1
|
Dungan CM, Brightwell CR, Wen Y, Zdunek CJ, Latham CM, Thomas NT, Zagzoog AM, Brightwell BD, Nolt GL, Keeble AR, Watowich SJ, Murach KA, Fry CS. Muscle-Specific Cellular and Molecular Adaptations to Late-Life Voluntary Concurrent Exercise. FUNCTION 2022; 3:zqac027. [PMID: 35774589 PMCID: PMC9233305 DOI: 10.1093/function/zqac027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/06/2022] [Accepted: 05/08/2022] [Indexed: 01/07/2023] Open
Abstract
Murine exercise models can provide information on factors that influence muscle adaptability with aging, but few translatable solutions exist. Progressive weighted wheel running (PoWeR) is a simple, voluntary, low-cost, high-volume endurance/resistance exercise approach for training young mice. In the current investigation, aged mice (22-mo-old) underwent a modified version of PoWeR for 8 wk. Muscle functional, cellular, biochemical, transcriptional, and myonuclear DNA methylation analyses provide an encompassing picture of how muscle from aged mice responds to high-volume combined training. Mice run 6-8 km/d, and relative to sedentary mice, PoWeR increases plantarflexor muscle strength. The oxidative soleus of aged mice responds to PoWeR similarly to young mice in every parameter measured in previous work; this includes muscle mass, glycolytic-to-oxidative fiber type transitioning, fiber size, satellite cell frequency, and myonuclear number. The oxidative/glycolytic plantaris adapts according to fiber type, but with modest overall changes in muscle mass. Capillarity increases markedly with PoWeR in both muscles, which may be permissive for adaptability in advanced age. Comparison to published PoWeR RNA-sequencing data in young mice identified conserved regulators of adaptability across age and muscles; this includes Aldh1l1 which associates with muscle vasculature. Agrn and Samd1 gene expression is upregulated after PoWeR simultaneous with a hypomethylated promoter CpG in myonuclear DNA, which could have implications for innervation and capillarization. A promoter CpG in Rbm10 is hypomethylated by late-life exercise in myonuclei, consistent with findings in muscle tissue. PoWeR and the data herein are a resource for uncovering cellular and molecular regulators of muscle adaptation with aging.
Collapse
Affiliation(s)
- Cory M Dungan
- Department of Physical Therapy, University of Kentucky, Lexington 40536, KY, USA
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
| | - Camille R Brightwell
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Yuan Wen
- Department of Physical Therapy, University of Kentucky, Lexington 40536, KY, USA
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
| | | | - Christine M Latham
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Alyaa M Zagzoog
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Benjamin D Brightwell
- Kinesiology and Health Promotion Graduate Program, University of Kentucky, Lexington 40536, KY, USA
| | - Georgia L Nolt
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
| | - Alexander R Keeble
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| | - Stanley J Watowich
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston 77555, TX, USA
| | - Kevin A Murach
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Health, Human Performance, and Recreation, University of Arkansas, Fayetteville 72701, AR, USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville 72701, AR, USA
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington 40536, KY, USA
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington 40536, KY, USA
| |
Collapse
|
2
|
Jacquier A, Risson V, Simonet T, Roussange F, Lacoste N, Ribault S, Carras J, Theuriet J, Girard E, Grosjean I, Le Goff L, Kröger S, Meltoranta J, Bauché S, Sternberg D, Fournier E, Kostera-Pruszczyk A, O’Connor E, Eymard B, Lochmüller H, Martinat C, Schaeffer L. Severe congenital myasthenic syndromes caused by agrin mutations affecting secretion by motoneurons. Acta Neuropathol 2022; 144:707-731. [PMID: 35948834 PMCID: PMC9468088 DOI: 10.1007/s00401-022-02475-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 01/28/2023]
Abstract
Congenital myasthenic syndromes (CMS) are predominantly characterized by muscle weakness and fatigability and can be caused by a variety of mutations in genes required for neuromuscular junction formation and maintenance. Among them, AGRN encodes agrin, an essential synaptic protein secreted by motoneurons. We have identified severe CMS patients with uncharacterized p.R1671Q, p.R1698P and p.L1664P mutations in the LG2 domain of agrin. Overexpression in primary motoneurons cultures in vitro and in chick spinal motoneurons in vivo revealed that the mutations modified agrin trafficking, leading to its accumulation in the soma and/or in the axon. Expression of mutant agrins in cultured cells demonstrated accumulation of agrin in the endoplasmic reticulum associated with induction of unfolded protein response (UPR) and impaired secretion in the culture medium. Interestingly, evaluation of the specific activity of individual agrins on AChR cluster formation indicated that when secreted, mutant agrins retained a normal capacity to trigger the formation of AChR clusters. To confirm agrin accumulation and secretion defect, iPS cells were derived from a patient and differentiated into motoneurons. Patient iPS-derived motoneurons accumulated mutant agrin in the soma and increased XBP1 mRNA splicing, suggesting UPR activation. Moreover, co-cultures of patient iPS-derived motoneurons with myotubes confirmed the deficit in agrin secretion and revealed a reduction in motoneuron survival. Altogether, we report the first mutations in AGRN gene that specifically affect agrin secretion by motoneurons. Interestingly, the three patients carrying these mutations were initially suspected of spinal muscular atrophy (SMA). Therefore, in the presence of patients with a clinical presentation of SMA but without mutation in the SMN1 gene, it can be worth to look for mutations in AGRN.
Collapse
Affiliation(s)
- Arnaud Jacquier
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Valérie Risson
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France
| | - Thomas Simonet
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Florine Roussange
- grid.503216.30000 0004 0618 2124INSERM/UEPS UMR 861, Paris Saclay Université, I-STEM, 91100 Corbeil-Essonnes, France
| | - Nicolas Lacoste
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France
| | - Shams Ribault
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Service de Médecine Physique et de Réadaptation, Hôpital Henry Gabrielle, Hospices Civils de Lyon, 69230 Saint-Genis-Laval, France
| | - Julien Carras
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Julian Theuriet
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Emmanuelle Girard
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France
| | - Isabelle Grosjean
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France
| | - Laure Le Goff
- grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| | - Stephan Kröger
- Department of Physiological Genomics, Biomedical Center, Planegg, Martinsried, Germany
| | - Julia Meltoranta
- Department of Physiological Genomics, Biomedical Center, Planegg, Martinsried, Germany
| | - Stéphanie Bauché
- grid.462844.80000 0001 2308 1657Inserm U 1127, CNRS UMR 7225, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Épinière, ICM, Sorbonne Universités, 75013 Paris, France
| | - Damien Sternberg
- grid.462844.80000 0001 2308 1657Inserm U 1127, CNRS UMR 7225, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Épinière, ICM, Sorbonne Universités, 75013 Paris, France ,grid.411439.a0000 0001 2150 9058APHP, UF Cardiogénétique et Myogénétique, Service de Biochimie Métabolique, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Emmanuel Fournier
- grid.462844.80000 0001 2308 1657Inserm U 1127, CNRS UMR 7225, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Épinière, ICM, Sorbonne Universités, 75013 Paris, France ,grid.411439.a0000 0001 2150 9058AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France ,grid.462844.80000 0001 2308 1657Département d’Éthique de l’Université et des enseignements de Physiologie de la Faculté de Médecine Pitié-Salpêtrière, 75013 Paris, France
| | - Anna Kostera-Pruszczyk
- grid.13339.3b0000000113287408Department of Neurology, Medical University of Warsaw, Warsaw, Poland
| | - Emily O’Connor
- grid.28046.380000 0001 2182 2255Division of Neurology, Department of Medicine, Children’s Hospital of Eastern Ontario Research Institute, The Ottawa Hospital and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - Bruno Eymard
- grid.462844.80000 0001 2308 1657Inserm U 1127, CNRS UMR 7225, UPMC Université Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle Épinière, ICM, Sorbonne Universités, 75013 Paris, France ,grid.411439.a0000 0001 2150 9058AP-HP, Hôpital Pitié-Salpêtrière, 75013 Paris, France
| | - Hanns Lochmüller
- grid.28046.380000 0001 2182 2255Division of Neurology, Department of Medicine, Children’s Hospital of Eastern Ontario Research Institute, The Ottawa Hospital and Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - Cécile Martinat
- grid.503216.30000 0004 0618 2124INSERM/UEPS UMR 861, Paris Saclay Université, I-STEM, 91100 Corbeil-Essonnes, France
| | - Laurent Schaeffer
