was used to compare the composition of soluble endogenously secreted growth factors in this co‐culture with an a‐neural muscle culture. The levels of seven neurotrophic factors brain‐derived neurotrophic factor (BDNF), glial‐cell‐line‐derived neurotrophic factor (GDNF), insulin‐like growth factor‐binding protein‐3 (IGFBP‐3), insulin‐like growth factor‐1 (IGF‐1), neurotrophin‐3 (NT‐3), neurotrophin‐4 (NT‐4), and vascular endothelial growth factor (VEGF) were higher (p < 0.05) in the supernatant of NMJ culture compared to those in the supernatant of the a‐neural muscle culture. This indicates that the cross‐talk between muscle and motor neurons promotes the secretion of soluble growth factors contributing to the local microenvironment thereby providing a favourable regenerative niche for NMJs formation and maturation.
Collapse
Affiliation(s)
- Jasdeep Saini
- Musculoskeletal Science & Sports Medicine Research Centre, Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Alessandro Faroni
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.,Dept. of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Adam J Reid
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.,Dept. of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Vincent Mouly
- Center for Research in Myology, Sorbonne Université-INSERM, Paris, France
| | | | - Adam P Lightfoot
- Musculoskeletal Science & Sports Medicine Research Centre, Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Jamie S McPhee
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, UK
| | - Hans Degens
- Musculoskeletal Science & Sports Medicine Research Centre, Department of Life Sciences, Manchester Metropolitan University, Manchester, UK.,Lithuanian Sports University, Institute of Sport Science and Innovations, Kaunas, Lithuania
| | - Nasser Al-Shanti
- Musculoskeletal Science & Sports Medicine Research Centre, Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
155
|
Hui T, Jing H, Lai X. Neuromuscular junction-specific genes screening by deep RNA-seq analysis. Cell Biosci 2021; 11:81. [PMID: 33933147 PMCID: PMC8088568 DOI: 10.1186/s13578-021-00590-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/17/2021] [Indexed: 01/17/2023] Open
Abstract
Background Neuromuscular junctions (NMJs) are chemical synapses formed between motor neurons and skeletal muscle fibers and are essential for controlling muscle contraction. NMJ dysfunction causes motor disorders, muscle wasting, and even breathing difficulties. Increasing evidence suggests that many NMJ disorders are closely related to alterations in specific gene products that are highly concentrated in the synaptic region of the muscle. However, many of these proteins are still undiscovered. Thus, screening for NMJ-specific proteins is essential for studying NMJ and the pathogenesis of NMJ diseases. Results In this study, synaptic regions (SRs) and nonsynaptic regions (NSRs) of diaphragm samples from newborn (P0) and adult (3-month-old) mice were used for RNA-seq. A total of 92 and 182 genes were identified as differentially expressed between the SR and NSR in newborn and adult mice, respectively. Meanwhile, a total of 1563 genes were identified as differentially expressed between the newborn SR and adult SR. Gene Ontology (GO) enrichment analyses, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and gene set enrichment analysis (GSEA) of the DEGs were performed. Protein–protein interaction (PPI) networks were constructed using STRING and Cytoscape. Further analysis identified some novel proteins and pathways that may be important for NMJ development, maintenance and maturation. Specifically, Sv2b, Ptgir, Gabrb3, P2rx3, Dlgap1 and Rims1 may play roles in NMJ development. Hcn1 may localize to the muscle membrane to regulate NMJ maintenance. Trim63, Fbxo32 and several Asb family proteins may regulate muscle developmental-related processes. Conclusion Here, we present a complete dataset describing the spatiotemporal transcriptome changes in synaptic genes and important synaptic pathways. The neuronal projection-related pathway, ion channel activity and neuroactive ligand-receptor interaction pathway are important for NMJ development. The myelination and voltage-gated ion channel activity pathway may be important for NMJ maintenance. These data will facilitate the understanding of the molecular mechanisms underlying the development and maintenance of NMJ and the pathogenesis of NMJ disorders.
Collapse
Affiliation(s)
- Tiankun Hui
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Hongyang Jing
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Xinsheng Lai
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China. .,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
156
|
Abstract
PURPOSE OF REVIEW This review encompasses the main novelties regarding nonimmune mechanisms implicated in the pathogenesis of idiopathic inflammatory myopathies (IIM). RECENT FINDINGS In recent years, growing data support a role for endoplasmic-reticulum (ER) stress as a propagator of muscular damage, together with the release of interferon type I and reactive oxygen species in hypoxemic muscle fibers. Other studies evaluating the relationship between autophagy and Toll-like receptors (TLRs) in IIM subtypes have shown increased TLR3 and TLR4 expression in fibers of IIM patients and colocalization with LC3, an autophagy marker, submitting autophagy as a likely player in IIM pathogenesis. Most novel evidences concern the potential role of denervation of the neuromuscular junction in IIM, possibly connected to hyperexpression of MHC-I, and trafficking of extracellular vesicles, which may represent a connection between nonimmune and immune-mediated mechanisms of muscle inflammation and damage. SUMMARY Nonimmune mechanisms contribute to the pathogenesis of IIM, likely cooperating with immune-mediated inflammation. Consistent data were released for ER stress, autophagy, mitochondrial dysfunction and hypoxia; in addition to, neuromuscular denervation and extracellular vesicles have been proposed as thoughtful links between muscle inflammation, damage and atrophy. Further understanding of nonimmune abnormalities and potential reversible pathways is needed to improve the management of IIM.
Collapse
|
157
|
Gender-specific analysis for the association between trunk muscle mass and spinal pathologies. Sci Rep 2021; 11:7816. [PMID: 33837250 PMCID: PMC8035169 DOI: 10.1038/s41598-021-87334-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/24/2021] [Indexed: 12/25/2022] Open
Abstract
We investigated the relationship between trunk muscle mass and spinal pathologies by gender. This multicenter cross-sectional study included patients aged ≥ 30 years who visited a spinal outpatient clinic. Trunk and appendicular muscle mass were measured using bioelectrical impedance analysis. The Oswestry Disability Index (ODI), visual analog scale (VAS) score for low back pain, sagittal vertical axis (SVA), and EuroQol 5 Dimension (EQ5D) score were investigated to evaluate spinal pathology. The association between trunk muscle mass and these parameters was analyzed by gender using a non-linear regression model adjusted for patients’ demographics. We investigated the association between age and trunk muscle mass. We included 781 men and 957 women. Trunk muscle mass differed significantly between men and women, although it decreased with age after age 70 in both genders. Lower trunk muscle mass was significantly associated with ODI, SVA, and EQ5D score deterioration in both genders; its association with VAS was significant only in men. Most parameters deteriorated when trunk muscle mass was < 26 kg in men and < 19 kg in women. Lower trunk muscle mass was associated with lumbar disability, spinal imbalance, and poor quality of life in both genders, with significant difference in muscle mass.
Collapse
|
158
|
Pinto CG, Leite APS, Sartori AA, Tibúrcio FC, Barraviera B, Junior RSF, Filadelpho AL, de Carvalho SC, Matheus SMM. Heterologous fibrin biopolymer associated to a single suture stitch enables the return of neuromuscular junction to its mature pattern after peripheral nerve injury. Injury 2021; 52:731-737. [PMID: 33902866 DOI: 10.1016/j.injury.2020.10.070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/26/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
Denervation leads to severe atrophy of neuromuscular junction (NMJ) structure including decrease of the expression of fundamental proteins. Up to now, conventional suture has been the gold standard method used to correct this injury. Fibrin sealant is one of the alternatives proposed to optimize this method. This study verified if the association of fibrin sealant - Heterologous Fibrin Biopolymer (HFB) and a single suture stitch promotes return of morphology and NMJ structure to mature pattern after peripheral nerve injury. Forty Wistar rats were distributed into 4 groups: Sham-Control (SC), Denervated-Control (DC), Suture-Lesion (SL) and Suture-Lesion + HFB (SFS). In SC group only the right sciatic nerve identification was done. In DC, SL and SFS groups fixation of nerve stumps on musculature immediately after neurotmesis was performed. After seven days, stump reconnection with 3 stitches in SL and a single stitch associated with HFB in SFS were done. After sixty days right soleus muscles were prepared for nicotinic acetylcholine receptors (nAChRs) and nerve terminal confocal analyses, and for nAChRs (α1, ε e γ), S100, Agrin, LRP-4, MMP-3, Rapsyn western blotting analyses. SC group presented normal morphology. In DC group it was observed flattening of NMJ, fragmentation of nAChRs and tangled nerve terminals. The majority of the parameters of SL and SFS groups presented values in between SC and DC groups. There was an increase of relative planar area in these groups (SL and SFS) highlighting that there was less nAChRs fragmentation and the values of protein expression showed return of nAChRs to mature pattern. Use of HFB associated with a single suture stitch decreased surgical time, minimized suture injuries, did not alter nerve regeneration and presented potential to reestablish the NMJ apparatus. These consolidated results encourage surgeons to develop future clinical trials to install definitively this new approach both for reconstructive surgery and neurosurgery.
Collapse
Affiliation(s)
- Carina Guidi Pinto
- Graduate Program in Surgery and Translational Medicine, Medical School, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil; Department of Structural and Functional Biology (Anatomy Sector), Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Ana Paula Silveira Leite
- Graduate Program in Surgery and Translational Medicine, Medical School, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil; Department of Structural and Functional Biology (Anatomy Sector), Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Arthur Alves Sartori
- Department of Structural and Functional Biology (Anatomy Sector), Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Felipe Cantore Tibúrcio
- Graduate Program in Surgery and Translational Medicine, Medical School, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil; Department of Structural and Functional Biology (Anatomy Sector), Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Benedito Barraviera
- Center for the Studies of Venoms and Venomous Animals (CEVAP), São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - Rui Seabra Ferreira Junior
- Center for the Studies of Venoms and Venomous Animals (CEVAP), São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | - André Luis Filadelpho
- Department of Structural and Functional Biology (Anatomy Sector), Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil
| | | | - Selma Maria Michelin Matheus
- Department of Structural and Functional Biology (Anatomy Sector), Institute of Biosciences, São Paulo State University (Unesp), Botucatu, São Paulo, Brazil.
| |
Collapse
|
159
|
DePew AT, Mosca TJ. Conservation and Innovation: Versatile Roles for LRP4 in Nervous System Development. J Dev Biol 2021; 9:9. [PMID: 33799485 PMCID: PMC8006230 DOI: 10.3390/jdb9010009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
As the nervous system develops, connections between neurons must form to enable efficient communication. This complex process of synaptic development requires the coordination of a series of intricate mechanisms between partner neurons to ensure pre- and postsynaptic differentiation. Many of these mechanisms employ transsynaptic signaling via essential secreted factors and cell surface receptors to promote each step of synaptic development. One such cell surface receptor, LRP4, has emerged as a synaptic organizer, playing a critical role in conveying extracellular signals to initiate diverse intracellular events during development. To date, LRP4 is largely known for its role in development of the mammalian neuromuscular junction, where it functions as a receptor for the synaptogenic signal Agrin to regulate synapse development. Recently however, LRP4 has emerged as a synapse organizer in the brain, where new functions for the protein continue to arise, adding further complexity to its already versatile roles. Additional findings indicate that LRP4 plays a role in disorders of the nervous system, including myasthenia gravis, amyotrophic lateral sclerosis, and Alzheimer's disease, demonstrating the need for further study to understand disease etiology. This review will highlight our current knowledge of how LRP4 functions in the nervous system, focusing on the diverse developmental roles and different modes this essential cell surface protein uses to ensure the formation of robust synaptic connections.
