Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.746
Filter
1.
J Vis Exp ; (199)2023 09 15.
Article in English | MEDLINE | ID: mdl-37782083

ABSTRACT

The Drosophila neuromuscular junction (NMJ) has emerged as a valuable model system in the field of neuroscience. The application of confocal microscopy at the Drosophila NMJ enables researchers to acquire synaptic information, encompassing both quantitative data on synapse abundance and detailed insights into their morphology. However, the diffuse distribution and limited visual range of the TEM present challenges for the ultrastructural analysis. This study introduces an innovative and efficient sample preparation method that surpasses the conventional approach. The procedure begins by placing a metal mesh at the base of a flat-bottomed bottle or test tube, followed by positioning fixed larvae samples onto the mesh. An additional mesh is placed over the samples, ensuring that they are positioned between the two meshes. The fixed samples are thoroughly dehydrated and infiltrated before proceeding with the embedding procedure. Then embedding of the samples in epoxy resin is performed in a flat sheet manner, which allows for the preparation of muscles for positioning and sectioning. Benefiting from these steps, all the muscles of Drosophila larvae can be visualized under light microscopy, therey facilitating subsequent positioning and sectioning. Excess resin is removed after locating the 6th and 7th muscles of body segments A2 and A3. Serial ultra-thin sectioning of the 6th or 7th muscle is performed.


Subject(s)
Drosophila , Neuromuscular Junction , Animals , Larva , Microscopy, Electron, Transmission , Neuromuscular Junction/ultrastructure , Synapses
2.
J Genet Genomics ; 50(3): 163-177, 2023 03.
Article in English | MEDLINE | ID: mdl-36473687

ABSTRACT

Formation and plasticity of neural circuits rely on precise regulation of synaptic growth. At Drosophila neuromuscular junction (NMJ), Bone Morphogenetic Protein (BMP) signaling is critical for many aspects of synapse formation and function. The evolutionarily conserved retromer complex and its associated GTPase-activating protein TBC1D5 are critical regulators of membrane trafficking and cellular signaling. However, their functions in regulating the formation of NMJ are less understood. Here, we report that TBC1D5 is required for inhibition of synaptic growth, and loss of TBC1D5 leads to abnormal presynaptic terminal development, including excessive satellite boutons and branch formation. Ultrastructure analysis reveals that the size of synaptic vesicles and the density of subsynaptic reticulum are increased in TBC1D5 mutant boutons. Disruption of interactions of TBC1D5 with Rab7 and retromer phenocopies the loss of TBC1D5. Unexpectedly, we find that TBC1D5 is functionally linked to Rab6, in addition to Rab7, to regulate synaptic growth. Mechanistically, we show that loss of TBC1D5 leads to upregulated BMP signaling by increasing the protein level of BMP type II receptor Wishful Thinking (Wit) at NMJ. Overall, our data establish that TBC1D5 in coordination with retromer constrains synaptic growth by regulating Rab7 activity, which negatively regulates BMP signaling through inhibiting Wit level.


Subject(s)
Drosophila Proteins , GTPase-Activating Proteins , Animals , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Signal Transduction/physiology , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Synapses/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Receptors, Cell Surface
3.
Elife ; 102021 08 27.
Article in English | MEDLINE | ID: mdl-34448452

ABSTRACT

Skeletal muscles are composed of hundreds of multinucleated muscle fibers (myofibers) whose myonuclei are regularly positioned all along the myofiber's periphery except the few ones clustered underneath the neuromuscular junction (NMJ) at the synaptic zone. This precise myonuclei organization is altered in different types of muscle disease, including centronuclear myopathies (CNMs). However, the molecular machinery regulating myonuclei position and organization in mature myofibers remains largely unknown. Conversely, it is also unclear how peripheral myonuclei positioning is lost in the related muscle diseases. Here, we describe the microtubule-associated protein, MACF1, as an essential and evolutionary conserved regulator of myonuclei positioning and maintenance, in cultured mammalian myotubes, in Drosophila muscle, and in adult mammalian muscle using a conditional muscle-specific knockout mouse model. In vitro, we show that MACF1 controls microtubules dynamics and contributes to microtubule stabilization during myofiber's maturation. In addition, we demonstrate that MACF1 regulates the microtubules density specifically around myonuclei, and, as a consequence, governs myonuclei motion. Our in vivo studies show that MACF1 deficiency is associated with alteration of extra-synaptic myonuclei positioning and microtubules network organization, both preceding NMJ fragmentation. Accordingly, MACF1 deficiency results in reduced muscle excitability and disorganized triads, leaving voltage-activated sarcoplasmic reticulum Ca2+ release and maximal muscle force unchanged. Finally, adult MACF1-KO mice present an improved resistance to fatigue correlated with a strong increase in mitochondria biogenesis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Microfilament Proteins/metabolism , Microtubules/metabolism , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Neuromuscular Junction/metabolism , Organelle Biogenesis , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/ultrastructure , Excitation Contraction Coupling , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microtubules/genetics , Microtubules/ultrastructure , Mitochondria, Muscle/genetics , Mitochondria, Muscle/ultrastructure , Muscle Fatigue , Muscle Fibers, Skeletal/ultrastructure , Muscle Strength , Myoblasts, Skeletal/ultrastructure , Neuromuscular Junction/genetics , Neuromuscular Junction/ultrastructure , Time Factors
4.
J Cell Biol ; 220(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34213535