- Pathophysiology and Genetics of Neuron and Muscle, Faculté de Médecine Lyon Est, CNRS UMR 5261, INSERM U1315, Université Lyon1, Lyon, France ,grid.413852.90000 0001 2163 3825Hospices Civils de Lyon, Groupement Est, Bron, France
| |
Collapse
|
3
|
Spendiff S, Howarth R, McMacken G, Davey T, Quinlan K, O'Connor E, Slater C, Hettwer S, Mäder A, Roos A, Horvath R, Lochmüller H. Modulation of the Acetylcholine Receptor Clustering Pathway Improves Neuromuscular Junction Structure and Muscle Strength in a Mouse Model of Congenital Myasthenic Syndrome. Front Mol Neurosci 2021; 13:594220. [PMID: 33390901 PMCID: PMC7773664 DOI: 10.3389/fnmol.2020.594220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022] Open
Abstract
Introduction: Congenital myasthenic syndromes (CMS) are a diverse group of inherited neuromuscular disorders characterized by a failure of synaptic transmission at the neuromuscular junction (NMJ). CMS often present early with fatigable weakness and can be fatal through respiratory complications. The AGRN gene is one of over 30 genes known to harbor mutations causative for CMS. In this study, we aimed to determine if a compound (NT1654), developed to stimulate the acetylcholine receptor (AChR) clustering pathway, would benefit a mouse model of CMS caused by a loss-of-function mutation in Agrn (Agrnnmf380 mouse). Methods:Agrnnmf380 mice received an injection of either NT1654 or vehicle compound daily, with wild-type litter mates used for comparison. Animals were weighed daily and underwent grip strength assessments. After 30 days of treatment animals were sacrificed, and muscles collected. Investigations into NMJ and muscle morphology were performed on collected tissue. Results: While minimal improvements in NMJ ultrastructure were observed with electron microscopy, gross NMJ structure analysis using fluorescent labelling and confocal microscopy revealed extensive postsynaptic improvements in Agrnnmf380 mice with NT1654 administration, with variables frequently returning to wild type levels. An improvement in muscle weight and myofiber characteristics helped increase forelimb grip strength and body weight. Conclusions: We conclude that NT1654 restores NMJ postsynaptic structure and improves muscle strength through normalization of muscle fiber composition and the prevention of atrophy. We hypothesize this occurs through the AChR clustering pathway in Agrnnmf380 mice. Future studies should investigate if this may represent a viable treatment option for patients with CMS, especially those with mutations in proteins of the AChR clustering pathway.
Collapse
Affiliation(s)
- Sally Spendiff
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Rachel Howarth
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Grace McMacken
- Department of Neurosciences, Royal Victoria Hospital, Belfast, United Kingdom
| | - Tracey Davey
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kaitlyn Quinlan
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Emily O'Connor
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Clarke Slater
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | - Andreas Roos
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.,Department of Paediatric Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.,Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Neuropediatrics and Muscle Disorders, Medical Center - University of Freiburg, Freiburg, Germany.,Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| |
Collapse
|
4
|
Sanz B, Albillos Sanchez A, Tangey B, Gilmore K, Yue Z, Liu X, Wallace G. Light Cross-Linkable Marine Collagen for Coaxial Printing of a 3D Model of Neuromuscular Junction Formation. Biomedicines 2020; 9:16. [PMID: 33375335 PMCID: PMC7823301 DOI: 10.3390/biomedicines9010016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022] Open
Abstract
Collagen is a major component of the extracellular matrix (ECM) that modulates cell adhesion, growth, and migration, and has been utilised in tissue engineering applications. However, the common terrestrial sources of collagen carry the risk of zoonotic disease transmission and there are religious barriers to the use of bovine and porcine products in many cultures. Marine based collagens offer an attractive alternative and have so far been under-utilized for use as biomaterials for tissue engineering. Marine collagen can be extracted from fish waste products, therefore industry by-products offer an economical and environmentally sustainable source of collagen. In a handful of studies, marine collagen has successfully been methacrylated to form collagen methacrylate (ColMA). Our work included the extraction, characterization and methacrylation of Red Snapper collagen, optimisation of conditions for neural cell seeding and encapsulation using the unmodified collagen, thermally cross-linked, and the methacrylated collagen with UV-induced cross-linking. Finally, the 3D co-axial printing of neural and skeletal muscle cell cultures as a model for neuromuscular junction (NMJ) formation was investigated. Overall, the results of this study show great potential for a novel NMJ in vitro 3D bioprinted model that, with further development, could provide a low-cost, customizable, scalable and quick-to-print platform for drug screening and to study neuromuscular junction physiology and pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gordon Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Squires Way, Wollongong, New South Wales 2500, Australia; (B.S.); (A.A.S.); (B.T.); (K.G.); (Z.Y.); (X.L.)
| |
Collapse
|
5
|
Affiliation(s)
- Minkyung Kang
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA
| | - Yao Yao
- From the Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA
| |
Collapse
|
6
|
Wang B, Li Y, Sui M, Qi Q, Wang T, Liu D, Zhou M, Zheng Y, Zhu LQ, Zhang B. Identification of the downstream molecules of agrin/Dok-7 signaling in muscle. FASEB J 2020; 34:5144-5161. [PMID: 32043676 DOI: 10.1096/fj.201901693rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 01/15/2020] [Accepted: 01/29/2020] [Indexed: 11/11/2022]
Abstract
The development of the neuromuscular junction depends on signaling processes that involve protein phosphorylation. Motor neuron releases agrin to activate muscle protein Dok-7, a key tyrosine kinase essential for the formation of a mature and functional neuromuscular junction. However, the signaling cascade downstream of Dok-7 remains poorly understood. In this study, we combined the clustered regularly interspaced short palindromic repeats/Cas9 technique and quantitative phosphoproteomics analysis to study the tyrosine phosphorylation events triggered by agrin/Dok-7. We found tyrosine phosphorylation level of 36 proteins increased specifically by agrin stimulation. In Dok-7 mutant myotubes, however, 13 of the 36 proteins failed to be enhanced by agrin stimulation, suggesting that these 13 proteins are Dok-7-dependent tyrosine-phosphorylated proteins, could work as downstream molecules of agrin/Dok-7 signaling. We validated one of the proteins, Anxa3, by in vitro and in vivo assays. Knocking down of Anxa3 in the cultured myotubes inhibited agrin-induced AChR clustering, whereas reduction of Anxa3 in mouse muscles induced abnormal postsynaptic development. Collectively, our phosphoproteomics analysis provides novel insights into the complicated signaling network downstream of agrin/Dok-7.
Collapse
Affiliation(s)
- Beibei Wang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Li
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Sui
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Qinqin Qi
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Wang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Liu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Meiling Zhou
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Yunjie Zheng
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling-Qiang Zhu
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.,Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Li S, Wang Y, Jiang D, Ni D, Kutyreff CJ, Barnhart TE, Engle JW, Cai W. Spatiotemporal Distribution of Agrin after Intrathecal Injection and Its Protective Role in Cerebral Ischemia/Reperfusion Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902600. [PMID: 32076591 PMCID: PMC7029627 DOI: 10.1002/advs.201902600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/18/2019] [Indexed: 05/30/2023]
Abstract
Intrathecal injection, drugs transporting along perivascular spaces, represents an important route for maintaining blood-brain barrier (BBB) integrity after cerebral ischemia/reperfusion (I/R) injury. However, after being directly injected into cerebrospinal fluid (CSF), the temporal and spatial changes in the distribution of therapeutic protein drugs have remained unknown. Here, with positron emission tomography (PET) imaging, the uptake of 89Zr-agrin is noninvasively and dynamically monitored. These data demonstrate the time-activity curve of drugs in the brain subregions and their spatial distribution in different organs after intrathecal administration. Furthermore, agrin treatment effectively inhibits BBB disruption by reducing the loss of tight-junctional proteins. Importantly, the infarct volume is reduced; the number of apoptotic neurons is decreased; and neurological function is improved in mouse I/R injury models. Thus, intrathecal injection of agrin provides the basis for a new strategy to research and develop protein drugs for reducing the aggravation of I/R injury.