Collapse
Affiliation(s)
| | - Timothy J. Mosca
- Department of Neuroscience, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| |
Collapse
|
160
|
Narayanan N, Lengemann P, Kim KH, Kuang L, Sobreira T, Hedrick V, Aryal UK, Kuang S, Deng M. Harnessing nerve-muscle cell interactions for biomaterials-based skeletal muscle regeneration. J Biomed Mater Res A 2021; 109:289-299. [PMID: 32490576 DOI: 10.1002/jbm.a.37022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 12/30/2022]
Abstract
Nerve cells secrete neurotrophic factors that play a critical role in neuronal survival, proliferation, and regeneration. However, their role in regulating myoblast behavior and skeletal muscle repair remains largely unexplored. In the present study, we investigated the effects of PC12 secreted signaling factors in modulating C2C12 myoblast behavior under physiologically relevant conditions. We showed that PC12 conditioned media modulated myoblast proliferation and differentiation in both 2D culture and 3D aligned electrospun fiber scaffold system in a dose-dependent manner. We further developed a biomimetic, tunable hydrogel consisting of hyaluronic acid, chondroitin sulfate, and polyethylene glycol as a 3D matrix encapsulating PC12 cells. The hydrogel-encapsulated PC12 cells promoted survival and proliferation of myoblasts in co-culture. Further proteomics analysis identified a total of 2,088 proteins from the secretome of the encapsulated PC12 cells and revealed the biological role and overlapping functions of nerve-secreted proteins for skeletal muscle regeneration, potentially through regulating myoblast behavior, nerve function, and angiogenesis. These experiments provide insights into the nerve-muscle interactions and pave the way for developing advanced biomaterials strategies incorporating nerve cell secretome for accelerated skeletal muscle regeneration.
Collapse
Affiliation(s)
- Naagarajan Narayanan
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, USA
| | - Paul Lengemann
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, USA
| | - Kun Ho Kim
- Department of Animal Science, Purdue University, West Lafayette, Indiana, USA
| | - Liangju Kuang
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, USA
| | - Tiago Sobreira
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, USA
| | - Victoria Hedrick
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, USA
| | - Uma K Aryal
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, USA
| | - Shihuan Kuang
- Department of Animal Science, Purdue University, West Lafayette, Indiana, USA
| | - Meng Deng
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana, USA
- Bindley Bioscience Center, Purdue University, West Lafayette, Indiana, USA
- School of Materials Engineering, Purdue University, West Lafayette, Indiana, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
161
|
Börsch A, Ham DJ, Mittal N, Tintignac LA, Migliavacca E, Feige JN, Rüegg MA, Zavolan M. Molecular and phenotypic analysis of rodent models reveals conserved and species-specific modulators of human sarcopenia. Commun Biol 2021; 4:194. [PMID: 33580198 PMCID: PMC7881157 DOI: 10.1038/s42003-021-01723-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Sarcopenia, the age-related loss of skeletal muscle mass and function, affects 5-13% of individuals aged over 60 years. While rodents are widely-used model organisms, which aspects of sarcopenia are recapitulated in different animal models is unknown. Here we generated a time series of phenotypic measurements and RNA sequencing data in mouse gastrocnemius muscle and analyzed them alongside analogous data from rats and humans. We found that rodents recapitulate mitochondrial changes observed in human sarcopenia, while inflammatory responses are conserved at pathway but not gene level. Perturbations in the extracellular matrix are shared by rats, while mice recapitulate changes in RNA processing and autophagy. We inferred transcription regulators of early and late transcriptome changes, which could be targeted therapeutically. Our study demonstrates that phenotypic measurements, such as muscle mass, are better indicators of muscle health than chronological age and should be considered when analyzing aging-related molecular data.
Collapse
Affiliation(s)
- Anastasiya Börsch
- Biozentrum, University of Basel and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Daniel J Ham
- Biozentrum, University of Basel, Basel, Switzerland
| | - Nitish Mittal
- Biozentrum, University of Basel and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Lionel A Tintignac
- Department of Biomedicine, Pharmazentrum, University of Basel, Basel, Switzerland
| | | | - Jérôme N Feige
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| | | | - Mihaela Zavolan
- Biozentrum, University of Basel and Swiss Institute of Bioinformatics, Basel, Switzerland.
| |
Collapse
|
162
|
Martinez-Pena y Valenzuela I, Akaaboune M. The Metabolic Stability of the Nicotinic Acetylcholine Receptor at the Neuromuscular Junction. Cells 2021; 10:cells10020358. [PMID: 33572348 PMCID: PMC7916148 DOI: 10.3390/cells10020358] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 11/16/2022] Open
Abstract
The clustering and maintenance of nicotinic acetylcholine receptors (AChRs) at high density in the postsynaptic membrane is a hallmark of the mammalian neuromuscular junction (NMJ). The regulation of receptor density/turnover rate at synapses is one of the main thrusts of neurobiology because it plays an important role in synaptic development and synaptic plasticity. The state-of-the-art imaging revealed that AChRs are highly dynamic despite the overall structural stability of the NMJ over the lifetime of the animal. This review highlights the work on the metabolic stability of AChRs at developing and mature NMJs and discusses the role of synaptic activity and the regulatory signaling pathways involved in the dynamics of AChRs.
Collapse
Affiliation(s)
| | - Mohammed Akaaboune
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA;
- Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence: ; Tel.: +1-73-(46)-478512; Fax: +1-73-(46)-470884
| |
Collapse
|
163
|
Skoglund E, Grönholdt-Klein M, Rullman E, Thornell LE, Strömberg A, Hedman A, Cederholm T, Ulfhake B, Gustafsson T. Longitudinal Muscle and Myocellular Changes in Community-Dwelling Men Over Two Decades of Successful Aging-The ULSAM Cohort Revisited. J Gerontol A Biol Sci Med Sci 2021; 75:654-663. [PMID: 31002330 DOI: 10.1093/gerona/glz068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Indexed: 12/25/2022] Open
Abstract
Participants of the population-based Uppsala longitudinal study of adult men (ULSAM) cohort reaching more than 88 years of age (survivors, S) were investigated at age 70, 82, and 88-90 and compared at 70 years with non-survivors (NS) not reaching 82 years. Body composition, muscle mass and muscle histology were remarkably stable over 18 years of advanced aging in S. Analysis of genes involved in muscle remodeling showed that S had higher mRNA levels of myogenic differentiation factors (Myogenin, MyoD), embryonic myosin (eMyHC), enzymes involved in regulated breakdown of myofibrillar proteins (Smad2, Trim32, MuRF1,) and NCAM compared with healthy adult men (n = 8). S also had higher mRNA levels of eMyHC, Smad 2, MuRF1 compared with NS. At 88 years, S expressed decreased levels of Myogenin, MyoD, eMyHC, NCAM and Smad2 towards those seen in NS at 70 years. The gene expression pattern of S at 70 years was likely beneficial since they maintained muscle fiber histology and appendicular lean body mass until advanced age. The expression pattern at 88 years may indicate a diminished muscle remodeling coherent with a decline of reinnervation capacity and/or plasticity at advanced age.
Collapse
Affiliation(s)
- Elisabeth Skoglund
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden.,Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Sweden.,Department of Integrative Medical Biology, Umeå University, Sweden
| | | | - Eric Rullman
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Anna Strömberg
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Anu Hedman
- Heart Centre East-Tallinn Central Hospital, Estonia
| | - Tommy Cederholm
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Sweden
| | - Brun Ulfhake
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Gustafsson
- Department of Laboratory Medicine, Division of Clinical Physiology, Karolinska Institutet, and Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
164
|
Fabiś J, Danilewicz M, Niedzielski KR, Waszczykowski M, Fabiś-Strobin A, Bogucki A. The eccentric mechanotransduction, neuro-muscular transmission, and structural reversibility of muscle fatty infiltration. An experimental advanced disuse muscle-wasting model of rabbit supraspinatus. Arch Med Sci 2021; 17:1400-1407. [PMID: 34522269 PMCID: PMC8425242 DOI: 10.5114/aoms/131956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/26/2020] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Full-thickness rotator cuff tear is present in almost 50% of patients over age 65 years, and its degree is known to be a good predictor of the severity of muscle-wasting (MW) sarcopaenia, also known as fatty degeneration (FD). A FD CT grade > 2° is recognized as a borderline of its reversibility. A disuse model of supraspinatus FD (grade 2) in rabbits provides clinically relevant data. Therefore, the present study evaluates the correlation between eccentric mechanotransduction, neuromuscular transmission (NT), and reversibility of muscle fatty infiltration (MFI) in rabbit supraspinatus FD > 2°. MATERIAL AND METHODS The supraspinatus tendon was detached from the greater tubercle, infraspinatus, and subscapularis in 16 rabbits. The tendon was reinserted after 12 weeks, and the animals were euthanized 24 weeks after reconstruction. MFI was measured in the middle part of the supraspinatus. Single-fibre EMG (SFEMG) examination of the supraspinatus NT was performed on 4 animals. RESULTS The power of analysis was 99%. Significant differences in MFI volume were found between the operated (4.6 ±1.1%) and the opposite control sides (2.91 ±0.61%) (p < 0.001). SFEMG revealed no significant differences between the disuse and the control supraspinatus muscles (p > 0.05); however, 6.5% of the examined muscle fibres exhibited NT disorders combined with blockade of conduction in 2.5% of muscle fibres. CONCLUSIONS Critical MFI in a disuse model of rabbit supraspinatus FD, CT grade > 2°, is substantially reversible by eccentric training despite subclinical impairment of neuromuscular transmission. In addition, 0.63% reversal of MFI is correlated with 1% hypertrophy of type I and II muscle fibre diameter.