ABSTRACT

The extracellular matrix has emerged as an active component of chemical synapses regulating synaptic formation, maintenance, and homeostasis. The heparan sulfate proteoglycan (HSPG) syndecans are known to regulate cellular and axonal migration in the brain. They are also enriched at synapses, but their synaptic functions remain more elusive. Here, we show that SDN-1, the sole orthologue of syndecan in C. elegans, is absolutely required for the synaptic clustering of homomeric α7-like acetylcholine receptors (AChRs) and regulates the synaptic content of heteromeric AChRs. SDN-1 is concentrated at neuromuscular junctions (NMJs) by the neurally secreted synaptic organizer Ce-Punctin/MADD-4, which also activates the transmembrane netrin receptor DCC. Those cooperatively recruit the FARP and CASK orthologues that localize α7-like-AChRs at cholinergic NMJs through physical interactions. Therefore, SDN-1 stands at the core of the cholinergic synapse organization by bridging the extracellular synaptic determinants to the intracellular synaptic scaffold that controls the postsynaptic receptor content.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , Receptors, Cholinergic/metabolism , Synapses/metabolism , Syndecans/metabolism , Acetylcholine/metabolism , Angiopoietin-Like Protein 4/genetics , Angiopoietin-Like Protein 4/metabolism , Animals , Brain/cytology , Brain/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , DCC Receptor/genetics , DCC Receptor/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Gene Expression Regulation , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Nerve Tissue Proteins/genetics , Neuromuscular Junction/ultrastructure , Neurons/cytology , Neurons/metabolism , Receptors, Cholinergic/genetics , Synapses/ultrastructure , Synaptic Transmission/genetics , Syndecans/genetics
5.
Biomed Pharmacother ; 137: 111357, 2021 05.
Article in English | MEDLINE | ID: mdl-33724918

ABSTRACT

3,4-Diaminopyridine (3,4-DAP) and its phosphate form, 3,4-DAPP have been used efficiently in the past years to treat muscular weakness in myasthenic syndromes with neuromuscular junctions (NMJs) impairment. Pompe disease (PD), an autosomal recessive metabolic disorder due to a defect of the lysosomal enzyme α-glucosidase (GAA), presents some secondary symptoms that are related to neuromuscular transmission dysfunction, resulting in endurance and strength failure. In order to evaluate whether 3,4-DAPP could have a beneficial effect on this pathology, we took advantage of a transient zebrafish PD model that we previously generated and characterized. We investigated presynaptic and postsynaptic structures, NMJs at the electron microscopy level, and zebrafish behavior, before and after treatment with 3,4-DAPP. After drug administration, we observed an increase in the number of acetylcholine receptors an increment in the percentage of NMJs with normal structure and amelioration in embryo behavior, with recovery of typical movements that were lost in the embryo PD model. Our results revealed early NMJ impairment in Pompe zebrafish model with improvement after administration of 3,4-DAPP, suggesting its potential use as symptomatic drug in patients with Pompe disease.