Collapse
Affiliation(s)
- Shiyong Li
- Department of RehabilitationSecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
- Department of NeurologySecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Ye Wang
- Department of NeurologySecond Affiliated Hospital of Nanchang UniversityNanchangJiangxi330006China
| | - Dawei Jiang
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Dalong Ni
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Christopher J. Kutyreff
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Todd E. Barnhart
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Jonathan W. Engle
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| | - Weibo Cai
- Departments of Radiology and Medical PhysicsUniversity of Wisconsin–MadisonMadisonWI53705USA
| |
Collapse
|
8
|
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
|
9
|
Luo B, Tian L, Chen N, Ramakrishna S, Thakor N, Yang IH. Electrospun nanofibers facilitate better alignment, differentiation, and long-term culture in an in vitro model of the neuromuscular junction (NMJ). Biomater Sci 2019; 6:3262-3272. [PMID: 30402630 DOI: 10.1039/c8bm00720a] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The neuromuscular junction (NMJ) is a specialized synapse between motor neurons and the muscle fibers they innervate. Due to the complexity of various signalling molecules and pathways, in vivo NMJs are difficult to study. Therefore, in vitro motor neuron-muscle co-culture plays a pivotal role in studying the mechanisms of NMJ formation associated with neurodegenerative diseases. There is a growing need to develop novel methodologies that can be used to develop long-term cultures of NMJs. To date, there have been few studies on NMJ development and long-term maintenance of the system, which is also the main challenge for the current in vitro models of NMJs. In this study, we demonstrate a long-term co-culture system of primary embryonic motor neurons from Sprague-Dawley rats and C2C12 cells on both random and aligned electrospun polylactic acid (PLA) nanofibrous scaffolds. This is the first study to explore the role of electrospun nanofibers in the long-term maintenance of NMJs. PLA nanofibrous scaffolds provide better contact guidance for C2C12 cells aligning along the fibers, thus guiding myotube formation. We can only maintain the co-culture system on a conventional glass substrate for 2 weeks, whilst 55% and 70% of the cells still survived on random and aligned PLA substrates after 7 weeks. Our nanofiber-based long-term co-culture system is used as an important tool for the fundamental research of NMJs.
Collapse
Affiliation(s)
- Baiwen Luo
- Singapore Institute for Neurotechnology, National University of Singapore, 28 Medical Drive, #05-COR, Singapore 119077. inhong.yang.@ku.ac.ae
| | | | | | | | | | | |
Collapse
|
10
|
Causes and consequences of age-related changes at the neuromuscular junction. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
11
|
MacDonald R, Barbat-Artigas S, Cho C, Peng H, Shang J, Moustaine A, Carbonetto S, Robitaille R, Chalifour LE, Paudel H. A Novel Egr-1-Agrin Pathway and Potential Implications for Regulation of Synaptic Physiology and Homeostasis at the Neuromuscular Junction. Front Aging Neurosci 2017; 9:258. [PMID: 28824419 PMCID: PMC5541023 DOI: 10.3389/fnagi.2017.00258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/19/2017] [Indexed: 12/11/2022] Open
Abstract
Synaptic transmission requires intricate coordination of the components involved in processing of incoming signals, formation and stabilization of synaptic machinery, neurotransmission and in all related signaling pathways. Changes to any of these components cause synaptic imbalance and disruption of neuronal circuitry. Extensive studies at the neuromuscular junction (NMJ) have greatly aided in the current understanding of synapses and served to elucidate the underlying physiology as well as associated adaptive and homeostatic processes. The heparan sulfate proteoglycan agrin is a vital component of the NMJ, mediating synaptic formation and maintenance in both brain and muscle, but very little is known about direct control of its expression. Here, we investigated the relationship between agrin and transcription factor early growth response-1 (Egr-1), as Egr-1 regulates the expression of many genes involved in synaptic homeostasis and plasticity. Using chromatin immunoprecipitation (ChIP), cell culture with cell lines derived from brain and muscle, and animal models, we show that Egr-1 binds to the AGRN gene locus and suppresses its expression. When compared with wild type (WT), mice deficient in Egr-1 (Egr-1−/−) display a marked increase in AGRN mRNA and agrin full-length and cleavage fragment protein levels, including the 22 kDa, C-terminal fragment in brain and muscle tissue homogenate. Because agrin is a crucial component of the NMJ, we explored possible physiological implications of the Egr-1-agrin relationship. In the diaphragm, Egr-1−/− mice display increased NMJ motor endplate density, individual area and area of innervation. In addition to increased density, soleus NMJs also display an increase in fragmented and faint endplates in Egr-1−/− vs. WT mice. Moreover, the soleus NMJ electrophysiology of Egr-1−/− mice revealed increased quantal content and motor testing showed decreased movement and limb muscle strength compared with WT. This study provides evidence for the potential involvement of a novel Egr-1-agrin pathway in synaptic homeostatic and compensatory mechanisms at the NMJ. Synaptic homeostasis is greatly affected by the process of aging. These and other data suggest that changes in Egr-1 expression may directly or indirectly promote age-related pathologies.
Collapse
Affiliation(s)
- Ryen MacDonald
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada
| | | | - Chulmin Cho
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada
| | - Huashan Peng
- Center for Research in NeuroscienceMontreal, QC, Canada
| | - Jijun Shang
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada
| | - Ayman Moustaine
- Département de neurosciences, Université de MontréalMontreal, QC, Canada
| | - Salvatore Carbonetto
- Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada.,Center for Research in NeuroscienceMontreal, QC, Canada.,Department of Medicine, McGill UniversityMontreal, QC, Canada
| | - Richard Robitaille
- Département de neurosciences, Université de MontréalMontreal, QC, Canada
| | - Lorraine E Chalifour
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Department of Medicine, McGill UniversityMontreal, QC, Canada
| | - Hemant Paudel
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada.,Department of Medicine, McGill UniversityMontreal, QC, Canada
| |
Collapse
|
12
|
Vezina-Audette R, Tremblay M, Carbonetto S. Laminin is instructive and calmodulin dependent kinase II is non-permissive for the formation of complex aggregates of acetylcholine receptors on myotubes in culture. Matrix Biol 2016; 57-58:106-123. [PMID: 27964993 DOI: 10.1016/j.matbio.2016.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 11/16/2016] [Indexed: 12/11/2022]
Abstract
Previous work has shown that myotubes cultured on laminin-coated substrates form complex aggregates of synaptic proteins that are similar in shape and composition to neuromuscular junctions (NMJs). Here we show that laminin instructs the location of complex aggregates which form only on the lower surface when laminin is coated onto culture dishes but over the entire cell when laminin is added in solution. Silencing of myotubes by agents that block electrical activity (tetrodotoxin, verapamil) or by inhibitors of calmodulin dependent kinase (CaMKII) render the myotube permissive for the formation of complex aggregates. Treatment with laminin alone will facilitate the formation of complex aggregates hours later when myotubes are made permissive by inhibiting CaMKII. The AChR agonist carbachol disperses pre formed aggregates suggesting that non-permissiveness may involve active dispersal of AChRs. The permissive period requires ongoing protein synthesis. The latter may reflect a requirement for rapsyn, which turns over rapidly, and is necessary for aggregation. Consistent with this geldanamycin, an agent that increases rapsyn turnover disrupts complex aggregates. Agrin is well known to induce small clusters of AChRs but does not induce complex aggregates even though aggregate formation requires MuSK, a receptor tyrosine kinase activated by agrin. Dystroglycan (DG) is the major laminin receptor mediating complex aggregate formation with some contribution from β1 integrins. In addition, there is a pool of CaMKII associated with DG. We discuss how these permissive and instructive mechanisms bear on NMJ formation in vivo.