Collapse
Affiliation(s)
- Jarosław Fabiś
- Department of Arthroscopy, Minimally Invasive Surgery and Sports Traumatology Medical University of Lodz, Lodz, Poland
| | - Marian Danilewicz
- Morphometry Division, Department of Pathology, Medical University of Lodz, Lodz, Poland
| | - Kryspin R. Niedzielski
- Clinic of Orthopaedic and Traumatology Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | - Michał Waszczykowski
- Department of Arthroscopy, Minimally Invasive Surgery and Sports Traumatology Medical University of Lodz, Lodz, Poland
| | - Anna Fabiś-Strobin
- Clinic of Orthopaedic and Traumatology Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | - Andrzej Bogucki
- Department of Extrapyramidal Diseases, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
165
|
Lu S, Lyu Z, Wang Z, Kou Y, Liu C, Li S, Hu M, Zhu H, Wang W, Zhang C, Kuan YS, Liu YW, Chen J, Tian J. Lipin 1 deficiency causes adult-onset myasthenia with motor neuron dysfunction in humans and neuromuscular junction defects in zebrafish. Theranostics 2021; 11:2788-2805. [PMID: 33456573 PMCID: PMC7806489 DOI: 10.7150/thno.53330] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/12/2020] [Indexed: 12/03/2022] Open
Abstract
Lipin 1 is an intracellular protein acting as a phosphatidic acid phosphohydrolase enzyme controlling lipid metabolism. Human recessive mutations in LPIN1 cause recurrent, early-onset myoglobinuria, a condition normally associated with muscle pain and weakness. Whether and how lipin 1 deficiency in humans leads to peripheral neuropathy is yet unclear. Herein, two novel compound heterozygous mutations in LPIN1 with neurological disorders, but no myoglobinuria were identified in an adult-onset syndromic myasthenia family. The present study sought to explore the pathogenic mechanism of LPIN1 in muscular and neural development. Methods: The clinical diagnosis of the proband was compared to the known 48 cases of LPIN1 recessive homozygous mutations. Whole-exome sequencing was carried out on the syndromic myasthenia family to identify the causative gene. The pathogenesis of lipin 1 deficiency during somitogenesis and neurogenesis was investigated using the zebrafish model. Whole-mount in situ hybridization, immunohistochemistry, birefringence analysis, touch-evoke escape response and locomotion assays were performed to observe in vivo the changes in muscles and neurons. The conservatism of the molecular pathways regulated by lipin 1 was evaluated in human primary glioblastoma and mouse myoblast cells by siRNA knockdown, drug treatment, qRT-PCR and Western blotting analysis. Results: The patient exhibited adult-onset myasthenia accompanied by muscle fiber atrophy and nerve demyelination without myoglobinuria. Two novel heterozygous mutations, c.2047A>C (p.I683L) and c.2201G>A (p.R734Q) in LPIN1, were identified in the family and predicted to alter the tertiary structure of LPIN1 protein. Lipin 1 deficiency in zebrafish embryos generated by lpin1 morpholino knockdown or human LPIN1 mutant mRNA injections reproduced the myotomes defects, a reduction both in primary motor neurons and secondary motor neurons projections, morphological changes of post-synaptic clusters of acetylcholine receptors, and myelination defects, which led to reduced touch-evoked response and abnormalities of swimming behaviors. Loss of lipin 1 function in zebrafish and mammalian cells also exhibited altered expression levels of muscle and neuron markers, as well as abnormally enhanced Notch signaling, which was partially rescued by the specific Notch pathway inhibitor DAPT. Conclusions: These findings pointed out that the compound heterozygous mutations in human LPIN1 caused adult-onset syndromic myasthenia with peripheral neuropathy. Moreover, zebrafish could be used to model the neuromuscular phenotypes due to the lipin 1 deficiency, where a novel pathological role of over-activated Notch signaling was discovered and further confirmed in mammalian cell lines.
Collapse
|
166
|
Kim M, Franke V, Brandt B, Lowenstein ED, Schöwel V, Spuler S, Akalin A, Birchmeier C. Single-nucleus transcriptomics reveals functional compartmentalization in syncytial skeletal muscle cells. Nat Commun 2020; 11:6375. [PMID: 33311457 PMCID: PMC7732842 DOI: 10.1038/s41467-020-20064-9] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 11/09/2020] [Indexed: 11/25/2022] Open
Abstract
Syncytial skeletal muscle cells contain hundreds of nuclei in a shared cytoplasm. We investigated nuclear heterogeneity and transcriptional dynamics in the uninjured and regenerating muscle using single-nucleus RNA-sequencing (snRNAseq) of isolated nuclei from muscle fibers. This revealed distinct nuclear subtypes unrelated to fiber type diversity, previously unknown subtypes as well as the expected ones at the neuromuscular and myotendinous junctions. In fibers of the Mdx dystrophy mouse model, distinct subtypes emerged, among them nuclei expressing a repair signature that were also abundant in the muscle of dystrophy patients, and a nuclear population associated with necrotic fibers. Finally, modifications of our approach revealed the compartmentalization in the rare and specialized muscle spindle. Our data identifies nuclear compartments of the myofiber and defines a molecular roadmap for their functional analyses; the data can be freely explored on the MyoExplorer server ( https://shiny.mdc-berlin.de/MyoExplorer/ ).
Collapse
Affiliation(s)
- Minchul Kim
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Vedran Franke
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Bettina Brandt
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Elijah D Lowenstein
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Verena Schöwel
- Muscle Research Unit, Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Altuna Akalin
- Berlin Institute for Medical Systems Biology, Max Delbrueck Center for Molecular Medicine, Berlin, Germany.
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
167
|
Yan M, Guo A, Chen P, Jing H, Ren D, Zhong Y, Wu Y, Fei E, Lai X, Zou S, Wang S. LRP4 LDLα repeats of astrocyte enhance dendrite arborization of the neuron. Mol Brain 2020; 13:166. [PMID: 33302985 PMCID: PMC7730773 DOI: 10.1186/s13041-020-00708-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
The low-density lipoprotein receptor-related protein 4 (LRP4) is essential for inducing the neuromuscular junction (NMJ) formation in muscle fibers, and LRP4 plays a critical role in dendritic development and synaptogenesis in the central nervous system (CNS). As a single transmembrane protein, LRP4 contains an enormously sizeable extracellular domain (ECD), containing multiple LDLα repeats in the N-terminal of ECD. LRP4 only with extracellular domain acts as a similar mechanism of full-length LRP4 in muscles to stimulate acetylcholine receptor clustering. In this study, we elucidated that LDLα repeats of LRP4 maintained the body weight and survival rate. Dendritic branches of the pyramidal neurons in Lrp4-null mice with LRP4 LDLα repeats residue were more than in Lrp4-null mice without residual LRP4 domain. Supplement with conditioned medium from LRP4 LDLα overexpression cells, the primary culture pyramidal neurons achieved strong dendritic arborization ability. Besides, astrocytes with LRP4 LDLα repeats residue could promote pyramidal neuronal dendrite arborization in the primary co-cultured system. These observations signify that LRP4 LDLα repeats play a prominent underlying role in dendrite arborization.
Collapse
Affiliation(s)
- Min Yan
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,School of Basic Medical Sciences, Nanchang University, Nanchang, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Amin Guo
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Peng Chen
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Hongyang Jing
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Dongyan Ren
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Yanzi Zhong
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Yongqiang Wu
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Erkang Fei
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Xinsheng Lai
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Suqi Zou
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China
| | - Shunqi Wang
- School of Life Sciences, Nanchang University, Nanchang, Jiangxi, China. .,Institute of Life Sciences, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
168
|
Bao Z, Cui C, Chow SKH, Qin L, Wong RMY, Cheung WH. AChRs Degeneration at NMJ in Aging-Associated Sarcopenia-A Systematic Review. Front Aging Neurosci 2020; 12:597811. [PMID: 33362532 PMCID: PMC7759742 DOI: 10.3389/fnagi.2020.597811] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/19/2020] [Indexed: 12/16/2022] Open
Abstract
Sarcopenia is an aging process with a decline of skeletal muscle mass and function, which is a challenging public health problem with reduced quality of life in patients. The endplate, the post-synaptic part of the neuromuscular junction (NMJ), occupies 0.1% of the myofiber surface area only, but is composed of millions of acetylcholine receptors (AChRs) that are efficient in binding to acetylcholine (ACh) and triggering skeletal muscle contraction. This systematic review aims to examine aging-associated alterations of post-synaptic AChRs, including morphology, function and related gene expression. A systematic literature search was conducted in PubMed, Embase and Web of Science with relevant keywords by two independent reviewers. Original pre-clinical and clinical studies regarding AChRs changes during aging with available full text and written in English were included. Information was extracted from the included studies for further review. In total, 30 articles were included. Various parameters assessing AChRs alterations by radioassay, immunofluorescence, electrophysiology and mechanical test were reported. Endplate fragmentation and denervation were common in old skeletal muscles during aging. To ensure efficient NMJ transmission and force generation, type I or IIb muscle fibers tended to have increased ACh quanta releasing after electrical stimulations, while type IIa muscle fibers tended to have stronger binding between ACh and AChRs, but the overall function of AChRs was reduced during aging. Alterations of AChRs area depended on muscle type, species and the progress of muscle atrophy and type I muscles fibers tended to demonstrate enlarging AChRs areas. Myogenic regulator factors (MRFs) can regulate the expression of AChRs subunits, while decreased MRF4 may lead to expression changes of AChRs subunits during aging. Sarcoglycan-α can delay low-density lipoprotein receptor-related protein 4 (LRP4) degradation. This protein was increased in old muscles but still cannot suppress the degradation of LRP4. Investigating the role of these AChRs-related genes in the process of aging may provide a potential target to treat sarcopenia.
Collapse
Affiliation(s)
- Zhengyuan Bao
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Can Cui
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Simon Kwoon-Ho Chow
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Ling Qin
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Ronald Man Yeung Wong
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing-Hoi Cheung
- Department of Orthopaedics and Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
169
|
Bauché S, Sureau A, Sternberg D, Rendu J, Buon C, Messéant J, Boëx M, Furling D, Fauré J, Latypova X, Gelot AB, Mayer M, Mary P, Whalen S, Fournier E, Cloix I, Remerand G, Laffargue F, Nougues MC, Fontaine B, Eymard B, Isapof A, Strochlic L. New recessive mutations in SYT2 causing severe presynaptic congenital myasthenic syndromes. NEUROLOGY-GENETICS 2020; 6:e534. [PMID: 33659639 PMCID: PMC7803339 DOI: 10.1212/nxg.0000000000000534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/25/2020] [Indexed: 11/15/2022]
Abstract
Objective To report the identification of 2 new homozygous recessive mutations in the synaptotagmin 2 (SYT2) gene as the genetic cause of severe and early presynaptic forms of congenital myasthenic syndromes (CMSs). Methods Next-generation sequencing identified new homozygous intronic and frameshift mutations in the SYT2 gene as a likely cause of presynaptic CMS. We describe the clinical and electromyographic patient phenotypes, perform ex vivo splicing analyses to characterize the effect of the intronic mutation on exon splicing, and analyze the functional impact of this variation at the neuromuscular junction (NMJ). Results The 2 infants presented a similar clinical phenotype evoking first a congenital myopathy characterized by muscle weakness and hypotonia. Next-generation sequencing allowed to the identification of 1 homozygous intronic mutation c.465+1G>A in patient 1 and another homozygous frameshift mutation c.328_331dup in patient 2, located respectively in the 5' splice donor site of SYT2 intron 4 and in exon 3. Functional studies of the intronic mutation validated the abolition of the splice donor site of exon 4 leading to its skipping. In-frame skipping of exon 4 that encodes part of the C2A calcium-binding domain of SYT2 is associated with a loss-of-function effect resulting in a decrease of neurotransmitter release and severe pre- and postsynaptic NMJ defects. Conclusions This study identifies new homozygous recessive SYT2 mutations as the underlying cause of severe and early presynaptic form of CMS expanding the genetic spectrum of recessive SYT2-related CMS associated with defects in neurotransmitter release.