Subject(s)
Amifampridine/therapeutic use , Glycogen Storage Disease Type II/drug therapy , Neuromuscular Junction/drug effects , Animals , Behavior, Animal , Embryo, Nonmammalian , Motor Activity/drug effects , Muscle Fibers, Skeletal/drug effects , Neuromuscular Junction/ultrastructure , Receptors, Cholinergic/drug effects , Zebrafish , alpha-Glucosidases/metabolism
6.
J Vis Exp ; (166)2020 12 03.
Article in English | MEDLINE | ID: mdl-33346186

ABSTRACT

The neuromuscular junction (NMJ) is a specialized synapse that transmits action potentials from the motor neuron to skeletal muscle for mechanical movement. The architecture of the NMJ structure influences the functions of the neuron, the muscle and the mutual interaction. Previous studies have reported many strategies by co-culturing the motor neurons and myotubes to generate NMJ in vitro with complex induction process and long culture period but have struggled to recapitulate mature NMJ morphology and function. Our in vitro NMJ induction system is constructed by differentiating human iPSC in a single culture dish. By switching the myogenic and neurogenic induction medium for induction, the resulting NMJ contained pre- and post- synaptic components, including motor neurons, skeletal muscle and Schwann cells in the one month culture. The functional assay of NMJ also showed that the myotubes contraction can be triggered by Ca++ then inhibited by curare, an acetylcholine receptor (AChR) inhibitor, in which the stimulating signal is transmitted through NMJ. This simple and robust approach successfully derived the complex structure of NMJ with functional connectivity. This in vitro human NMJ, with its integrated structures and function, has promising potential for studying pathological mechanisms and compound screening.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neuromuscular Junction/cytology , Animals , Curare , Extracellular Matrix/metabolism , Humans , Induced Pluripotent Stem Cells/ultrastructure , Muscle Contraction/physiology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Neuromuscular Junction/ultrastructure , Schwann Cells/cytology , Schwann Cells/metabolism
7.
Biol Open ; 9(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-33148607

ABSTRACT

The amyotrophic lateral sclerosis (ALS) neurodegenerative disorder has been associated with multiple genetic lesions, including mutations in the gene for fused in sarcoma (FUS), a nuclear-localized RNA/DNA-binding protein. Neuronal expression of the pathological form of FUS proteins in Caenorhabditis elegans results in mislocalization and aggregation of FUS in the cytoplasm, and leads to impairment of motility. However, the mechanisms by which the mutant FUS disrupts neuronal health and function remain unclear. Here we investigated the impact of ALS-associated FUS on motor neuron health using correlative light and electron microscopy, electron tomography, and electrophysiology. We show that ectopic expression of wild-type or ALS-associated human FUS impairs synaptic vesicle docking at neuromuscular junctions. ALS-associated FUS led to the emergence of a population of large, electron-dense, and filament-filled endosomes. Electrophysiological recording revealed reduced transmission from motor neurons to muscles. Together, these results suggest a pathological effect of ALS-causing FUS at synaptic structure and function organization.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Amyotrophic Lateral Sclerosis/etiology , Gene Expression , Mutation , Neuromuscular Junction/genetics , Neuromuscular Junction/physiopathology , RNA-Binding Protein FUS/genetics , Synaptic Transmission/genetics , Animals , Caenorhabditis elegans , Disease Models, Animal , Disease Susceptibility , Endosomes/metabolism , Endosomes/ultrastructure , Humans , Motor Neurons/metabolism , Motor Neurons/ultrastructure , Neuromuscular Junction/pathology , Neuromuscular Junction/ultrastructure , Synaptic Potentials
8.
PLoS Genet ; 16(11): e1009179, 2020 11.
Article in English | MEDLINE | ID: mdl-33175853

ABSTRACT

Gene therapy approaches for DMD using recombinant adeno-associated viral (rAAV) vectors to deliver miniaturized (or micro) dystrophin genes to striated muscles have shown significant progress. However, concerns remain about the potential for immune responses against dystrophin in some patients. Utrophin, a developmental paralogue of dystrophin, may provide a viable treatment option. Here we examine the functional capacity of an rAAV-mediated microutrophin (µUtrn) therapy in the mdx4cv mouse model of DMD. We found that rAAV-µUtrn led to improvement in dystrophic histopathology & mostly restored the architecture of the neuromuscular and myotendinous junctions. Physiological studies of tibialis anterior muscles indicated peak force maintenance, with partial improvement of specific force. A fundamental question for µUtrn therapeutics is not only can it replace critical functions of dystrophin, but whether full-length utrophin impacts the therapeutic efficacy of the smaller, highly expressed µUtrn. As such, we found that µUtrn significantly reduced the spacing of the costameric lattice relative to full-length utrophin. Further, immunostaining suggested the improvement in dystrophic pathophysiology was largely influenced by favored correction of fast 2b fibers. However, unlike µUtrn, µdystrophin (µDys) expression did not show this fiber type preference. Interestingly, µUtrn was better able to protect 2a and 2d fibers in mdx:utrn-/- mice than in mdx4cv mice where the endogenous full-length utrophin was most prevalent. Altogether, these data are consistent with the role of steric hindrance between full-length utrophin & µUtrn within the sarcolemma. Understanding the stoichiometry of this effect may be important for predicting clinical efficacy.