Collapse
Affiliation(s)
- Raphael Vezina-Audette
- Centre for Research in Neuroscience, and Dept. of Neurology, McGill University Health Centre, 1650, Cedar Ave., Montreal, Quebec, H3G 1A4, Canada
| | - Mathieu Tremblay
- Centre for Research in Neuroscience, and Dept. of Neurology, McGill University Health Centre, 1650, Cedar Ave., Montreal, Quebec, H3G 1A4, Canada
| | - Salvatore Carbonetto
- Centre for Research in Neuroscience, and Dept. of Neurology, McGill University Health Centre, 1650, Cedar Ave., Montreal, Quebec, H3G 1A4, Canada.
| |
Collapse
|
13
|
Lozano R, Gilmore KJ, Thompson BC, Stewart EM, Waters AM, Romero-Ortega M, Wallace GG. Electrical stimulation enhances the acetylcholine receptors available for neuromuscular junction formation. Acta Biomater 2016; 45:328-339. [PMID: 27554016 DOI: 10.1016/j.actbio.2016.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 07/18/2016] [Accepted: 08/05/2016] [Indexed: 01/17/2023]
Abstract
Neuromuscular junctions (NMJ) are specialized synapses that link motor neurons with muscle fibers. These sites are fundamental to human muscle activity, controlling swallowing and breathing amongst many other vital functions. Study of this synapse formation is an essential area in neuroscience; the understanding of how neurons interact and control their targets during development and regeneration are fundamental questions. Existing data reveals that during initial stages of development neurons target and form synapses driven by biophysical and biochemical cues, and during later stages they require electrical activity to develop their functional interactions. The aim of this study was to investigate the effect of exogenous electrical stimulation (ES) electrodes directly in contact with cells, on the number and size of acetylcholine receptor (AChR) clusters available for NMJ formation. We used a novel in vitro model that utilizes a flexible electrical stimulation system and allows the systematic testing of several stimulation parameters simultaneously as well as the use of alternative electrode materials such as conductive polymers to deliver the stimulation. Functionality of NMJs under our co-culture conditions was demonstrated by monitoring changes in the responses of primary myoblasts to chemical stimulants that specifically target neuronal signaling. Our results suggest that biphasic electrical stimulation at 250Hz, 100μs pulse width and current density of 1mA/cm2 for 8h, applied via either gold-coated mylar or the conductive polymer PPy, significantly increased the number and size of AChRs clusters available for NMJ formation. This study supports the beneficial use of direct electrical stimulation as a strategic therapy for neuromuscular disorders. STATEMENT OF SIGNIFICANCE The beneficial effects of electrical stimulation (ES) on human cells in vitro and in vivo have long been known. Although the effects of stimulation are clear and the therapeutic benefits are known, no uniform parameters exist with regard to the duration, frequency and amplitude of the ES. To this end, we are answering several important questions on the parameters for ES of nerve and muscle monocultures and co-cultures by probing the effects on the enhancement of acetylcholine receptors (AChR) clustering available for neuromuscular junction formation using a conductive platform. This work opens the possibility to combine electrical stimulus delivered via conductive polymer substrates, from which biomolecules could also be delivered, providing opportunities to further enhance the therapeutic effect.
Collapse
Affiliation(s)
- Rodrigo Lozano
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, AIIM Facility, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Kerry J Gilmore
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, AIIM Facility, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Brianna C Thompson
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, AIIM Facility, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Elise M Stewart
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, AIIM Facility, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Aaron M Waters
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, AIIM Facility, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Mario Romero-Ortega
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Gordon G Wallace
- Intelligent Polymer Research Institute, ARC Centre of Excellence for Electromaterials Science, AIIM Facility, University of Wollongong, Wollongong, NSW 2522, Australia.
| |
Collapse
|
14
|
Neill T, Schaefer L, Iozzo RV. Decoding the Matrix: Instructive Roles of Proteoglycan Receptors. Biochemistry 2015; 54:4583-98. [PMID: 26177309 DOI: 10.1021/acs.biochem.5b00653] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The extracellular matrix is a dynamic repository harboring instructive cues that embody substantial regulatory dominance over many evolutionarily conserved intracellular activities, including proliferation, apoptosis, migration, motility, and autophagy. The matrix also coordinates and parses hierarchical information, such as angiogenesis, tumorigenesis, and immunological responses, typically providing the critical determinants driving each outcome. We provide the first comprehensive review focused on proteoglycan receptors, that is, signaling transmembrane proteins that use secreted proteoglycans as ligands, in addition to their natural ligands. The majority of these receptors belong to an exclusive subset of receptor tyrosine kinases and assorted cell surface receptors that specifically bind, transduce, and modulate fundamental cellular processes following interactions with proteoglycans. The class of small leucine-rich proteoglycans is the most studied so far and constitutes the best understood example of proteoglycan-receptor interactions. Decorin and biglycan evoke autophagy and immunological responses that deter, suppress, or exacerbate pathological conditions such as tumorigenesis, angiogenesis, and chronic inflammatory disease. Basement membrane-associated heparan sulfate proteoglycans (perlecan, agrin, and collagen XVIII) represent a unique cohort and provide proteolytically cleaved bioactive fragments for modulating cellular behavior. The receptors that bind the genuinely multifactorial and multivalent proteoglycans represent a nexus in understanding basic biological pathways and open new avenues for therapeutic and pharmacological intervention.
Collapse
Affiliation(s)
- Thomas Neill
- †Department of Pathology, Anatomy and Cell Biology and Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - Liliana Schaefer
- ‡Department of Pharmacology, Goethe University, 60590 Frankfurt, Germany
| | - Renato V Iozzo
- †Department of Pathology, Anatomy and Cell Biology and Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| |
Collapse
|
15
|
Tintignac LA, Brenner HR, Rüegg MA. Mechanisms Regulating Neuromuscular Junction Development and Function and Causes of Muscle Wasting. Physiol Rev 2015; 95:809-52. [DOI: 10.1152/physrev.00033.2014] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The neuromuscular junction is the chemical synapse between motor neurons and skeletal muscle fibers. It is designed to reliably convert the action potential from the presynaptic motor neuron into the contraction of the postsynaptic muscle fiber. Diseases that affect the neuromuscular junction may cause failure of this conversion and result in loss of ambulation and respiration. The loss of motor input also causes muscle wasting as muscle mass is constantly adapted to contractile needs by the balancing of protein synthesis and protein degradation. Finally, neuromuscular activity and muscle mass have a major impact on metabolic properties of the organisms. This review discusses the mechanisms involved in the development and maintenance of the neuromuscular junction, the consequences of and the mechanisms involved in its dysfunction, and its role in maintaining muscle mass during aging. As life expectancy is increasing, loss of muscle mass during aging, called sarcopenia, has emerged as a field of high medical need. Interestingly, aging is also accompanied by structural changes at the neuromuscular junction, suggesting that the mechanisms involved in neuromuscular junction maintenance might be disturbed during aging. In addition, there is now evidence that behavioral paradigms and signaling pathways that are involved in longevity also affect neuromuscular junction stability and sarcopenia.
Collapse
Affiliation(s)
- Lionel A. Tintignac
- Biozentrum, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland; and INRA, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France
| | - Hans-Rudolf Brenner
- Biozentrum, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland; and INRA, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France
| | - Markus A. Rüegg
- Biozentrum, University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland; and INRA, UMR866 Dynamique Musculaire et Métabolisme, Montpellier, France
| |
Collapse
|
16
|
Abstract
The neuromuscular junction (NMJ) is a synapse between motor neurons and skeletal muscle fibers, and is critical for control of muscle contraction. Its formation requires neuronal agrin that acts by binding to LRP4 to stimulate MuSK. Mutations have been identified in agrin, MuSK, and LRP4 in patients with congenital myasthenic syndrome, and patients with myasthenia gravis develop antibodies against agrin, LRP4, and MuSK. However, it remains unclear whether the agrin signaling pathway is critical for NMJ maintenance because null mutation of any of the three genes is perinatal lethal. In this study, we generated imKO mice, a mutant strain whose LRP4 gene can be deleted in muscles by doxycycline (Dox) treatment. Ablation of the LRP4 gene in adult muscle enabled studies of its role in NMJ maintenance. We demonstrate that Dox treatment of P30 mice reduced muscle strength and compound muscle action potentials. AChR clusters became fragmented with diminished junctional folds and synaptic vesicles. The amplitude and frequency of miniature endplate potentials were reduced, indicating impaired neuromuscular transmission and providing cellular mechanisms of adult LRP4 deficiency. We showed that LRP4 ablation led to the loss of synaptic agrin and the 90 kDa fragments, which occurred ahead of other prejunctional and postjunctional components, suggesting that LRP4 may regulate the stability of synaptic agrin. These observations demonstrate that LRP4 is essential for maintaining the structural and functional integrity of the NMJ and that loss of muscle LRP4 in adulthood alone is sufficient to cause myasthenic symptoms.