Collapse
Affiliation(s)
- Stéphanie Bauché
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Alain Sureau
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Damien Sternberg
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - John Rendu
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Céline Buon
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Julien Messéant
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Myriam Boëx
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Denis Furling
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Julien Fauré
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Xénia Latypova
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Antoinette Bernabe Gelot
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Michèle Mayer
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Pierre Mary
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Sandra Whalen
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Emmanuel Fournier
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Isabelle Cloix
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Ganaelle Remerand
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Fanny Laffargue
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Marie-Christine Nougues
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Bertrand Fontaine
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Bruno Eymard
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Arnaud Isapof
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| | - Laure Strochlic
- Sorbonne Université, INSERM, UMRS974, Centre de Recherche en Myologie, Hôpital de la Pitié-Salpêtrière, Paris, (S.B., A.S., C. B., J.M., M.B., D.F., E. F., B.F., B.E., A.I., L.S.); CHU APHP (D.S., J.R., J.F., X.L., A.B.G., M.M., P.M., S.W., E.F., I.C., G.R., F.L., M.C.N., B.F., B.E., A.I.); Aix-Marseille University, INSERM, INMED, Campus de Luminy, Marseille, France (A.B.G.); UFR Cardiogénétique et Myogénétique, Hôpital de la Pitié-Salpêtrière, APHP, Paris (D.S.); UF de génétique clinique, CRMR Anomalies du développement et syndromes malformatifs, APHP, Hôpital Armand Trousseau, Paris, France (S.W.); Université de Grenoble Alpes, INSERM, CHU Grenoble Alpes, GIN (J.R., J.F., X.L.); CHU Clermont Ferrand (I.C., G.R., F.L.); and Reference Centre for Neuromuscular Pathologies "Nord/Est/Ile-de France" Paris (A.B.G., M.M., P.M., S.W., M.C.N., B.F., B.E., A.I.)
| |
Collapse
|
170
|
Blasco A, Gras S, Mòdol-Caballero G, Tarabal O, Casanovas A, Piedrafita L, Barranco A, Das T, Pereira SL, Navarro X, Rueda R, Esquerda JE, Calderó J. Motoneuron deafferentation and gliosis occur in association with neuromuscular regressive changes during ageing in mice. J Cachexia Sarcopenia Muscle 2020; 11:1628-1660. [PMID: 32691534 PMCID: PMC7749545 DOI: 10.1002/jcsm.12599] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The cellular mechanisms underlying the age-associated loss of muscle mass and function (sarcopenia) are poorly understood, hampering the development of effective treatment strategies. Here, we performed a detailed characterization of age-related pathophysiological changes in the mouse neuromuscular system. METHODS Young, adult, middle-aged, and old (1, 4, 14, and 24-30 months old, respectively) C57BL/6J mice were used. Motor behavioural and electrophysiological tests and histological and immunocytochemical procedures were carried out to simultaneously analyse structural, molecular, and functional age-related changes in distinct cellular components of the neuromuscular system. RESULTS Ageing was not accompanied by a significant loss of spinal motoneurons (MNs), although a proportion (~15%) of them in old mice exhibited an abnormally dark appearance. Dark MNs were also observed in adult (~9%) and young (~4%) animals, suggesting that during ageing, some MNs undergo early deleterious changes, which may not lead to MN death. Old MNs were depleted of cholinergic and glutamatergic inputs (~40% and ~45%, respectively, P < 0.01), suggestive of age-associated alterations in MN excitability. Prominent microgliosis and astrogliosis [~93% (P < 0.001) and ~100% (P < 0.0001) increase vs. adults, respectively] were found in old spinal cords, with increased density of pro-inflammatory M1 microglia and A1 astroglia (25-fold and 4-fold increase, respectively, P < 0.0001). Ageing resulted in significant reductions in the nerve conduction velocity and the compound muscle action potential amplitude (~30%, P < 0.05, vs. adults) in old distal plantar muscles. Compared with adult muscles, old muscles exhibited significantly higher numbers of both denervated and polyinnervated neuromuscular junctions, changes in fibre type composition, higher proportion of fibres showing central nuclei and lipofuscin aggregates, depletion of satellite cells, and augmented expression of different molecules related to development, plasticity, and maintenance of neuromuscular junctions, including calcitonin gene-related peptide, growth associated protein 43, agrin, fibroblast growth factor binding protein 1, and transforming growth factor-β1. Overall, these alterations occurred at varying degrees in all the muscles analysed, with no correlation between the age-related changes observed and myofiber type composition or muscle topography. CONCLUSIONS Our data provide a global view of age-associated neuromuscular changes in a mouse model of ageing and help to advance understanding of contributing pathways leading to development of sarcopenia.
Collapse
Affiliation(s)
- Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Guillem Mòdol-Caballero
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, CIBERNED, Bellaterra, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, 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, 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, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | | | - Tapas Das
- Abbott Nutrition Research and Development, Columbus, OH, USA
| | | | - Xavier Navarro
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, CIBERNED, Bellaterra, Spain
| | - Ricardo Rueda
- Abbott Nutrition Research and Development, Granada, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, 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, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| |
Collapse
|
171
|
Wang W, Xu C, Ma X, Zhang X, Xie P. Intensive Care Unit-Acquired Weakness: A Review of Recent Progress With a Look Toward the Future. Front Med (Lausanne) 2020; 7:559789. [PMID: 33330523 PMCID: PMC7719824 DOI: 10.3389/fmed.2020.559789] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
Intensive care unit-acquired weakness (ICU-AW), a common neuromuscular complication associated with patients in the ICU, is a type of skeletal muscle dysfunction that commonly occurs following sepsis, mobility restriction, hyperglycemia, and the use of glucocorticoids or neuromuscular blocking agents. ICU-AW can lead to delayed withdrawal of mechanical ventilation and extended hospitalization. Patients often have poor prognosis, limited mobility, and severely affected quality of life. Currently, its pathogenesis is uncertain, with unavailability of specific drugs or targeted therapies. ICU-AW has gained attention in recent years. This manuscript reviews the current research status of the epidemiology, pathogenesis, diagnosis, and treatment methods for ICU-AW and speculates the novel perspectives for future research.
Collapse
Affiliation(s)
- Wenkang Wang
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| | - Chuanjie Xu
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| | - Xinglong Ma
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| | - Xiaoming Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine Houston, Houston, TX, United States
| | - Peng Xie
- Department of Critical Care Medicine of the Third Affiliated Hospital (The First People's Hospital of Zunyi), Zunyi Medical University, Zunyi, China
| |
Collapse
|
172
|
Castellanos-Montiel MJ, Velasco I, Escobedo-Avila I. Modeling the neuromuscular junction in vitro: an approach to study neuromuscular junction disorders. Ann N Y Acad Sci 2020; 1488:3-15. [PMID: 33040338 DOI: 10.1111/nyas.14504] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022]
Abstract
The neuromuscular junction (NMJ) is a specialized structure that works as an interface to translate the action potential of the presynaptic motor neuron (MN) in the contraction of the postsynaptic myofiber. The design of appropriate experimental models is essential to have efficient and reliable approaches to study NMJ development and function, but also to generate conditions that recapitulate distinct features of diseases. Initial studies relied on the use of tissue slices maintained under the same environment and in which single motor axons were difficult to trace. Later, MNs and muscle cells were obtained from primary cultures or differentiation of progenitors and cocultured as monolayers; however, the tissue architecture was lost. Current approaches include self-assembling 3D structures or the incorporation of biomaterials with cells to generate engineered tissues, although the incorporation of Schwann cells remains a challenge. Thus, numerous investigations have established different NMJ models, some of which are quite complex and challenging. Our review summarizes the in vitro models that have emerged in recent years to coculture MNs and skeletal muscle, trying to mimic the healthy and diseased NMJ. We expect our review may serve as a reference for choosing the appropriate experimental model for the required purposes of investigation.
Collapse
Affiliation(s)
- María José Castellanos-Montiel
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.,Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montréal, Quebec, Canada
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.,Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico City, Mexico
| | - Itzel Escobedo-Avila
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
173
|
Ng SY, Ljubicic V. Recent insights into neuromuscular junction biology in Duchenne muscular dystrophy: Impacts, challenges, and opportunities. EBioMedicine 2020; 61:103032. [PMID: 33039707 PMCID: PMC7648118 DOI: 10.1016/j.ebiom.2020.103032] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/28/2020] [Accepted: 09/11/2020] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common and relentless form of muscular dystrophy. The pleiotropic effects of dystrophin deficiency include remarkable impacts on neuromuscular junction (NMJ) structure and function. Some of these alterations contribute to the severe muscle wasting and weakness that distinguish DMD, while others attempt to compensate for them. Experimental approaches that correct NMJ biology in pre-clinical models of DMD attenuate disease progression and improve functional outcomes, which suggests that targeting the NMJ may be an effective therapeutic strategy for DMD patients. The objectives of this review are to 1) survey the distinctions in NMJ structure, function, and gene expression in the dystrophic context as compared to the healthy condition, and 2) summarize the efforts, opportunities and challenges to correct NMJ biology in DMD. This information will expand our basic understanding of neuromuscular biology and may be useful for designing novel NMJ-targeted drug or behavioural strategies to mitigate the dystrophic pathology and other disorders of the neuromuscular system.
Collapse
Affiliation(s)
- Sean Y Ng
- Department of Kinesiology, McMaster University, Hamilton L8S 4L8, Ontario, Canada
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton L8S 4L8, Ontario, Canada.
| |
Collapse
|
174
|
Cetin H, Beeson D, Vincent A, Webster R. The Structure, Function, and Physiology of the Fetal and Adult Acetylcholine Receptor in Muscle. Front Mol Neurosci 2020; 13:581097. [PMID: 33013323 PMCID: PMC7506097 DOI: 10.3389/fnmol.2020.581097] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/13/2020] [Indexed: 12/31/2022] Open
Abstract
The neuromuscular junction (NMJ) is a highly developed synapse linking motor neuron activity with muscle contraction. A complex of molecular cascades together with the specialized NMJ architecture ensures that each action potential arriving at the motor nerve terminal is translated into an action potential in the muscle fiber. The muscle-type nicotinic acetylcholine receptor (AChR) is a key molecular component located at the postsynaptic muscle membrane responsible for the generation of the endplate potential (EPP), which usually exceeds the threshold potential necessary to activate voltage-gated sodium channels and triggers a muscle action potential. Two AChR isoforms are found in mammalian muscle. The fetal isoform is present in prenatal stages and is involved in the development of the neuromuscular system whereas the adult isoform prevails thereafter, except after denervation when the fetal form is re-expressed throughout the muscle. This review will summarize the structural and functional differences between the two isoforms and outline congenital and autoimmune myasthenic syndromes that involve the isoform specific AChR subunits.
Collapse
Affiliation(s)
- Hakan Cetin
- Department of Neurology, Medical University of Vienna, Vienna, Austria.,Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Richard Webster
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
175
|
Castets P, Ham DJ, Rüegg MA. The TOR Pathway at the Neuromuscular Junction: More Than a Metabolic Player? Front Mol Neurosci 2020; 13:162. [PMID: 32982690 PMCID: PMC7485269 DOI: 10.3389/fnmol.2020.00162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/05/2020] [Indexed: 12/18/2022] Open
Abstract
The neuromuscular junction (NMJ) is the chemical synapse connecting motor neurons and skeletal muscle fibers. NMJs allow all voluntary movements, and ensure vital functions like breathing. Changes in the structure and function of NMJs are hallmarks of numerous pathological conditions that affect muscle function including sarcopenia, the age-related loss of muscle mass and function. However, the molecular mechanisms leading to the morphological and functional perturbations in the pre- and post-synaptic compartments of the NMJ remain poorly understood. Here, we discuss the role of the metabolic pathway associated to the kinase TOR (Target of Rapamycin) in the development, maintenance and alterations of the NMJ. This is of particular interest as the TOR pathway has been implicated in aging, but its role at the NMJ is still ill-defined. We highlight the respective functions of the two TOR-associated complexes, TORC1 and TORC2, and discuss the role of localized protein synthesis and autophagy regulation in motor neuron terminals and sub-synaptic regions of muscle fibers and their possible effects on NMJ maintenance.