Subject(s)
Genetic Therapy/methods , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/therapy , Utrophin/therapeutic use , Animals , Dependovirus/genetics , Disease Models, Animal , Dystrophin/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred mdx , Microscopy, Electron , Muscle Contraction , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Neuromuscular Junction/pathology , Neuromuscular Junction/ultrastructure , Sarcolemma/pathology , Sarcolemma/ultrastructure , Utrophin/genetics
9.
Cell Rep ; 32(1): 107866, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32640231

ABSTRACT

Glutamate receptor auxiliary proteins control receptor distribution and function, ultimately controlling synapse assembly, maturation, and plasticity. At the Drosophila neuromuscular junction (NMJ), a synapse with both pre- and postsynaptic kainate-type glutamate receptors (KARs), we show that the auxiliary protein Neto evolved functionally distinct isoforms to modulate synapse development and homeostasis. Using genetics, cell biology, and electrophysiology, we demonstrate that Neto-α functions on both sides of the NMJ. In muscle, Neto-α limits the size of the postsynaptic receptor field. In motor neurons (MNs), Neto-α controls neurotransmitter release in a KAR-dependent manner. In addition, Neto-α is both required and sufficient for the presynaptic increase in neurotransmitter release in response to reduced postsynaptic sensitivity. This KAR-independent function of Neto-α is involved in activity-induced cytomatrix remodeling. We propose that Drosophila ensures NMJ functionality by acquiring two Neto isoforms with differential expression patterns and activities.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Homeostasis , Membrane Proteins/metabolism , Neuromuscular Junction/metabolism , Synapses/metabolism , Animals , Calcium/metabolism , Drosophila Proteins/chemistry , Drosophila melanogaster/ultrastructure , Membrane Proteins/chemistry , Neuromuscular Junction/ultrastructure , Post-Synaptic Density/ultrastructure , Protein Domains , Receptors, Glutamate/metabolism
10.
Methods Mol Biol ; 2143: 145-157, 2020.
Article in English | MEDLINE | ID: mdl-32524478

ABSTRACT

We describe here an organotypic culture system we have used to investigate mechanisms that maintain structure and function of axon terminals at the neuromuscular junction (NMJ). We developed this by taking advantage of the slow Wallerian degeneration phenotype in mutant Wlds mice, using these to compare preservation of NMJs with degeneration in nerve-muscle preparations from wild-type mice. We take hind limb tibial nerve/flexor digitorum brevis and lumbrical muscles and incubate them in mammalian physiological saline at 32 °C for 24-48 h. Integrity of NMJs can then be compared using a combination of electrophysiological and morphological techniques. We illustrate our method with data showing synaptic preservation ex vivo in nerve-muscle explants from Sarm-1 null-mutant mice. The ex vivo assays of NMJ integrity we describe here may therefore be useful for detailed investigation of synaptic maintenance and degeneration.


Subject(s)
Neuromuscular Junction/physiology , Organ Culture Techniques/methods , Wallerian Degeneration/physiopathology , Animals , Armadillo Domain Proteins/deficiency , Axons/physiology , Cytoskeletal Proteins/deficiency , Dissection/methods , Electrophysiology/methods , Female , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Muscle, Skeletal , Neuromuscular Junction/ultrastructure , Organ Culture Techniques/instrumentation , Synapses/ultrastructure , Tibial Nerve
11.
Methods Mol Biol ; 2143: 311-320, 2020.
Article in English | MEDLINE | ID: mdl-32524490

ABSTRACT

The fruit fly Drosophila melanogaster has been a powerful model to study axonal biology including axon degeneration and regeneration (Brace et al., J Neurosci 34:8398-8410, 2014; Valakh et al. J Neurosci 33:17863-17,873, 2013; Xiong and Collins J Neurosci 32:610-615, 2012; Xiong et al. 191:211-223, 2010). Both adult and larval injury models have been developed in the fruit fly. This chapter focuses on in vivo and ex vivo methods developed for studying axon degeneration in Drosophila larvae. Additional models have been developed in the adult fly including injury models of olfactory receptor neurons in the brain and a model of axonal degeneration of sensory axons in the wing (Fang and Bonini, Annu Rev. Cell Dev Biol 28:575-597, 2012; Hoopfer et al. Neuron 50:883-895, 2006; Neukomm et al. Proc Natl Acad Sci U S A 111:9965-9970, 2014).