Collapse
|
17
|
Osses N, Henríquez JP. Bone morphogenetic protein signaling in vertebrate motor neurons and neuromuscular communication. Front Cell Neurosci 2015; 8:453. [PMID: 25674047 PMCID: PMC4307192 DOI: 10.3389/fncel.2014.00453] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/15/2014] [Indexed: 01/28/2023] Open
Abstract
An accurate communication between motor neurons and skeletal muscle fibers is required for the proper assembly, growth and maintenance of neuromuscular junctions (NMJs). Several signaling and extracellular matrix molecules play stimulatory and inhibitory roles on the assembly of functional synapses. Studies in Drosophila have revealed crucial functions for early morphogens, such as members of the Wnt and Bone Morphogenetic Proteins (BMP) signaling pathways, during the assembly and maturation of the NMJ. Here, we bring together recent findings that led us to propose that BMPs also work in vertebrate organisms as diffusible cues to communicate motor neurons and skeletal muscles.
Collapse
Affiliation(s)
- Nelson Osses
- BMP Research Group, Institute of Chemistry, Faculty of Sciences, Pontificia Universidad Católica de Valparaíso Valparaíso, Chile
| | - Juan P Henríquez
- Laboratory of Developmental Neurobiology, Department of Cell Biology, Faculty of Biological Sciences, Millennium Nucleus of Regenerative Biology, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción Concepción, Chile
| |
Collapse
|
18
|
Zhang Y, Lin S, Karakatsani A, Rüegg MA, Kröger S. Differential regulation of AChR clustering in the polar and equatorial region of murine muscle spindles. Eur J Neurosci 2014; 41:69-78. [PMID: 25377642 DOI: 10.1111/ejn.12768] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 09/29/2014] [Accepted: 10/02/2014] [Indexed: 12/16/2022]
Abstract
Intrafusal fibers of muscle spindles are innervated in the central region by afferent sensory axons and at both polar regions by efferent γ-motoneurons. We previously demonstrated that both neuron-muscle contact sites contain cholinergic synapse-like specialisation, including aggregates of the nicotinic acetylcholine receptor (AChR). In this study we tested the hypothesis that agrin and its receptor complex (consisting of LRP4 and the tyrosine kinase MuSK) are involved in the aggregation of AChRs in muscle spindles, similar to their role at the neuromuscular junction. We show that agrin, MuSK and LRP4 are concentrated at the contact site between the intrafusal fibers and the sensory- and γ-motoneuron, respectively, and that they are expressed in the cell bodies of proprioceptive neurons in dorsal root ganglia. Moreover, agrin and LRP4, but not MuSK, are expressed in γ-motoneuron cell bodies in the ventral horn of the spinal cord. In agrin- and in MuSK-deficient mice, AChR aggregates are absent from the polar regions. In contrast, the subcellular concentration of AChRs in the central region where the sensory neuron contacts the intrafusal muscle fiber is apparently unaffected. Skeletal muscle-specific expression of miniagrin in agrin(-/-) mice in vivo is sufficient to restore the formation of γ-motoneuron endplates. These results show that agrin and MuSK are major determinants during the formation of γ-motoneuron endplates but appear dispensable for the aggregation of AChRs at the central region. Our results therefore suggest different molecular mechanisms for AChR clustering within two domains of intrafusal fibers.
Collapse
Affiliation(s)
- Yina Zhang
- Department of Physiological Genomics, Ludwig-Maximilians-University, Pettenkoferstrasse 12, D-80336, Munich, Germany; Helmholtz Center Munich, Neuherberg, Germany
| | | | | | | | | |
Collapse
|
19
|
The MuSK activator agrin has a separate role essential for postnatal maintenance of neuromuscular synapses. Proc Natl Acad Sci U S A 2014; 111:16556-61. [PMID: 25368159 DOI: 10.1073/pnas.1408409111] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The motoneural control of skeletal muscle contraction requires the neuromuscular junction (NMJ), a midmuscle synapse between the motor nerve and myotube. The formation and maintenance of NMJs are orchestrated by the muscle-specific receptor tyrosine kinase (MuSK). Motor neuron-derived agrin activates MuSK via binding to MuSK's coreceptor Lrp4, and genetic defects in agrin underlie a congenital myasthenic syndrome (an NMJ disorder). However, MuSK-dependent postsynaptic differentiation of NMJs occurs in the absence of a motor neuron, indicating a need for nerve/agrin-independent MuSK activation. We previously identified the muscle protein Dok-7 as an essential activator of MuSK. Although NMJ formation requires agrin under physiological conditions, it is dispensable for NMJ formation experimentally in the absence of the neurotransmitter acetylcholine, which inhibits postsynaptic specialization. Thus, it was hypothesized that MuSK needs agrin together with Lrp4 and Dok-7 to achieve sufficient activation to surmount inhibition by acetylcholine. Here, we show that forced expression of Dok-7 in muscle enhanced MuSK activation in mice lacking agrin or Lrp4 and restored midmuscle NMJ formation in agrin-deficient mice, but not in Lrp4-deficient mice, probably due to the loss of Lrp4-dependent presynaptic differentiation. However, these NMJs in agrin-deficient mice rapidly disappeared after birth, and postsynaptic specializations emerged ectopically throughout myotubes whereas exogenous Dok-7-mediated MuSK activation was maintained. These findings demonstrate that the MuSK activator agrin plays another role essential for the postnatal maintenance, but not for embryonic formation, of NMJs and also for the postnatal, but not prenatal, midmuscle localization of postsynaptic specializations, providing physiological and pathophysiological insight into NMJ homeostasis.
Collapse
|
20
|
Steiner E, Enzmann GU, Lyck R, Lin S, Rüegg MA, Kröger S, Engelhardt B. The heparan sulfate proteoglycan agrin contributes to barrier properties of mouse brain endothelial cells by stabilizing adherens junctions. Cell Tissue Res 2014; 358:465-79. [PMID: 25107608 PMCID: PMC4210653 DOI: 10.1007/s00441-014-1969-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 07/17/2014] [Indexed: 01/13/2023]
Abstract
Barrier characteristics of brain endothelial cells forming the blood–brain barrier (BBB) are tightly regulated by cellular and acellular components of the neurovascular unit. During embryogenesis, the accumulation of the heparan sulfate proteoglycan agrin in the basement membranes ensheathing brain vessels correlates with BBB maturation. In contrast, loss of agrin deposition in the vasculature of brain tumors is accompanied by the loss of endothelial junctional proteins. We therefore wondered whether agrin had a direct effect on the barrier characteristics of brain endothelial cells. Agrin increased junctional localization of vascular endothelial (VE)-cadherin, β-catenin, and zonula occludens-1 (ZO-1) but not of claudin-5 and occludin in the brain endothelioma cell line bEnd5 without affecting the expression levels of these proteins. This was accompanied by an agrin-induced reduction of the paracellular permeability of bEnd5 monolayers. In vivo, the lack of agrin also led to reduced junctional localization of VE-cadherin in brain microvascular endothelial cells. Taken together, our data support the notion that agrin contributes to barrier characteristics of brain endothelium by stabilizing the adherens junction proteins VE-cadherin and β-catenin and the junctional protein ZO-1 to brain endothelial junctions.