Collapse
Affiliation(s)
- Perrine Castets
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | | |
Collapse
|
176
|
Rocha LC, Jacob CDS, Barbosa GK, Pimentel Neto J, Krause Neto W, Gama EF, Ciena AP. Remodeling of the skeletal muscle and postsynaptic component after short-term joint immobilization and aquatic training. Histochem Cell Biol 2020; 154:621-628. [PMID: 32797254 DOI: 10.1007/s00418-020-01910-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2020] [Indexed: 12/16/2022]
Abstract
Joint immobilization is commonly used as a conservative treatment for osteoarticular and musculotendinous traumas. However, joint immobilization might elicit degenerative effects on the neuromuscular system and muscle atrophy. For this reason, the choice of strategies that mitigate these effects is essential in the post-immobilization period. Therefore, this study aimed to investigate the impact of aquatic training on the morphology of muscle fibers and motor endplates of the gastrocnemius muscle in the post-immobilization period. Male Wistar rats (90 days old) were divided into groups: Sedentary: no procedure; Immobilization: joint immobilization protocol (10 days); Immobilization/non-training: joint immobilization protocol (10 days) followed by four weeks without exercise intervention; Immobilization/training: joint immobilization protocol (10 days) and post-immobilization aquatic training (4 weeks). After the procedures, we quantified the cross-sectional area (CSA), volume and numerical density of different myofibers types, and total and stained area and perimeter of the motor endplate. We demonstrate the following main results: (a) short-term joint immobilization resulted in myofibers atrophy; however, we verified a small change in the postsynaptic component; (b) the period of inactivity after immobilization caused severe changes in the motor endplate (lower stained area, stained perimeter, total area, and total perimeter) and maintenance of muscle atrophy due to immobilization; (c) the prescription of post-immobilization exercise proved to be effective in restoring muscle morphology and inducing plasticity in the motor endplate. We conclude that short-term joint immobilization (10 days) results in atrophy type I and II myofibers, in addition to a decline in the total perimeter of the motor endplate. Besides, the post-immobilization period appears to be decisive in muscle and postsynaptic remodeling. Thus, aquatic training is effective in stimulating adjustments associated with muscle hypertrophy and plasticity of the motor endplate during the post-immobilization period.
Collapse
Affiliation(s)
- Lara Caetano Rocha
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), Avenue 24A, n 1515, Rio Claro, SP, 13506-900, Brazil
| | - Carolina Dos Santos Jacob
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), Avenue 24A, n 1515, Rio Claro, SP, 13506-900, Brazil
| | - Gabriela Klein Barbosa
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), Avenue 24A, n 1515, Rio Claro, SP, 13506-900, Brazil
| | - Jurandyr Pimentel Neto
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), Avenue 24A, n 1515, Rio Claro, SP, 13506-900, Brazil
| | - Walter Krause Neto
- Laboratory of Morphoquantitative Studies and Immunohistochemistry, Department of Physical Education, São Judas Tadeu University, São Paulo, SP, Brazil
| | - Eliane Florencio Gama
- Laboratory of Morphoquantitative Studies and Immunohistochemistry, Department of Physical Education, São Judas Tadeu University, São Paulo, SP, Brazil
| | - Adriano Polican Ciena
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), Avenue 24A, n 1515, Rio Claro, SP, 13506-900, Brazil.
| |
Collapse
|
177
|
DOK7 Gene Therapy Enhances Neuromuscular Junction Innervation and Motor Function in Aged Mice. iScience 2020; 23:101385. [PMID: 32758427 PMCID: PMC7452162 DOI: 10.1016/j.isci.2020.101385] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/21/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Muscle denervation at the neuromuscular junction (NMJ), the essential synapse between motor neuron and skeletal muscle, is associated with age-related motor impairment. Therefore, improving muscle innervation at aged NMJs may be an effective therapeutic strategy for treating the impairment. We previously demonstrated that the muscle protein Dok-7 plays an essential role in NMJ formation, and, indeed, its forced expression in muscle enlarges NMJs. Moreover, therapeutic administration of an adeno-associated virus vector encoding human Dok-7 (DOK7 gene therapy) suppressed muscle denervation and enhanced motor activity in a mouse model of amyotrophic lateral sclerosis (ALS). Here, we show that DOK7 gene therapy significantly enhances motor function and muscle strength together with NMJ innervation in aged mice. Furthermore, the treated mice showed greatly increased compound muscle action potential (CMAP) amplitudes compared with the controls, suggesting enhanced neuromuscular transmission. Thus, therapies aimed at enhancing NMJ innervation have potential for treating age-related motor impairment. DOK7 gene therapy enhances motor function and muscle strength in aged (≥2 years) mice DOK7 gene therapy enhances neuromuscular junction (NMJ) innervation in aged mice DOK7 gene therapy increases compound muscle action potential amplitudes in aged mice Enhancing NMJ innervation in the elderly may strengthen muscles and motor activities
Collapse
|
178
|
Lahiri S, Kim H, Garcia-Perez I, Reza MM, Martin KA, Kundu P, Cox LM, Selkrig J, Posma JM, Zhang H, Padmanabhan P, Moret C, Gulyás B, Blaser MJ, Auwerx J, Holmes E, Nicholson J, Wahli W, Pettersson S. The gut microbiota influences skeletal muscle mass and function in mice. Sci Transl Med 2020; 11:11/502/eaan5662. [PMID: 31341063 DOI: 10.1126/scitranslmed.aan5662] [Citation(s) in RCA: 356] [Impact Index Per Article: 71.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/30/2018] [Accepted: 02/11/2019] [Indexed: 12/25/2022]
Abstract
The functional interactions between the gut microbiota and the host are important for host physiology, homeostasis, and sustained health. We compared the skeletal muscle of germ-free mice that lacked a gut microbiota to the skeletal muscle of pathogen-free mice that had a gut microbiota. Compared to pathogen-free mouse skeletal muscle, germ-free mouse skeletal muscle showed atrophy, decreased expression of insulin-like growth factor 1, and reduced transcription of genes associated with skeletal muscle growth and mitochondrial function. Nuclear magnetic resonance spectrometry analysis of skeletal muscle, liver, and serum from germ-free mice revealed multiple changes in the amounts of amino acids, including glycine and alanine, compared to pathogen-free mice. Germ-free mice also showed reduced serum choline, the precursor of acetylcholine, the key neurotransmitter that signals between muscle and nerve at neuromuscular junctions. Reduced expression of genes encoding Rapsyn and Lrp4, two proteins important for neuromuscular junction assembly and function, was also observed in skeletal muscle from germ-free mice compared to pathogen-free mice. Transplanting the gut microbiota from pathogen-free mice into germ-free mice resulted in an increase in skeletal muscle mass, a reduction in muscle atrophy markers, improved oxidative metabolic capacity of the muscle, and elevated expression of the neuromuscular junction assembly genes Rapsyn and Lrp4 Treating germ-free mice with short-chain fatty acids (microbial metabolites) partly reversed skeletal muscle impairments. Our results suggest a role for the gut microbiota in regulating skeletal muscle mass and function in mice.
Collapse
Affiliation(s)
- Shawon Lahiri
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden. .,Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Hyejin Kim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Isabel Garcia-Perez
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Sir Alexander Fleming Building, Imperial College London, London SW72AZ, UK
| | - Musarrat Maisha Reza
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Katherine A Martin
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Parag Kundu
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Singapore Center for Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore, Singapore
| | - Laura M Cox
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joel Selkrig
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Joram M Posma
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Sir Alexander Fleming Building, Imperial College London, London SW72AZ, UK
| | - Hongbo Zhang
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | - Catherine Moret
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Balázs Gulyás
- Department of Neuroscience and Mental Health, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin J Blaser
- Department of Medicine, New York University School of Medicine, New York, NY 10016, USA.,Medical Service, VA New York Harbor Healthcare System, New York, NY 10010, USA
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Elaine Holmes
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Sir Alexander Fleming Building, Imperial College London, London SW72AZ, UK
| | - Jeremy Nicholson
- Australian National Phenome Center, Murdoch University, WA 6150, Australia
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,INRA ToxAlim Integrative Toxicology and Metabolism UMR1331, Chemin de Tournefeuille, Toulouse Cedex, France
| | - Sven Pettersson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden. .,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Singapore Center for Environmental Life Sciences Engineering (SCELSE), Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
179
|
Nango H, Kosuge Y, Sato M, Shibukawa Y, Aono Y, Saigusa T, Ito Y, Ishige K. Highly Efficient Conversion of Motor Neuron-Like NSC-34 Cells into Functional Motor Neurons by Prostaglandin E 2. Cells 2020; 9:cells9071741. [PMID: 32708195 PMCID: PMC7409148 DOI: 10.3390/cells9071741] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/10/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022] Open
Abstract
Motor neuron diseases are a group of progressive neurological disorders that degenerate motor neurons. The neuroblastoma × spinal cord hybrid cell line NSC-34 is widely used as an experimental model in studies of motor neuron diseases. However, the differentiation efficiency of NSC-34 cells to neurons is not always sufficient. We have found that prostaglandin E2 (PGE2) induces morphological differentiation in NSC-34 cells. The present study investigated the functional properties of PGE2-differentiated NSC-34 cells. Retinoic acid (RA), a widely-used agent inducing cell differentiation, facilitated neuritogenesis, which peaked on day 7, whereas PGE2-induced neuritogenesis took only 2 days to reach the same level. Whole-cell patch-clamp recordings showed that the current threshold of PGE2-treated cell action potentials was lower than that of RA-treated cells. PGE2 and RA increased the protein expression levels of neuronal differentiation markers, microtubule-associated protein 2c and synaptophysin, and to the same extent, motor neuron-specific markers HB9 and Islet-1. On the other hand, protein levels of choline acetyltransferase and basal release of acetylcholine in PGE2-treated cells were higher than in RA-treated cells. These results suggest that PGE2 is a rapid and efficient differentiation-inducing factor for the preparation of functionally mature motor neurons from NSC-34 cells.
Collapse
Affiliation(s)
- Hiroshi Nango
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba 274-8555, Japan; (H.N.); (Y.I.)
| | - Yasuhiro Kosuge
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba 274-8555, Japan; (H.N.); (Y.I.)
- Correspondence: (Y.K.); (K.I.); Tel.: +81-47-465-4027 (Y.K.)
| | - Masaki Sato
- Department of Physiology, Tokyo Dental College, 2-9-18 Kanda-Misakicho, Chiyoda-ku, Tokyo 101-0061, Japan; (M.S.); (Y.S.)
- Department of Biology Tokyo Dental College, 2-9-7 Kandasurugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Yoshiyuki Shibukawa
- Department of Physiology, Tokyo Dental College, 2-9-18 Kanda-Misakicho, Chiyoda-ku, Tokyo 101-0061, Japan; (M.S.); (Y.S.)
| | - Yuri Aono
- Department of Pharmacology, School of Dentistry at Matsudo, Nihon University, 2-870-1 Sakaechonishi, Matsudo-shi, Chiba 271-8587, Japan; (Y.A.); (T.S.)
| | - Tadashi Saigusa
- Department of Pharmacology, School of Dentistry at Matsudo, Nihon University, 2-870-1 Sakaechonishi, Matsudo-shi, Chiba 271-8587, Japan; (Y.A.); (T.S.)
| | - Yoshihisa Ito
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba 274-8555, Japan; (H.N.); (Y.I.)
- Pharmacy Education Center, Yokohama University of Pharmacy, 601 Matanocho, Totuka-ku, Yokohama 245-0066, Japan
| | - Kumiko Ishige
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba 274-8555, Japan; (H.N.); (Y.I.)