Subject(s)
Axons/physiology , Disease Models, Animal , Drosophila melanogaster/drug effects , Nerve Degeneration , Animals , Axons/ultrastructure , Drosophila melanogaster/growth & development , Drosophila melanogaster/physiology , Fluorescent Antibody Technique, Indirect/methods , Genes, Reporter , Larva , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/physiopathology , Neuromuscular Junction/ultrastructure , Regeneration
12.
J Neurosci ; 40(14): 2817-2827, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32122953

ABSTRACT

Perturbations to postsynaptic glutamate receptors (GluRs) trigger retrograde signaling to precisely increase presynaptic neurotransmitter release, maintaining stable levels of synaptic strength, a process referred to as homeostatic regulation. However, the structural change of homeostatic regulation remains poorly defined. At wild-type Drosophila neuromuscular junction synapse, there is one Bruchpilot (Brp) ring detected by superresolution microscopy at active zones (AZs). In the present study, we report multiple Brp rings (i.e., multiple T-bars seen by electron microscopy) at AZs of both male and female larvae when GluRs are reduced. At GluRIIC-deficient neuromuscular junctions, quantal size was reduced but quantal content was increased, indicative of homeostatic presynaptic potentiation. Consistently, multiple Brp rings at AZs were observed in the two classic synaptic homeostasis models (i.e., GluRIIA mutant and pharmacological blockade of GluRIIA activity). Furthermore, postsynaptic overexpression of the cell adhesion protein Neuroligin 1 partially rescued multiple Brp rings phenotype. Our study thus supports that the formation of multiple Brp rings at AZs might be a structural basis for synaptic homeostasis.SIGNIFICANCE STATEMENT Synaptic homeostasis is a conserved fundamental mechanism to maintain efficient neurotransmission of neural networks. Active zones (AZs) are characterized by an electron-dense cytomatrix, which is largely composed of Bruchpilot (Brp) at the Drosophila neuromuscular junction synapses. It is not clear how the structure of AZs changes during homeostatic regulation. To address this question, we examined the structure of AZs by superresolution microscopy and electron microscopy during homeostatic regulation. Our results reveal multiple Brp rings at AZs of glutamate receptor-deficient neuromuscular junction synapses compared with single Brp ring at AZs in wild type (WT). We further show that Neuroligin 1-mediated retrograde signaling regulates multiple Brp ring formation at glutamate receptor-deficient synapses. This study thus reveals a regulatory mechanism for synaptic homeostasis.


Subject(s)
Homeostasis/physiology , Neuromuscular Junction/physiology , Neuromuscular Junction/ultrastructure , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission/physiology , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila , Drosophila Proteins/metabolism , Female , Male , Receptors, Glutamate/metabolism
13.
Muscle Nerve ; 60(6): 790-800, 2019 12.
Article in English | MEDLINE | ID: mdl-31531871

ABSTRACT

INTRODUCTION: Reduced expression of the vesicular acetylcholine transporter (VAChT) leads to changes in the distribution and shape of synaptic vesicles (SVs) at neuromuscular junctions (NMJs), suggesting vesicular acetylcholine (ACh) as a key component of synaptic structure and function. It is poorly understood how long-term changes in cholinergic transmission contribute to age- and disease-related degeneration in the motor system. METHODS: In this study we performed confocal imaging, electrophysiology, electron microscopy, and analyses of respiratory mechanics of the diaphragm NMJ components in 12-month-old wild-type (WT) and VAChTKDHOM mice. RESULTS: Diaphragms of NMJs of the VAChTKDHOM mice were similar to those in WT mice in number, colocalization, and fragmentation of pre-/postsynaptic components. However, they had increased spontaneous SV exocytosis, miniature endplate potential frequency, and diminished MEPP amplitude. No impairment in respiratory mechanics at rest was observed, probably due to the large neurotransmission safety factor of the diaphragm. DISCUSSION: The present findings help us to understand the consequences of reduced ACh release at the NMJs during aging.