Collapse
Affiliation(s)
- Esther Steiner
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | | | | | | | | | | | | |
Collapse
|
21
|
Hettwer S, Lin S, Kucsera S, Haubitz M, Oliveri F, Fariello RG, Ruegg MA, Vrijbloed JW. Injection of a soluble fragment of neural agrin (NT-1654) considerably improves the muscle pathology caused by the disassembly of the neuromuscular junction. PLoS One 2014; 9:e88739. [PMID: 24520420 PMCID: PMC3919806 DOI: 10.1371/journal.pone.0088739] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 01/10/2014] [Indexed: 01/13/2023] Open
Abstract
Treatment of neuromuscular diseases is still an unsolved problem. Evidence over the last years strongly indicates the involvement of malformation and dysfunction of neuromuscular junctions in the development of such medical conditions. Stabilization of NMJs thus seems to be a promising approach to attenuate the disease progression of muscle wasting diseases. An important pathway for the formation and maintenance of NMJs is the agrin/Lrp4/MuSK pathway. Here we demonstrate that the agrin biologic NT-1654 is capable of activating the agrin/Lrp4/MuSK system in vivo, leading to an almost full reversal of the sarcopenia-like phenotype in neurotrypsin-overexpressing (SARCO) mice. We also show that injection of NT-1654 accelerates muscle re-innervation after nerve crush. This report demonstrates that a systemically administered agrin fragment has the potential to counteract the symptoms of neuromuscular disorders.
Collapse
Affiliation(s)
| | - Shuo Lin
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
22
|
Ko IK, Lee BK, Lee SJ, Andersson KE, Atala A, Yoo JJ. The effect of in vitro formation of acetylcholine receptor (AChR) clusters in engineered muscle fibers on subsequent innervation of constructs in vivo. Biomaterials 2013; 34:3246-55. [PMID: 23391495 DOI: 10.1016/j.biomaterials.2013.01.029] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 01/04/2013] [Indexed: 11/16/2022]
Abstract
Timely innervation of muscle tissue is critical in the recovery of function, and this time-sensitive process relies heavily on the host tissue microenvironment after implantation. However, restoration of muscle tissue mass and function has been a challenge. We investigated whether pre-forming acetylcholine receptor (AChR) clusters on engineered muscle fibers using an AChR cluster-inducing factor (agrin) prior to implantation would facilitate established contacts between implanted muscle tissues and nerves and result in rapid innervation of engineered muscle in vivo. We showed that agrin treatment significantly increased the formation of AChR clusters on culture differentiated myotubes (C2C12), enhanced contacts with nerves in vitro and in vivo, and increased angiogenesis. Pre-fabrication of AChR clusters on engineered skeletal muscle using a released neurotrophic factor can accelerate innervations following implantation in vivo. This technique has considerable potential for enhancing muscle tissue function.
Collapse
Affiliation(s)
- In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | | | | | | |
Collapse
|
23
|
Chao T, Frump D, Lin M, Caiozzo VJ, Mozaffar T, Steward O, Gupta R. Matrix metalloproteinase 3 deletion preserves denervated motor endplates after traumatic nerve injury. Ann Neurol 2012; 73:210-23. [DOI: 10.1002/ana.23781] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 08/29/2012] [Accepted: 09/24/2012] [Indexed: 12/13/2022]
Affiliation(s)
- Tom Chao
- Department of Orthopaedic Surgery; University of California; Irvine, Irvine; CA
| | - Derek Frump
- Department of Orthopaedic Surgery; University of California; Irvine, Irvine; CA
| | - Michael Lin
- Department of Orthopaedic Surgery; University of California; Irvine, Irvine; CA
| | - Vincent J. Caiozzo
- Department of Orthopaedic Surgery; University of California; Irvine, Irvine; CA
| | | | | | | |
Collapse
|
24
|
Steiner E, Enzmann GU, Lin S, Ghavampour S, Hannocks MJ, Zuber B, Rüegg MA, Sorokin L, Engelhardt B. Loss of astrocyte polarization upon transient focal brain ischemia as a possible mechanism to counteract early edema formation. Glia 2012; 60:1646-59. [DOI: 10.1002/glia.22383] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 06/07/2012] [Indexed: 11/10/2022]
|
25
|
Punga AR, Ruegg MA. Signaling and aging at the neuromuscular synapse: lessons learnt from neuromuscular diseases. Curr Opin Pharmacol 2012; 12:340-6. [PMID: 22365504 DOI: 10.1016/j.coph.2012.02.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 01/26/2012] [Accepted: 02/02/2012] [Indexed: 12/30/2022]
Abstract
The neuromuscular junction (NMJ) is a specialized synapse between motor neurons and skeletal muscle with a complex signaling network that assures highly reliable neuromuscular transmission. Diseases of the NMJ cause skeletal muscle fatigue and include inherited and acquired disorders that affect presynaptic, intrasynaptic or postsynaptic components. Moreover, fragmentation of the NMJ contributes to sarcopenia, the loss of muscle mass during aging. Studies from recent years indicate that the formation and stabilization of NMJs differs between various muscles and that this difference affects their response under pathological conditions. This review summarizes the most important mechanisms involved in the development, maintenance and dysfunction of the NMJ and it discusses their significance in myasthenic disorders and aging and as targets for possible future treatment of NMJ dysfunction.
Collapse
Affiliation(s)
- Anna Rostedt Punga
- Institute of Neuroscience, Department of Clinical Neurophysiology, Uppsala University Hospital, Uppsala, Sweden
| | | |
Collapse
|
26
|
Ngo ST, Cole RN, Sunn N, Phillips WD, Noakes PG. Neuregulin-1 potentiates agrin-induced acetylcholine receptor clustering through muscle-specific kinase phosphorylation. J Cell Sci 2012; 125:1531-43. [PMID: 22328506 DOI: 10.1242/jcs.095109] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
At neuromuscular synapses, neural agrin (n-agrin) stabilizes embryonic postsynaptic acetylcholine receptor (AChR) clusters by signalling through the muscle-specific kinase (MuSK) complex. Live imaging of cultured myotubes showed that the formation and disassembly of primitive AChR clusters is a dynamic and reversible process favoured by n-agrin, and possibly other synaptic signals. Neuregulin-1 is a growth factor that can act through muscle ErbB receptor kinases to enhance synaptic gene transcription. Recent studies suggest that neuregulin-1-ErbB signalling can modulate n-agrin-induced AChR clustering independently of its effects on transcription. Here we report that neuregulin-1 increased the size of developing AChR clusters when injected into muscles of embryonic mice. We investigated this phenomenon using cultured myotubes, and found that in the ongoing presence of n-agrin, neuregulin-1 potentiates AChR clustering by increasing the tyrosine phosphorylation of MuSK. This potentiation could be blocked by inhibiting Shp2, a postsynaptic tyrosine phosphatase known to modulate the activity of MuSK. Our results provide new evidence that neuregulin-1 modulates the signaling activity of MuSK and hence might function as a second-order regulator of postsynaptic AChR clustering at the neuromuscular synapse. Thus two classic synaptic signalling systems (neuregulin-1 and n-agrin) converge upon MuSK to regulate postsynaptic differentiation.
Collapse
Affiliation(s)
- Shyuan T Ngo
- School of Biomedical Sciences, University of Queensland, St. Lucia, 4072, Queensland, Australia
| | | | | | | | | |
Collapse
|
27
|
Bian W, Bursac N. Soluble miniagrin enhances contractile function of engineered skeletal muscle. FASEB J 2011; 26:955-65. [PMID: 22075647 DOI: 10.1096/fj.11-187575] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neural agrin plays a pleiotropic role in skeletal muscle innervation and maturation, but its specific effects on the contractile function of aneural engineered muscle remain unknown. In this study, neonatal rat skeletal myoblasts cultured within 3-dimensional engineered muscle tissue constructs were treated with 10 nM soluble recombinant miniagrin and assessed using histological, biochemical, and functional assays. Depending on the treatment duration and onset time relative to the stage of myogenic differentiation, miniagrin was found to induce up to 1.7-fold increase in twitch and tetanus force amplitude. This effect was associated with the 2.3-fold up-regulation of dystrophin gene expression at 6 d after agrin removal and enhanced ACh receptor (AChR) cluster formation, but no change in cell number, expression of muscle myosin, or important aspects of intracellular Ca(2+) handling. In muscle constructs with endogenous ACh levels suppressed by the application of α-NETA, miniagrin increased AChR clustering and twitch force amplitude but failed to improve intracellular Ca(2+) handling and increase tetanus-to-twitch ratio. Overall, our studies suggest that besides its synaptogenic function that could promote integration of engineered muscle constructs in vivo, neural agrin can directly promote the contractile function of aneural engineered muscle via mechanisms distinct from those involving endogenous ACh.