- Correspondence: (Y.K.); (K.I.); Tel.: +81-47-465-4027 (Y.K.)
| |
Collapse
|
180
|
Chen A, Bai L, Zhong K, Shu X, Wang A, Xiao Y, Zhang K, Shen C. APC2
CDH1
negatively regulates agrin signaling by promoting the ubiquitination and proteolytic degradation of DOK7. FASEB J 2020; 34:12009-12023. [PMID: 32687671 DOI: 10.1096/fj.202000485r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 01/28/2023]
Affiliation(s)
- Aizhong Chen
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Lei Bai
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Keke Zhong
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Xiaoqiu Shu
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Ailian Wang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Yatao Xiao
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Kejing Zhang
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
| | - Chengyong Shen
- The First Affiliated Hospital Institute of Translational Medicine School of Medicine Zhejiang University Zhejiang China
- Department of Neurobiology Key Laboratory of Medical Neurobiology of Zhejiang Province School of Medicine Zhejiang University Zhejiang China
| |
Collapse
|
181
|
Henze H, Jung MJ, Ahrens HE, Steiner S, von Maltzahn J. Skeletal muscle aging – Stem cells in the spotlight. Mech Ageing Dev 2020; 189:111283. [DOI: 10.1016/j.mad.2020.111283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/04/2020] [Accepted: 06/01/2020] [Indexed: 01/09/2023]
|
182
|
Xing G, Xiong WC, Mei L. Rapsyn as a signaling and scaffolding molecule in neuromuscular junction formation and maintenance. Neurosci Lett 2020; 731:135013. [DOI: 10.1016/j.neulet.2020.135013] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/23/2020] [Indexed: 12/20/2022]
|
183
|
Belotti E, Schaeffer L. Regulation of Gene expression at the neuromuscular Junction. Neurosci Lett 2020; 735:135163. [PMID: 32553805 DOI: 10.1016/j.neulet.2020.135163] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 06/11/2020] [Accepted: 06/14/2020] [Indexed: 01/08/2023]
Abstract
Gene expression in skeletal muscle is profoundly changed upon innervation. 50 years of research on the neuromuscular system have greatly increased our understanding of the mechanisms underlying these changes. By controlling the expression and the activity of key transcription factors, nerve-evoked electrical activity in the muscle fiber positively and negatively regulates the expression of hundreds of genes. Innervation also compartmentalizes gene expression into synaptic and extra-synaptic regions of muscle fibers. In addition, electrically-evoked, release of several factors (e.g. Agrin, Neuregulin, Wnt ligands) induce the clustering of synaptic proteins and of a few muscle nuclei. The sub-synaptic nuclei acquire a particular chromatin organization and develop a specific gene expression program dedicated to building and maintaining a functional neuromuscular synapse. Deciphering synapse-specific, transcriptional regulation started with the identification of the N-box, a six base pair element present in the promoters of the acetylcholine δ and ε subunits. Most genes with synapse-specific expression turned out to contain at least one N-box in their promoters. The N-box is a response element for the synaptic signals Agrin and Neuregulins as well as a binding site for transcription factors of the Ets family. The Ets transcription factors GABP and Erm are implicated in the activation of post-synaptic genes via the N-box. In muscle fibers, Erm expression is restricted to the NMJ whereas GABP is expressed in all muscle nuclei but phosphorylated and activated by the JNK and ERK signaling pathways in response to Agrin and Neuregulins. Post-synaptic gene expression also correlates with chromatin modifications at the genomic level as evidenced by the strong enrichment of decondensed chromatin and acetylated histones in sub-synaptic nuclei. Here we discuss these transcriptional pathways for synaptic specialization at NMJs.
Collapse
Affiliation(s)
- Edwige Belotti
- INMG, Inserm U1217, CNRS UMR5310, Université Lyon 1, Université De Lyon, Lyon, France
| | - Laurent Schaeffer
- INMG, Inserm U1217, CNRS UMR5310, Université Lyon 1, Université De Lyon, Lyon, France; Centre De Biotechnologie Cellulaire, Hospices Civils De Lyon, Lyon, France.
| |
Collapse
|
184
|
Chen PJ, Zelada D, Belhasan DC, Akaaboune M. Phosphorylation of α-dystrobrevin is essential for αkap accumulation and acetylcholine receptor stability. J Biol Chem 2020; 295:10677-10688. [PMID: 32532815 DOI: 10.1074/jbc.ra120.013952] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/08/2020] [Indexed: 01/26/2023] Open
Abstract
The maintenance of a high density of the acetylcholine receptor (AChR) is the hallmark of the neuromuscular junction. Muscle-specific anchoring protein (αkap) encoded within the calcium/calmodulin-dependent protein kinase IIα (CAMK2A) gene is essential for the maintenance of AChR clusters both in vivo and in cultured muscle cells. The underlying mechanism by which αkap is maintained and regulated remains unknown. Here, using human cell lines, fluorescence microscopy, and pulldown and immunoblotting assays, we show that α-dystrobrevin (α-dbn), an intracellular component of the dystrophin glycoprotein complex, directly and robustly promotes the stability of αkap in a concentration-dependent manner. Mechanistically, we found that the phosphorylatable tyrosine residues of α-dbn are essential for the stability of α-dbn itself and its interaction with αkap, with substitution of three tyrosine residues in the α-dbn C terminus with phenylalanine compromising the αkap-α-dbn interaction and significantly reducing both αkap and α-dbn accumulation. Moreover, the αkap-α-dbn interaction was critical for αkap accumulation and stability. We also found that the absence of either αkap or α-dbn markedly reduces AChRα accumulation and that overexpression of α-dbn or αkap in cultured muscle cells promotes the formation of large agrin-induced AChR clusters. Collectively, these results indicate that the stability of αkap and α-dbn complex plays an important role in the maintenance of high-level expression of AChRs.
Collapse
Affiliation(s)
- Po-Ju Chen
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Diego Zelada
- Department of Cell Biology, Universidad de Concepción, Concepción, Chile
| | - Dina Cheryne Belhasan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Mohammed Akaaboune
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA .,Program in Neuroscience, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
185
|
Prieto-Gómez V, Navarro-Brazález B, Sánchez-Méndez Ó, de-la-Villa P, Sánchez-Sánchez B, Torres-Lacomba M. Electromyographic Analysis of Shoulder Neuromuscular Activity in Women Following Breast Cancer Treatment: A Cross-Sectional Descriptive Study. J Clin Med 2020; 9:jcm9061804. [PMID: 32531893 PMCID: PMC7355794 DOI: 10.3390/jcm9061804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/30/2020] [Accepted: 06/08/2020] [Indexed: 11/19/2022] Open
Abstract
Certain secondary effects derived from medical treatment in breast cancer can favor the development of alterations in shoulder biomechanics. To the best of our knowledge, persistent peripheral pain as a key factor for the development of neuromuscular activity impairments has not been analyzed. A cross-sectional descriptive study was carried out. A total of 90 women were included and allocated to three groups: (i) 30 women with persistent peripheral pain after breast cancer treatment, (ii) 30 women without pain after breast cancer treatment, and (iii) 30 healthy women. Surface electromyography was employed to measure the onset and amplitude of the muscle activity of three shoulder movements. Statistically significant differences were found in the neuromuscular activity for all the muscles and shoulder movements among women with persistent pain versus healthy women (i.e., amplitude muscle activity variable p < 0.001). Statistically significant differences were also observed in the neuromuscular activity for certain muscles in shoulder movements among women with persistent pain versus women without pain, as well as between women without pain versus healthy women. Therefore, following breast cancer treatment, women showed alterations in their shoulder neuromuscular activity, which were more significant if persistent pain existed. These findings may contribute to developing a selective therapeutic exercise program that optimizes the shoulder neuromuscular activity in women after breast cancer treatment.
Collapse
|
186
|
Behera J, Ison J, Tyagi SC, Tyagi N. The role of gut microbiota in bone homeostasis. Bone 2020; 135:115317. [PMID: 32169602 PMCID: PMC8457311 DOI: 10.1016/j.bone.2020.115317] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/27/2020] [Accepted: 03/10/2020] [Indexed: 02/08/2023]
Abstract
The gut microbiota (GM) is referred to as the second gene pool of the human body and a commensal, symbiotic, and pathogenic microorganism living in our intestines. The knowledge of the complex interaction between intestinal microbiota and health outcomes is a novel and rapidly expanding the field. Earlier studies have reported that the microbial communities affect the cellular responses and shape many aspects of physiology and pathophysiology within the body, including muscle and bone metabolism (formation and resorption). GM influences the skeletal homeostasis via affecting the host metabolism, immune function, hormone secretion, and the gut-brain axis. The premise of this review is to discuss the role of GM on bone homeostasis and skeletal muscle mass function. This review also opens up new perspectives for pathophysiological studies by establishing the presence of a 'microbiota-skeletal' axis and raising the possibility of innovative new treatments for skeletal development.
Collapse
Affiliation(s)
- Jyotirmaya Behera
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Jessica Ison
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Suresh C Tyagi
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Neetu Tyagi
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
187
|
Reduced TUBA1A Tubulin Causes Defects in Trafficking and Impaired Adult Motor Behavior. eNeuro 2020; 7:ENEURO.0045-20.2020. [PMID: 32184299 PMCID: PMC7218002 DOI: 10.1523/eneuro.0045-20.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/03/2020] [Indexed: 12/30/2022] Open
Abstract
Newly born neurons express high levels of TUBA1A α-tubulin to assemble microtubules for neurite extension and to provide tracks for intracellular transport. In the adult brain, Tuba1a expression decreases dramatically. A mouse that harbors a loss-of-function mutation in the gene encoding TUBA1A (Tuba1aND/+) allows us to ask whether TUBA1A is important for the function of mature neurons. α-Tubulin levels are about half of wild type in juvenile Tuba1aND/+ brains, but are close to normal in older animals. In postnatal day (P)0 cultured neurons, reduced TUBA1A allows for assembly of less microtubules in axons resulting in more pausing during organelle trafficking. While Tuba1aND/+ mouse behavior is indistinguishable from wild-type siblings at weaning, Tuba1aND/+ mice develop adult-onset ataxia. Neurons important for motor function in Tuba1aND/+ remain indistinguishable from wild-type with respect to morphology and number and display no evidence of axon degeneration. Tuba1aND/+ neuromuscular junction (NMJ) synapses are the same size as wild-type before the onset of ataxia, but are reduced in size in older animals. Together, these data indicate that the TUBA1A-rich microtubule tracks that are assembled during development are essential for mature neuron function and maintenance of synapses over time.