Subject(s)
Aging/pathology , Diaphragm/ultrastructure , Myasthenic Syndromes, Congenital/pathology , Neuromuscular Junction/ultrastructure , Synaptic Vesicles/ultrastructure , Acetylcholine/metabolism , Aging/metabolism , Animals , Diaphragm/metabolism , Diaphragm/physiopathology , Disease Models, Animal , Endocytosis , Excitatory Postsynaptic Potentials/physiology , Exocytosis , Gene Knockdown Techniques , Mice , Microscopy, Confocal , Microscopy, Electron, Transmission , Motor Endplate , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/metabolism , Myasthenic Syndromes, Congenital/physiopathology , Neuromuscular Junction/metabolism , Neuromuscular Junction/physiopathology , Respiratory Mechanics/physiology , Synaptic Transmission , Synaptic Vesicles/metabolism , Vesicular Acetylcholine Transport Proteins/genetics
14.
Acta Neuropathol Commun ; 7(1): 147, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31514753

ABSTRACT

The coordinated movement of organisms relies on efficient nerve-muscle communication at the neuromuscular junction. After peripheral nerve injury or neurodegeneration, motor neurons and Schwann cells increase the expression of the p75NTR pan-neurotrophin receptor. Even though p75NTR targeting has emerged as a promising therapeutic strategy to delay peripheral neuronal damage progression, the effects of long-term p75NTR inhibition at the mature neuromuscular junction have not been elucidated. We performed quantitative neuroanathomical analyses of the neuromuscular junction in p75NTR null mice by laser confocal and electron microscopy, which were complemented with electromyography, locomotor tests, and pharmacological intervention studies. Mature neuromuscular synapses of p75NTR null mice show impaired postsynaptic organization and ultrastructural complexity, which correlate with altered synaptic function at the levels of nerve activity-induced muscle responses, muscle fiber structure, force production, and locomotor performance. Our results on primary myotubes and denervated muscles indicate that muscle-derived p75NTR does not play a major role on postsynaptic organization. In turn, motor axon terminals of p75NTR null mice display a strong reduction in the number of synaptic vesicles and active zones. According to the observed pre and postsynaptic defects, pharmacological acetylcholinesterase inhibition rescued nerve-dependent muscle response and force production in p75NTR null mice. Our findings revealing that p75NTR is required to organize mature neuromuscular junctions contribute to a comprehensive view of the possible effects caused by therapeutic attempts to target p75NTR.


Subject(s)
Motor Neurons/physiology , Neuromuscular Junction/physiology , Receptors, Nerve Growth Factor/physiology , Synaptic Vesicles/physiology , Animals , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Motor Neurons/ultrastructure , Neuromuscular Junction/ultrastructure , Receptors, Nerve Growth Factor/genetics , Synaptic Vesicles/ultrastructure
15.
JCI Insight ; 4(18)2019 09 19.
Article in English | MEDLINE | ID: mdl-31534050

ABSTRACT

The control of voluntary skeletal muscle contraction relies on action potentials, which send signals from the motor neuron through the neuromuscular junction (NMJ). Although dysfunction of the NMJ causes various neuromuscular diseases, a reliable in vitro system for disease modeling is currently unavailable. Here, we present a potentially novel 2-step, self-organizing approach for generating in vitro human NMJs from human induced pluripotent stem cells. Our simple and robust approach results in a complex NMJ structure that includes functional connectivity, recapitulating in vivo synapse formation. We used these in vitro NMJs to model the pathological features of spinal muscular atrophy, revealing the developmental and functional defects of NMJ formation and NMJ-dependent muscular contraction. Our differentiation system is therefore useful for investigating and understanding the physiology and pathology of human NMJs.


Subject(s)
Motor Neurons/pathology , Muscle Contraction/physiology , Muscular Atrophy, Spinal/pathology , Neuromuscular Junction/pathology , Survival of Motor Neuron 1 Protein/genetics , Cell Differentiation , Cell Line , Gene Knockdown Techniques , Humans , Induced Pluripotent Stem Cells/physiology , Microscopy, Electron , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/physiopathology , Neuromuscular Junction/genetics , Neuromuscular Junction/growth & development , Neuromuscular Junction/ultrastructure , Optogenetics , Proof of Concept Study
16.
eNeuro ; 6(4)2019.
Article in English | MEDLINE | ID: mdl-31387877