Collapse
Affiliation(s)
- Weining Bian
- Department of Anesthesia and Medicine and Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
28
|
Punga AR, Lin S, Oliveri F, Meinen S, Rüegg MA. Muscle-selective synaptic disassembly and reorganization in MuSK antibody positive MG mice. Exp Neurol 2011; 230:207-17. [DOI: 10.1016/j.expneurol.2011.04.018] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 04/15/2011] [Accepted: 04/21/2011] [Indexed: 11/25/2022]
|
29
|
Nestin negatively regulates postsynaptic differentiation of the neuromuscular synapse. Nat Neurosci 2011; 14:324-30. [PMID: 21278733 PMCID: PMC3069133 DOI: 10.1038/nn.2747] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 12/22/2010] [Indexed: 01/14/2023]
Abstract
Positive and negative regulation of neurotransmitter receptor aggregation on the postsynaptic membrane is a critical event during synapse formation. Acetylcholine (ACh) and agrin are two opposing signals that regulate ACh receptor (AChR) clustering during neuromuscular junction (NMJ) development. ACh induces dispersion of AChR clusters that are not stabilized by agrin via a cyclin-dependent kinase 5 (Cdk5)-mediated mechanism, but regulation of Cdk5 activation is poorly understood. Here we show that the intermediate filament protein nestin physically interacts with Cdk5 and is required for ACh-induced association of p35, the co-activator of Cdk5, with the muscle membrane. Blockade of nestin-dependent signaling inhibits ACh-induced Cdk5 activation and the dispersion of AChR clusters in cultured myotubes. Similar to the effects of Cdk5 gene inactivation, knockdown of nestin in agrin-deficient embryos significantly restores AChR clusters. These results suggest that nestin is required for ACh-induced, Cdk5-dependent dispersion of AChR clusters during NMJ development.
Collapse
|
30
|
Yurchenco PD. Basement membranes: cell scaffoldings and signaling platforms. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a004911. [PMID: 21421915 DOI: 10.1101/cshperspect.a004911] [Citation(s) in RCA: 638] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Basement membranes are widely distributed extracellular matrices that coat the basal aspect of epithelial and endothelial cells and surround muscle, fat, and Schwann cells. These extracellular matrices, first expressed in early embryogenesis, are self-assembled on competent cell surfaces through binding interactions among laminins, type IV collagens, nidogens, and proteoglycans. They form stabilizing extensions of the plasma membrane that provide cell adhesion and that act as solid-phase agonists. Basement membranes play a role in tissue and organ morphogenesis and help maintain function in the adult. Mutations adversely affecting expression of the different structural components are associated with developmental arrest at different stages as well as postnatal diseases of muscle, nerve, brain, eye, skin, vasculature, and kidney.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA.
| |
Collapse
|
31
|
Punga AR, Maj M, Lin S, Meinen S, Rüegg MA. MuSK levels differ between adult skeletal muscles and influence postsynaptic plasticity. Eur J Neurosci 2011; 33:890-8. [PMID: 21255125 DOI: 10.1111/j.1460-9568.2010.07569.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Muscle-specific tyrosine kinase (MuSK) is involved in the formation and maintenance of the neuromuscular junction (NMJ), and is necessary for NMJ integrity. As muscle involvement is strikingly selective in pathological conditions in which MuSK is targeted, including congenital myasthenic syndrome with MuSK mutation and MuSK antibody-seropositive myasthenia gravis, we hypothesized that the postsynaptic response to MuSK-agrin signalling differs between adult muscles. Transcript levels of postsynaptic proteins were compared between different muscles in wild-type adult mice. MuSK expression was high in the soleus and sternomastoid muscles and low in the extensor digitorum longus (EDL) and omohyoid muscles. The acetylcholine receptor (AChR) α subunit followed a similar expression pattern, whereas expression of Dok-7, Lrp4 and rapsyn was comparable between the muscles. We subsequently examined muscles in mice that overexpressed a miniaturized form of neural agrin or MuSK. In these transgenic mice, the soleus and sternomastoid muscles responded with formation of ectopic AChR clusters, whereas such clusters were almost absent in the EDL and omohyoid muscles. Electroporation of Dok-7 revealed its important role as an activator of MuSK in AChR cluster formation in adult muscles. Together, our findings indicate for the first time that adult skeletal muscles harbour different endogenous levels of MuSK and that these levels determine the ability to form ectopic AChR clusters upon overexpression of agrin or MuSK. We believe that these findings are important for our understanding of adult muscle plasticity and the selective muscle involvement in neuromuscular disorders in which MuSK is diminished.
Collapse
Affiliation(s)
- Anna R Punga
- Department of Neurobiology/Pharmacology, Biozentrum, University of Basel, Basel, Switzerland.
| | | | | | | | | |
Collapse
|
32
|
Barros CS, Franco SJ, Müller U. Extracellular matrix: functions in the nervous system. Cold Spring Harb Perspect Biol 2011; 3:a005108. [PMID: 21123393 DOI: 10.1101/cshperspect.a005108] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
An astonishing number of extracellular matrix glycoproteins are expressed in dynamic patterns in the developing and adult nervous system. Neural stem cells, neurons, and glia express receptors that mediate interactions with specific extracellular matrix molecules. Functional studies in vitro and genetic studies in mice have provided evidence that the extracellular matrix affects virtually all aspects of nervous system development and function. Here we will summarize recent findings that have shed light on the specific functions of defined extracellular matrix molecules on such diverse processes as neural stem cell differentiation, neuronal migration, the formation of axonal tracts, and the maturation and function of synapses in the peripheral and central nervous system.
Collapse
Affiliation(s)
- Claudia S Barros
- The Scripps Research Institute, Department of Cell Biology, Dorris Neuroscience Center, La Jolla, California 92037, USA
| | | | | |
Collapse
|
33
|
Abstract
The most impressive structural feature of the nervous system is the specificity of its synaptic connections. Even after axons have navigated long distances to reach target areas, they must still choose appropriate synaptic partners from the many potential partners within easy reach. In many cases, axons also select a particular domain of the postsynaptic cell on which to form a synapse. Thus, synapse formation is selective at both cellular and subcellular levels. Unsurprisingly, the nervous system uses multiple mechanisms to ensure proper connectivity; these include complementary labels, coordinated growth of synaptic partners, sorting of afferents, prohibition or elimination of inappropriate synapses, respecification of targets, and use of short-range guidance mechanisms or intermediate targets. Specification of any circuit is likely to involve integration of multiple mechanisms. Recent studies of vertebrate and invertebrate systems have led to the identification of molecules that mediate a few of these interactions.
Collapse
Affiliation(s)
- Joshua R Sanes
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, USA.
| | | |
Collapse
|
34
|
Huzé C, Bauché S, Richard P, Chevessier F, Goillot E, Gaudon K, Ben Ammar A, Chaboud A, Grosjean I, Lecuyer HA, Bernard V, Rouche A, Alexandri N, Kuntzer T, Fardeau M, Fournier E, Brancaccio A, Rüegg MA, Koenig J, Eymard B, Schaeffer L, Hantaï D. Identification of an agrin mutation that causes congenital myasthenia and affects synapse function. Am J Hum Genet 2009; 85:155-67. [PMID: 19631309 DOI: 10.1016/j.ajhg.2009.06.015] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2009] [Revised: 06/16/2009] [Accepted: 06/23/2009] [Indexed: 11/16/2022] Open
Abstract
We report the case of a congenital myasthenic syndrome due to a mutation in AGRN, the gene encoding agrin, an extracellular matrix molecule released by the nerve and critical for formation of the neuromuscular junction. Gene analysis identified a homozygous missense mutation, c.5125G>C, leading to the p.Gly1709Arg variant. The muscle-biopsy specimen showed a major disorganization of the neuromuscular junction, including changes in the nerve-terminal cytoskeleton and fragmentation of the synaptic gutters. Experiments performed in nonmuscle cells or in cultured C2C12 myotubes and using recombinant mini-agrin for the mutated and the wild-type forms showed that the mutated form did not impair the activation of MuSK or change the total number of induced acetylcholine receptor aggregates. A solid-phase assay using the dystrophin glycoprotein complex showed that the mutation did not affect the binding of agrin to alpha-dystroglycan. Injection of wild-type or mutated agrin into rat soleus muscle induced the formation of nonsynaptic acetylcholine receptor clusters, but the mutant protein specifically destabilized the endogenous neuromuscular junctions. Importantly, the changes observed in rat muscle injected with mutant agrin recapitulated the pre- and post-synaptic modifications observed in the patient. These results indicate that the mutation does not interfere with the ability of agrin to induce postsynaptic structures but that it dramatically perturbs the maintenance of the neuromuscular junction.