Collapse
|
188
|
Scaricamazza S, Salvatori I, Giacovazzo G, Loeffler JP, Renè F, Rosina M, Quessada C, Proietti D, Heil C, Rossi S, Battistini S, Giannini F, Volpi N, Steyn FJ, Ngo ST, Ferraro E, Madaro L, Coccurello R, Valle C, Ferri A. Skeletal-Muscle Metabolic Reprogramming in ALS-SOD1 G93A Mice Predates Disease Onset and Is A Promising Therapeutic Target. iScience 2020; 23:101087. [PMID: 32371370 PMCID: PMC7200935 DOI: 10.1016/j.isci.2020.101087] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/13/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
Patients with ALS show, in addition to the loss of motor neurons in the spinal cord, brainstem, and cerebral cortex, an abnormal depletion of energy stores alongside hypermetabolism. In this study, we show that bioenergetic defects and muscle remodeling occur in skeletal muscle of the SOD1G93A mouse model of ALS mice prior to disease onset and before the activation of muscle denervation markers, respectively. These changes in muscle physiology were followed by an increase in energy expenditure unrelated to physical activity. Finally, chronic treatment of SOD1G93A mice with Ranolazine, an FDA-approved inhibitor of fatty acid β-oxidation, led to a decrease in energy expenditure in symptomatic SOD1G93A mice, and this occurred in parallel with a robust, albeit temporary, recovery of the pathological phenotype. Metabolic switch use occurs early in the skeletal muscle of SOD1G93A mice Mitochondrial impairment precedes locomotor deficits and evokes catabolic pathways Sarcolipin upregulation in presymptomatic SOD1G93A mice precedes hypermetabolism Pharmacological modulation of hypermetabolism improves locomotor performance
Collapse
Affiliation(s)
- Silvia Scaricamazza
- University of Rome Tor Vergata, Department of Biology, Rome, Italy; IRCCS Fondazione Santa Lucia, Rome, Italy
| | | | | | - Jean Philippe Loeffler
- Université de Strasbourg, UMR_S 1118, Strasbourg, France; INSERM, U1118, Central and Peripheral Mechanisms of Neurodegeneration, Strasbourg, France
| | - Frederique Renè
- Université de Strasbourg, UMR_S 1118, Strasbourg, France; INSERM, U1118, Central and Peripheral Mechanisms of Neurodegeneration, Strasbourg, France
| | - Marco Rosina
- University of Rome Tor Vergata, Department of Biology, Rome, Italy
| | - Cyril Quessada
- Université de Strasbourg, UMR_S 1118, Strasbourg, France; INSERM, U1118, Central and Peripheral Mechanisms of Neurodegeneration, Strasbourg, France
| | | | | | - Simona Rossi
- University of Rome Tor Vergata, Department of Biology, Rome, Italy; National Research Council, Institute of Translational Pharmacology (IFT), Rome, Italy
| | - Stefania Battistini
- University of Siena, Department of Medical, Surgical and Neurological Science, Siena, Italy
| | - Fabio Giannini
- University of Siena, Department of Medical, Surgical and Neurological Science, Siena, Italy
| | - Nila Volpi
- University of Siena, Department of Medical, Surgical and Neurological Science, Siena, Italy
| | - Frederik J Steyn
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia; Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Shyuan T Ngo
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia; Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | | | - Luca Madaro
- IRCCS Fondazione Santa Lucia, Rome, Italy; DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome, Italy
| | - Roberto Coccurello
- IRCCS Fondazione Santa Lucia, Rome, Italy; National Research Council, Institute for Complex System (ISC), Rome, Italy
| | - Cristiana Valle
- IRCCS Fondazione Santa Lucia, Rome, Italy; National Research Council, Institute of Translational Pharmacology (IFT), Rome, Italy.
| | - Alberto Ferri
- IRCCS Fondazione Santa Lucia, Rome, Italy; National Research Council, Institute of Translational Pharmacology (IFT), Rome, Italy.
| |
Collapse
|
189
|
Busetto G, Cangiano A. Pre- and post-synaptic roles of action potential activity in synapse elimination revealed by using ectopic neuromuscular junction formation by a foreign nerve. Neurosci Lett 2020; 722:134835. [PMID: 32057925 DOI: 10.1016/j.neulet.2020.134835] [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: 12/13/2019] [Revised: 02/07/2020] [Accepted: 02/09/2020] [Indexed: 10/25/2022]
Abstract
The formation of the neuromuscular junction (nmj) is based on molecular cascades initiated by neural agrin as well as electrical activity in the neuromuscular structures. This review focuses on the latter factor, emphasizing the multiplicity of its mechanisms in the process of synapse elimination following initial polyneuronal innervation. Pre- and post-synaptic components of activity have in fact been identified through experiments on an adult model of nmj formation: ectopic reinnervation of the rat soleus muscle by the fibular nerve. Two activity-dependent elimination processes are thus compared: competition between distributed nmjs, which depends on evoked muscle impulse activity, and competition between axons converging on single nmjs, which instead depends on differences in the timing of impulses in the converging axons.
Collapse
Affiliation(s)
- Giuseppe Busetto
- Department of Neurosciences Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy; National Institute of Neuroscience, Verona, Italy
| | - Alberto Cangiano
- Department of Neurosciences Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134, Verona, Italy; National Institute of Neuroscience, Verona, Italy.
| |
Collapse
|
190
|
Pęziński M, Daszczuk P, Pradhan BS, Lochmüller H, Prószyński TJ. An improved method for culturing myotubes on laminins for the robust clustering of postsynaptic machinery. Sci Rep 2020; 10:4524. [PMID: 32161296 PMCID: PMC7066178 DOI: 10.1038/s41598-020-61347-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 02/20/2020] [Indexed: 01/03/2023] Open
Abstract
Motor neurons form specialized synapses with skeletal muscle fibers, called neuromuscular junctions (NMJs). Cultured myotubes are used as a simplified in vitro system to study the postsynaptic specialization of muscles. The stimulation of myotubes with the glycoprotein agrin or laminin-111 induces the clustering of postsynaptic machinery that contains acetylcholine receptors (AChRs). When myotubes are grown on laminin-coated surfaces, AChR clusters undergo developmental remodeling to form topologically complex structures that resemble mature NMJs. Needing further exploration are the molecular processes that govern AChR cluster assembly and its developmental maturation. Here, we describe an improved protocol for culturing muscle cells to promote the formation of complex AChR clusters. We screened various laminin isoforms and showed that laminin-221 was the most potent for inducing AChR clusters, whereas laminin-121, laminin-211, and laminin-221 afforded the highest percentages of topologically complex assemblies. Human primary myotubes that were formed by myoblasts obtained from patient biopsies also assembled AChR clusters that underwent remodeling in vitro. Collectively, these results demonstrate an advancement of culturing myotubes that can facilitate high-throughput screening for potential therapeutic targets for neuromuscular disorders.
Collapse
Affiliation(s)
- Marcin Pęziński
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Patrycja Daszczuk
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Bhola Shankar Pradhan
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.,Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Hanns Lochmüller
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Centre, University of Freiburg, Freiburg, Germany.,Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Tomasz J Prószyński
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland. .,Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland.
| |
Collapse
|
191
|
Perisynaptic schwann cells - The multitasking cells at the developing neuromuscular junctions. Semin Cell Dev Biol 2020; 104:31-38. [PMID: 32147379 DOI: 10.1016/j.semcdb.2020.02.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/27/2022]
Abstract
Neuromuscular junctions (NMJs) are specialized synapses in the peripheral nervous system that allow the transmission of neuronal impulses to skeletal muscles for their contraction. Due to its size and accessibility, the NMJ is a commonly used model for studying basic principles of synapse organization and function. Similar to synapses in the central nervous system, NMJs are composed of presynaptic axonal terminals, the postsynaptic machinery formed at the membrane of the muscle fibers, and the synapse-associated glial cells. The special glial cells at the NMJs are called terminal Schwann cells or perisynaptic Schwann cells (PSCs). Decades of studies on the NMJ, as well as the most recent discoveries, have revealed multiple functions for PSCs at different stages of synaptic formation, maintenance, and disassembly. This review summarizes major observations in the field.
Collapse
|
192
|
The role of the dystrophin glycoprotein complex on the neuromuscular system. Neurosci Lett 2020; 722:134833. [DOI: 10.1016/j.neulet.2020.134833] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/07/2020] [Accepted: 02/09/2020] [Indexed: 12/26/2022]
|
193
|
Qaisar R, Karim A, Elmoselhi AB. Muscle unloading: A comparison between spaceflight and ground-based models. Acta Physiol (Oxf) 2020; 228:e13431. [PMID: 31840423 DOI: 10.1111/apha.13431] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 12/09/2019] [Accepted: 12/12/2019] [Indexed: 12/12/2022]
Abstract
Prolonged unloading of skeletal muscle, a common outcome of events such as spaceflight, bed rest and hindlimb unloading, can result in extensive metabolic, structural and functional changes in muscle fibres. With advancement in investigations of cellular and molecular mechanisms, understanding of disuse muscle atrophy has significantly increased. However, substantial gaps exist in our understanding of the processes dictating muscle plasticity during unloading, which prevent us from developing effective interventions to combat muscle loss. This review aims to update the status of knowledge and underlying mechanisms leading to cellular and molecular changes in skeletal muscle during unloading. We have also discussed advances in the understanding of contractile dysfunction during spaceflights and in ground-based models of muscle unloading. Additionally, we have elaborated on potential therapeutic interventions that show promising results in boosting muscle mass and strength during mechanical unloading. Finally, we have identified key gaps in our knowledge as well as possible research direction for the future.
Collapse
Affiliation(s)
- Rizwan Qaisar
- Department of Basic Medical Sciences College of Medicine University of Sharjah Sharjah UAE
| | - Asima Karim
- Department of Basic Medical Sciences College of Medicine University of Sharjah Sharjah UAE
| | - Adel B. Elmoselhi
- Department of Basic Medical Sciences College of Medicine University of Sharjah Sharjah UAE
- Department of Physiology Michigan State University East Lansing MI USA
| |
Collapse
|
194
|
Estrada‐Bonilla YC, Castro de Souza‐Tomé PA, Faturi FM, Mendes‐Zambetta R, Lepesteur‐Gianlorenço AC, Croti G, Jones TA, Russo TL. Compensatory neuromuscular junction adaptations of forelimb muscles in focal cortical ischemia in rats. Brain Behav 2020; 10:e01472. [PMID: 32004425 PMCID: PMC7066338 DOI: 10.1002/brb3.1472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/17/2019] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION Upper limb movements are affected frequently by brain ischemia (BI). Mechanisms involved in recovery and compensatory movements have developed several studies. However, less attention is given to skeletal muscles, where neuromuscular junction (NMJ) has an important role on muscle tropism and functional performance. METHODS Animals were divided into two groups: control (C) and BI. Then, animals were skilled to perform single-pellet retrieval task, following these procedures: habituation, shaping, and single-pellet retrieval task. BI was induced using stereotaxic surgery in order to apply endothelin-1 in motor cortex, representative of movements of dominant paw. Reaching task performance was evaluated by single-pellet retrieval task 1 day before BI induction, 4 and 15 days after BI induction. After that, biceps, triceps, fingers flexor, and extensor muscles were extracted. NMJ was assessed in morphometric characteristics (total area, total perimeter, and feret). Muscle fiber cross-sectional area and connective tissue percentage were also evaluated for characterization. Student's t test was used for comparisons between C and BI groups. Tau Kendall's correlation was applied among variables from BI group. RESULTS An increase in all NMJ morphometric parameters, as well as increase of atrophy and fibrosis in BI group compared with C. There was a high level of direct correlation between mean values of NMJ morphometry with percentage of success in reaching task in BI group. CONCLUSION Brain ischemia-induced NMJ compensatory expansion, muscle atrophy, and fibrosis in forelimb muscles that are related to reaching performance.