ABSTRACT

Synapse formation can be promoted by intense activity. At the Drosophila larval neuromuscular junction (NMJ), new synaptic boutons can grow acutely in response to patterned stimulation. We combined confocal imaging with electron microscopy and tomography to investigate the initial stages of growth and differentiation of new presynaptic boutons at the Drosophila NMJ. We found that the new boutons can form rapidly in intact larva in response to intense crawling activity, and we observed two different patterns of bouton formation and maturation. The first pathway involves the growth of filopodia followed by a formation of boutons that are initially devoid of synaptic vesicles (SVs) but filled with filamentous matrix. The second pathway involves rapid budding of synaptic boutons packed with SVs, and these more mature boutons are sometimes capable of exocytosis/endocytosis. We demonstrated that intense activity predominantly promotes the second pathway, i.e., budding of more mature boutons filled with SVs. We also showed that this pathway depends on synapsin (Syn), a neuronal protein which reversibly associates with SVs and mediates their clustering via a protein kinase A (PKA)-dependent mechanism. Finally, we took advantage of the temperature-sensitive mutant sei to demonstrate that seizure activity can promote very rapid budding of new boutons filled with SVs, and this process occurs at scale of minutes. Altogether, these results demonstrate that intense activity acutely and selectively promotes rapid budding of new relatively mature presynaptic boutons filled with SVs, and that this process is regulated via a PKA/Syn-dependent pathway.


Subject(s)
Locomotion , Neuromuscular Junction/physiology , Presynaptic Terminals/physiology , Synaptic Vesicles/physiology , Animals , Animals, Genetically Modified , Cell Differentiation , Cyclic AMP-Dependent Protein Kinases/physiology , Drosophila , Drosophila Proteins/physiology , Female , Male , Neuromuscular Junction/cytology , Neuromuscular Junction/growth & development , Neuromuscular Junction/ultrastructure , Presynaptic Terminals/ultrastructure , Synapsins/physiology
17.
Cells ; 8(7)2019 07 02.
Article in English | MEDLINE | ID: mdl-31269763

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease of the neuromuscular junction (NMJ). Autoantibodies target key molecules at the NMJ, such as the nicotinic acetylcholine receptor (AChR), muscle-specific kinase (MuSK), and low-density lipoprotein receptor-related protein 4 (Lrp4), that lead by a range of different pathogenic mechanisms to altered tissue architecture and reduced densities or functionality of AChRs, reduced neuromuscular transmission, and therefore a severe fatigable skeletal muscle weakness. In this review, we give an overview of the history and clinical aspects of MG, with a focus on the structure and function of myasthenic autoantigens at the NMJ and how they are affected by the autoantibodies' pathogenic mechanisms. Furthermore, we give a short overview of the cells that are implicated in the production of the autoantibodies and briefly discuss diagnostic challenges and treatment strategies.


Subject(s)
Autoantibodies/immunology , Autoantigens/immunology , Muscle, Skeletal/pathology , Myasthenia Gravis/immunology , Neuromuscular Junction/pathology , Agrin/immunology , Agrin/metabolism , Animals , Autoantigens/metabolism , Humans , LDL-Receptor Related Proteins/immunology , LDL-Receptor Related Proteins/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/ultrastructure , Myasthenia Gravis/pathology , Neuromuscular Junction/immunology , Neuromuscular Junction/ultrastructure , Receptor Protein-Tyrosine Kinases/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cholinergic/immunology , Receptors, Cholinergic/metabolism , Receptors, Nicotinic/immunology , Receptors, Nicotinic/metabolism
18.
Neuromuscul Disord ; 29(7): 533-542, 2019 07.
Article in English | MEDLINE | ID: mdl-31230871

ABSTRACT

Acetylcholine receptor (AChR) clustering on the surface of muscle cells is a hallmark of postsynaptic differentiation at the vertebrate neuromuscular junction (NMJ). Even though the assembly of complex postsynaptic apparatuses is known to rely on both, pre- and postsynaptic signals, the identity of muscle-derived proteins modulating postsynaptic assembly and maintenance is still to be fully elucidated. Efficient gene transfer into muscle cells represents a powerful tool to analyze the contribution of muscle proteins on postsynaptic assembly and maintenance. Here, we describe a protocol that combines efficient electroporation of primary muscle satellite cells with the formation of aneural complex postsynaptic structures on the surface of myotubes. In vitro formed postsynaptic structures share various similarities with in vivo postsynaptic NMJ domains. While primary myotubes express increasing amounts of the ε AChR subunit, associated with NMJ maturation, surface AChR aggregates lack this AChR subunit. Our results also validate the functional expression of a luciferase reporter gene, as well as the response of complex postsynaptic structures to pharmacological treatment. Together, these methods in primary muscle cells are a valuable tool to perform a detailed and accurate analysis of the potential role of muscle-derived proteins on the maintenance of complex postsynaptic structures and to identify nerve-derived signals regulating functional NMJ maturation.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Gene Transfer Techniques , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/innervation , Muscle, Skeletal/physiology , Animals , Cell Differentiation/genetics , Cell Survival , DNA/genetics , Electroporation , Myoblasts , Neuromuscular Junction/physiology , Neuromuscular Junction/ultrastructure , Primary Cell Culture , Rats , Receptors, Cholinergic/metabolism , Satellite Cells, Skeletal Muscle
19.
Int J Mol Sci ; 20(11)2019 May 31.
Article in English | MEDLINE | ID: mdl-31159267