Collapse
MESH Headings
- Adult
- Agrin/chemistry
- Agrin/genetics
- Agrin/metabolism
- Animals
- Biopsy
- Cell Line
- DNA Mutational Analysis
- Dystroglycans/metabolism
- Female
- Humans
- Male
- Models, Chemical
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/surgery
- Muscle, Skeletal/ultrastructure
- Mutation, Missense
- Myasthenic Syndromes, Congenital/genetics
- Neuromuscular Junction/genetics
- Neuromuscular Junction/metabolism
- Neuromuscular Junction/physiology
- Neuromuscular Junction/ultrastructure
- Pedigree
- Protein Structure, Tertiary
- Rats
- Receptors, Cholinergic/genetics
- Receptors, Cholinergic/metabolism
- Receptors, Cholinergic/physiology
- Recombinant Proteins/chemistry
- Recombinant Proteins/metabolism
- Synapses/metabolism
Collapse
Affiliation(s)
- Caroline Huzé
- Equipe Différenciation Neuromusculaire, UMR 5239, Ecole Normale Supérieure Lyon, CNRS, Université Lyon 1, Lyon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Pawlikowski BT, Maimone MM. Formation of complex AChR aggregates in vitro requires alpha-dystrobrevin. Dev Neurobiol 2009; 69:326-38. [PMID: 19224566 DOI: 10.1002/dneu.20703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Efficient function at the neuromuscular junction requires high-density aggregates of acetylcholine receptors (AChRs) to be precisely aligned with the motor nerve terminal. A collaborative effort between the motor neuron and muscle intrinsic factors drives the formation and maintenance of these AChR aggregates. alpha-Dystrobrevin (alpha DB), a cytoplasmic protein found at the postsynaptic membrane, has been implicated in the regulation of AChR aggregate density and patterning. To investigate the contribution of alpha DB to the muscle intrinsic program regulating AChR aggregate development, we analyzed the formation of complex, pretzel-like AChR aggregates on primary muscle cell cultures derived from alpha DB knockout (alpha DB-KO) mice in the absence of nerve or agrin. In myotubes lacking alpha DB, complex AChR aggregates failed to form, whereas aggregates formed readily in wildtype myotubes. Five major isoforms of alpha DB are expressed in skeletal muscle: alpha DB1, alpha DB1(-), alpha DB2, alpha DB2(-), and alpha DB3. Expression of alpha DB1 or alpha DB1(-) in alpha DB-KO myotubes restored formation of complex AChR aggregates similar to those in wildtype myotubes. In contrast, individual expression of alpha DB2, alpha DB2(-), alpha DB3, or an alpha DB1 phosphorylation mutant resulted in the formation of few, if any, complex AChR aggregates. Collectively, these data suggest that alpha DB is a significant component of the muscle intrinsic program that mediates the formation of complex AChR aggregates and that alpha DB's tyrosine phosphorylation sites are of particular functional importance to this program. Although the muscle intrinsic program appears to influence synaptogenesis, the formation of complex mature AChR aggregates in alpha DB-KO mice (with the motor neuron present) suggests the motor neuron, not the muscle intrinsic program, is the major stimulus driving the maturation of AChRs from plaque to pretzel in vivo.
Collapse
Affiliation(s)
- Bradley T Pawlikowski
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York 13210, USA.
| | | |
Collapse
|
36
|
Iozzo RV, Zoeller JJ, Nyström A. Basement membrane proteoglycans: modulators Par Excellence of cancer growth and angiogenesis. Mol Cells 2009; 27:503-13. [PMID: 19466598 PMCID: PMC6712562 DOI: 10.1007/s10059-009-0069-0] [Citation(s) in RCA: 171] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Accepted: 04/25/2009] [Indexed: 01/13/2023] Open
Abstract
Proteoglycans located in basement membranes, the nanostructures underling epithelial and endothelial layers, are unique in several respects. They are usually large, elongated molecules with a collage of domains that share structural and functional homology with numerous extracellular matrix proteins, growth factors and surface receptors. They mainly carry heparan sulfate side chains and these contribute not only to storing and preserving the biological activity of various heparan sulfate-binding cytokines and growth factors, but also in presenting them in a more "active configuration" to their cognate receptors. Abnormal expression or deregulated function of these proteoglycans affect cancer and angiogenesis, and are critical for the evolution of the tumor microenvironment. This review will focus on the functional roles of the major heparan sulfate proteoglycans from basement membrane zones: perlecan, agrin and collagen XVIII, and on their roles in modulating cancer growth and angiogenesis.
Collapse
Affiliation(s)
- Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA.
| | | | | |
Collapse
|
37
|
Inoue A, Setoguchi K, Matsubara Y, Okada K, Sato N, Iwakura Y, Higuchi O, Yamanashi Y. Dok-7 activates the muscle receptor kinase MuSK and shapes synapse formation. Sci Signal 2009; 2:ra7. [PMID: 19244212 DOI: 10.1126/scisignal.2000113] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The formation of the neuromuscular junction (NMJ) is orchestrated by the muscle-specific receptor tyrosine kinase MuSK and by neural agrin, an extracellular activator of MuSK. We previously showed that the MuSK-interacting protein Dok-7 is essential for neuromuscular synaptogenesis, although the mechanisms by which Dok-7 regulates MuSK activity and promotes synapse formation have been unclear. Here, we show that Dok-7 directly interacts with the cytoplasmic portion of MuSK and activates the receptor tyrosine kinase, and that neural agrin requires Dok-7 to activate MuSK. In vivo overexpression of Dok-7 increased MuSK activation and promoted NMJ formation. Furthermore, Dok-7 was required for the localization of MuSK in the central region of muscle, which is essential for the correct formation of NMJs in this region. These observations indicate that Dok-7 positively regulates neuromuscular synaptogenesis by controlling MuSK activity, its distribution, and its responsiveness to neural agrin.
Collapse
Affiliation(s)
- Akane Inoue
- Division of Genetics and Department of Cancer Biology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The heparan sulfate proteoglycan agrin is best known for its essential role during formation, maintenance and regeneration of the neuromuscular junction. Mutations in agrin-interacting proteins are the genetic basis for a number of neuromuscular disorders. However, agrin is widely expressed in many tissues including neurons and glial cells of the brain, where its precise function is much less understood. Fewer synapses develop in brains that lack agrin, consistent with a function of agrin during CNS synaptogenesis. Recently, a specific transmembrane form of agrin (TM-agrin) was identified that is concentrated at that interneuronal synapses in the brain. Clustering or overexpression of TM-agrin leads to the formation of filopodia-like processes, which might be precursors for CNS synapses. Agrin is subject to defined and activity-dependent proteolytic cleavage by neurotrypsin at synapses and dysregulation of agrin processing might contribute to the development of mental retardation. This review summarizes what is known about the role of agrin during synapse formation at the neuromuscular junction and in the developing CNS and will discuss additional functions of agrin in the adult CNS, in particular during BBB formation, during recovery after traumatic brain injury and in the etiology of diseases, including Alzheimer’s disease and mental retardation.
Collapse
Affiliation(s)
- Stephan Kröger
- Department of Physiological Genomics, Ludwig-Maximilians University, Schillerstrasse 46, D-80336 Munich, Germany
| | - Heike Pfister
- Department of Physiological Genomics, Ludwig-Maximilians University, Schillerstrasse 46, D-80336 Munich, Germany
| |
Collapse
|