Collapse
Affiliation(s)
- Yisel Carolina Estrada‐Bonilla
- Physiotherapy Deparment – (DFisio)Laboratorio de Pesquisa em Fisioterapia Neurológica – LaFiNFederal University of São CarlosSão CarlosBrazil
- Body, Subjetct and Education Research GroupPhysical Culture, Sports and RecreationSaint Thomas UniversityBogotáDCColombia
| | - Paula Aiello Castro de Souza‐Tomé
- Physiotherapy Deparment – (DFisio)Laboratorio de Pesquisa em Fisioterapia Neurológica – LaFiNFederal University of São CarlosSão CarlosBrazil
| | - Fernanda María Faturi
- Physiotherapy Deparment – (DFisio)Laboratorio de Pesquisa em Fisioterapia Neurológica – LaFiNFederal University of São CarlosSão CarlosBrazil
| | - Rafaella Mendes‐Zambetta
- Physiotherapy Deparment – (DFisio)Laboratorio de Pesquisa em Fisioterapia Neurológica – LaFiNFederal University of São CarlosSão CarlosBrazil
| | - Anna Carolyna Lepesteur‐Gianlorenço
- Physiotherapy Deparment – (DFisio)Laboratorio de Pesquisa em Fisioterapia Neurológica – LaFiNFederal University of São CarlosSão CarlosBrazil
| | - Gabrielle Croti
- Physiotherapy Deparment – (DFisio)Laboratorio de Pesquisa em Fisioterapia Neurológica – LaFiNFederal University of São CarlosSão CarlosBrazil
| | - Theresa A. Jones
- Department of PsychologyInstitute for NeuroscienceUniversity of Texas at AustinAustinTXUSA
| | - Thiago Luiz Russo
- Physiotherapy Deparment – (DFisio)Laboratorio de Pesquisa em Fisioterapia Neurológica – LaFiNFederal University of São CarlosSão CarlosBrazil
| |
Collapse
|
195
|
Arakawa M, Wagatsuma A. 1α, 25(OH) 2D 3 regulates agrin-induced acetylcholine receptor clustering through upregulation of rapsyn expression in C2C12 myotubes. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30293-X. [PMID: 32081417 DOI: 10.1016/j.bbrc.2020.02.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 12/26/2022]
Abstract
The active form of vitamin D (1α, 25-dihydroxyvitamin D3 [1α, 25(OH)2D3], referred to as 1,25D) has been suggested to play a pivotal role in skeletal muscle function and metabolism. However, the mechanisms through which 1,25D functions in this tissue remain to be elucidated. Recent studies have shown that vitamin D signaling regulates neuromuscular maintenance and improves locomotion in mice. In the present study, we examined the effects of 1,25D on neuromuscular synaptogenesis by measuring clustering of acetylcholine receptors (AChRs) in C2C12 myotubes. 1,25D treatment enhanced the agrin-induced AChR clustering in myotubes compared to treatment with agrin alone. Furthermore, siRNA-mediated knockdown of the vitamin D receptor (VDR) decreased the agrin-induced AChR clustering. 1,25D increased the expression of rapsyn, which is necessary for AChR clustering, while demonstrating no effect on other neuromuscular junction-related genes. In addition, rapsyn expression was dependent on 1,25D-VDR signaling. These results suggest that 1,25D-VDR signaling may regulate rapsin expression, resulting in the up-regulation of agrin-induced AChR clustering.
Collapse
Affiliation(s)
- Masayuki Arakawa
- Laboratory of Virology, Institute of Microbial Chemistry, 5-14-23 Kamiosaki, Shinagawa-ku, Tokyo, 141-0021, Japan
| | - Akira Wagatsuma
- Laboratory of Muscle Biology, Tokyo Woman's Christian University, 2-6-1 Zempukuji, Suginami-ku, Tokyo, 167-8585, Japan.
| |
Collapse
|
196
|
Decreased Serum Levels of C-Terminal Agrin in Postmenopausal Women Following Resistance Training. J Aging Phys Act 2020; 28:73-80. [DOI: 10.1123/japa.2019-0066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/27/2019] [Accepted: 05/30/2019] [Indexed: 11/18/2022]
Abstract
Elevated circulating C-terminal agrin fragment (CAF) is a marker of neuromuscular junction degradation and sarcopenia. This study sought to determine if resistance training (RT) impacted the serum levels of CAF in perimenopausal (PERI-M) and postmenopausal (POST-M) women. A total of 35 women, either PERI-M or POST-M, participated in 10 weeks of RT. Body composition, muscle strength, and serum estradiol and CAF were determined before and after the RT. The data were analyzed with two-way analysis of variance (p ≤ .05). Upper body and lower body strength was significantly increased, by 81% and 73% and 86% and 79% for the PERI-M and POST-M participants, respectively; however, there were no significant changes in body composition. Estradiol was significantly less for the POST-M participants at pretraining compared with the PERI-M participants. CAF moderately increased by 22% for the PERI-M participants in response to RT, whereas it significantly decreased by 49% for the POST-M participants. Ten weeks of RT reduced the circulating CAF in the POST-M women and might play a role in attenuating degenerative neuromuscular junction changes.
Collapse
|
197
|
Vilquin JT, Bayer AC, Le Panse R, Berrih-Aknin S. The Muscle Is Not a Passive Target in Myasthenia Gravis. Front Neurol 2020; 10:1343. [PMID: 31920954 PMCID: PMC6930907 DOI: 10.3389/fneur.2019.01343] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/05/2019] [Indexed: 12/22/2022] Open
Abstract
Myasthenia gravis (MG) is a rare autoimmune disease mediated by pathogenic antibodies (Ab) directed against components of the neuromuscular junction (NMJ), mainly the acetylcholine receptor (AChR). The etiological mechanisms are not totally elucidated, but they include a combination of genetic predisposition, triggering event(s), and hormonal components. MG disease is associated with defective immune regulation, chronic cell activation, inflammation, and the thymus is frequently abnormal. MG is characterized by muscle fatigability that is very invalidating and can be life-threatening when respiratory muscles are affected. MG is not cured, and symptomatic treatments with acetylcholinesterase inhibitors and immunosuppressors are life-long medications associated with severe side effects (especially glucocorticoids). While the muscle is the ultimate target of the autoimmune attack, its place and role are not thoroughly described, and this mini-review will focus on the cascade of pathophysiologic mechanisms taking place at the NMJ and its consequences on the muscle biology, function, and regeneration in myasthenic patients, at the histological, cellular, and molecular levels. The fine structure of the synaptic cleft is damaged by the Ab binding that is coupled to focal complement-dependent lysis in the case of MG with anti-AChR antibodies. Cellular and molecular reactions taking place in the muscle involve several cell types as well as soluble factors. Finally, the regenerative capacities of the MG muscle tissue may be altered. Altogether, the studies reported in this review demonstrate that the muscle is not a passive target in MG, but interacts dynamically with its environment in several ways, activating mechanisms of compensation that limit the pathogenic mechanisms of the autoantibodies.
Collapse
Affiliation(s)
- Jean-Thomas Vilquin
- Sorbonne Université, INSERM, Association Institut de Myologie (AIM), Paris, France
| | | | - Rozen Le Panse
- Sorbonne Université, INSERM, Association Institut de Myologie (AIM), Paris, France
| | - Sonia Berrih-Aknin
- Sorbonne Université, INSERM, Association Institut de Myologie (AIM), Paris, France
| |
Collapse
|
198
|
Geremek M, Dudarewicz L, Obersztyn E, Paczkowska M, Smyk M, Sobecka K, Nowakowska B. Null variants in AGRN cause lethal fetal akinesia deformation sequence. Clin Genet 2019; 97:634-638. [PMID: 31730230 DOI: 10.1111/cge.13677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 01/19/2023]
Abstract
We present a case of lethal fetal akinesia deformation sequence (FADS) caused by a frameshift variant in trans with a 148 kbp deletion encompassing 3-36 exons of AGRN. Pathogenic variants in AGRN have been described in families with a form of congenital myasthenic syndrome (CMS), manifesting in the early childhood with variable fatigable muscle weakness. To the best of our knowledge, this is the first case of FADS caused by defects in AGRN gene. FADS has been reported to be caused by pathogenic variants in genes previously associated with CMS including these involved in endplate development and maintenance: MuSK, DOK7, and RAPSN. FADS seems to be the most severe form of CMS. None of the reported in the literature CMS cases associated with AGRN had two null variants, like the case presented herein. This indicates a strong genotype-phenotype correlation.
Collapse
Affiliation(s)
- Maciej Geremek
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Lech Dudarewicz
- Department of Medical Genetics, Polish Mother's Memorial Hospital, Łódź, Poland
| | - Ewa Obersztyn
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | | | - Marta Smyk
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Katarzyna Sobecka
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| | - Beata Nowakowska
- Department of Medical Genetics, Institute of Mother and Child, Warsaw, Poland
| |
Collapse
|
199
|
Rimer M. Extracellular signal-regulated kinases 1 and 2 regulate neuromuscular junction and myofiber phenotypes in mammalian skeletal muscle. Neurosci Lett 2019; 715:134671. [PMID: 31805372 DOI: 10.1016/j.neulet.2019.134671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 02/06/2023]
Abstract
The neuromuscular junction is the synapse between a motor neuron of the spinal cord and a skeletal muscle fiber in the periphery. Reciprocal interactions between these excitable cells, and between them and others cell types present within the muscle tissue, shape the development, homeostasis and plasticity of skeletal muscle. An important aim in the field is to understand the molecular mechanisms underlying these cellular interactions, which include identifying the nature of the signals and receptors involved but also of the downstream intracellular signaling cascades elicited by them. This review focuses on work that shows that skeletal muscle fiber-derived extracellular signal-regulated kinases 1 and 2 (ERK1/2), ubiquitous and prototypical intracellular mitogen-activated protein kinases, have modulatory roles in the maintenance of the neuromuscular synapse and in the acquisition and preservation of fiber type identity in skeletal muscle.
Collapse
Affiliation(s)
- Mendell Rimer
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center and Texas A&M Institute for Neuroscience, Bryan, TX 77807 USA.
| |
Collapse
|
200
|
Zanetti G, Negro S, Megighian A, Mattarei A, Lista F, Fillo S, Rigoni M, Pirazzini M, Montecucco C. A CXCR4 receptor agonist strongly stimulates axonal regeneration after damage. Ann Clin Transl Neurol 2019; 6:2395-2402. [PMID: 31725979 PMCID: PMC6917312 DOI: 10.1002/acn3.50926] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 12/21/2022] Open
Abstract
Objective To test whether the signaling axis CXCL12α‐CXCR4 is activated upon crush/cut of the sciatic nerve and to test the activity of NUCC‐390, a new CXCR4 agonist, in promoting nerve recovery from damage. Methods The sciatic nerve was either crushed or cut. Expression and localization of CXCL12α and CXCR4 were evaluated by imaging with specific antibodies. Their functional involvement in nerve regeneration was determined by antibody‐neutralization of CXCL12α, and by the CXCR4 specific antagonist AMD3100, using as quantitative read‐out the compound muscle action potential (CMAP). NUCC‐390 activity on nerve regeneration was determined by imaging and CMAP recordings. Results CXCR4 is expressed at the injury site within the axonal compartment, whilst its ligand CXCL12α is expressed in Schwann cells. The CXCL12α‐CXCR4 axis is involved in the recovery of neurotransmission of the injured nerve. More importantly, the small molecule NUCC‐390 is a strong promoter of the functional and anatomical recovery of the nerve, by acting very similarly to CXCL12α. This pharmacological action is due to the capability of NUCC‐390 to foster elongation of motor neuron axons both in vitro and in vivo. Interpretation Imaging and electrophysiological data provide novel and compelling evidence that the CXCL12α‐CXCR4 axis is involved in sciatic nerve repair after crush/cut. This makes NUCC‐390 a strong candidate molecule to stimulate nerve repair by promoting axonal elongation. We propose this molecule to be tested in other models of neuronal damage, to lay the basis for clinical trials on the efficacy of NUCC‐390 in peripheral nerve repair in humans.
Collapse
Affiliation(s)
- Giulia Zanetti
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Padua Neuroscience Center, University of Padua, Padua, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Silvia Fillo
- Scientific Department, Army Medical Center, Roma, Italy
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,CNR Institute of Neuroscience, Padua, Italy
| |
Collapse
|