ABSTRACT

Synaptic vesicles dock on the presynaptic plasma membrane of axon terminals and become ready to fuse with the presynaptic membrane or primed. Fusion of the vesicle membrane and presynaptic membrane results in the formation of a pore between the membranes, through which the vesicle's neurotransmitter is released into the synaptic cleft. A recent electron tomography study on frog neuromuscular junctions fixed at rest showed that there is no discernible gap between or merging of the membrane of docked synaptic vesicles with the presynaptic membrane, however, the extent of the contact area between the membrane of docked synaptic vesicles and the presynaptic membrane varies 10-fold with a normal distribution. The study also showed that when the neuromuscular junctions are fixed during repetitive electrical nerve stimulation, the portion of large contact areas in the distribution is reduced compared to the portion of small contact areas, suggesting that docked synaptic vesicles with the largest contact areas are greatly primed to fuse with the membrane. Furthermore, the finding of several hemifused synaptic vesicles among the docked vesicles was briefly reported. Here, the spatial relationship of 81 synaptic vesicles with the presynaptic membrane at active zones of the neuromuscular junctions fixed during stimulation is described in detail. For the most of the vesicles, the combined thickness of each of their contact sites was not different from the sum of the membrane thicknesses of the vesicle membrane and presynaptic membrane, similar to the docked vesicles at active zones of the resting neuromuscular junctions. However, the combined membrane thickness of a small portion of the vesicles was considerably less than the sum of the membrane thicknesses, indicating that the membranes at their contact sites were fixed in a state of hemifusion. Moreover, the hemifused vesicles were found to have large contact areas with the presynaptic membrane. These findings support the recently proposed hypothesis that, at frog neuromuscular junctions, docked synaptic vesicles with the largest contact areas are most primed for fusion with the presynaptic membrane, and that hemifusion is a fusion intermediate step of the vesicle membrane with the presynaptic membrane for synaptic transmission.


Subject(s)
Neuromuscular Junction/metabolism , Synaptic Membranes/metabolism , Synaptic Transmission , Synaptic Vesicles/metabolism , Animals , Anura , Models, Biological , Neuromuscular Junction/ultrastructure , Synaptic Membranes/ultrastructure , Synaptic Vesicles/ultrastructure
20.
Muscle Nerve ; 59(5): 611-618, 2019 05.
Article in English | MEDLINE | ID: mdl-30677149

ABSTRACT

INTRODUCTION: In motor neurons, cholera toxin B (CTB) binds to the cell-surface ganglioside GM1 and is internalized and transported via structurally unique components of plasma membranes (lipid rafts). METHODS: Lipid raft uptake by axon terminals adjoining type-identified rat diaphragm muscle fibers was investigated using CTB and confocal imaging. RESULTS: Lipid raft uptake increased significantly at higher frequency stimulation (80 Hz), compared with lower frequency (20 Hz) and unstimulated (0 Hz) conditions. The fraction of axon terminal occupied by CTB was ∼45% at 0- or 20-Hz stimulation, and increased to ∼65% at 80 Hz. Total CTB fluorescence intensity also increased (∼20%) after 80-Hz stimulation compared with 0 Hz. DISCUSSION: Evidence of increased lipid raft uptake at high stimulation frequencies supports an important role for lipid raft signaling at rat diaphragm muscle axon terminals, primarily for motor units physiologically activated at the higher frequencies. Muscle Nerve 59:611-611, 2019.


Subject(s)
Cholera Toxin/metabolism , Diaphragm/innervation , Membrane Microdomains/metabolism , Neuromuscular Junction/metabolism , Phrenic Nerve/metabolism , Presynaptic Terminals/metabolism , Animals , Electric Stimulation , Membrane Microdomains/ultrastructure , Microscopy, Confocal , Motor Neurons/metabolism , Neuromuscular Junction/ultrastructure , Phrenic Nerve/cytology , Phrenic Nerve/ultrastructure , Presynaptic Terminals/ultrastructure , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...