Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 651
Filter
1.
Cells ; 11(21)2022 10 24.
Article in English | MEDLINE | ID: mdl-36359747

ABSTRACT

Hereditary spastic paraplegia (HSP) is a heterogeneous group of genetic neurodegenerative disorders, characterized by progressive lower limb spasticity and weakness resulting from retrograde axonal degeneration of motor neurons (MNs). Here, we generated in vitro human neuromuscular junctions (NMJs) from five HSP patient-specific induced pluripotent stem cell (hiPSC) lines, by means of microfluidic strategy, to model disease-relevant neuropathologic processes. The strength of our NMJ model lies in the generation of lower MNs and myotubes from autologous hiPSC origin, maintaining the genetic background of the HSP patient donors in both cell types and in the cellular organization due to the microfluidic devices. Three patients characterized by a mutation in the SPG3a gene, encoding the ATLASTIN GTPase 1 protein, and two patients with a mutation in the SPG4 gene, encoding the SPASTIN protein, were included in this study. Differentiation of the HSP-derived lines gave rise to lower MNs that could recapitulate pathological hallmarks, such as axonal swellings with accumulation of Acetyl-α-TUBULIN and reduction of SPASTIN levels. Furthermore, NMJs from HSP-derived lines were lower in number and in contact point complexity, denoting an impaired NMJ profile, also confirmed by some alterations in genes encoding for proteins associated with microtubules and responsible for axonal transport. Considering the complexity of HSP, these patient-derived neuronal and skeletal muscle cell co-cultures offer unique tools to study the pathologic mechanisms and explore novel treatment options for rescuing axonal defects and diverse cellular processes, including membrane trafficking, intracellular motility and protein degradation in HSP.


Subject(s)
Induced Pluripotent Stem Cells , Neuromuscular Junction , Spastic Paraplegia, Hereditary , Humans , Adenosine Triphosphatases/metabolism , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/pathology , Neuromuscular Junction/cytology , Neuromuscular Junction/pathology , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/pathology , Spastin/metabolism
2.
Cells ; 10(12)2021 11 24.
Article in English | MEDLINE | ID: mdl-34943800

ABSTRACT

Motoneurons, skeletal muscle fibers, and Schwann cells form synapses, termed neuromuscular junctions (NMJs). These control voluntary body movement and are affected in numerous neuromuscular diseases. Therefore, a variety of NMJ in vitro models have been explored to enable mechanistic and pharmacological studies. So far, selective integration of Schwann cells in these models has been hampered, due to technical limitations. Here we present robust protocols for derivation of Schwann cells from human induced pluripotent stem cells (hiPSC) and their coculture with hiPSC-derived motoneurons and C2C12 muscle cells. Upon differentiation with tuned BMP signaling, Schwann cells expressed marker proteins, S100b, Gap43, vimentin, and myelin protein zero. Furthermore, they displayed typical spindle-shaped morphologies with long processes, which often aligned with motoneuron axons. Inclusion of Schwann cells in coculture experiments with hiPSC-derived motoneurons and C2C12 myoblasts enhanced myotube growth and affected size and number of acetylcholine receptor plaques on myotubes. Altogether, these data argue for the availability of a consistent differentiation protocol for Schwann cells and their amenability for functional integration into neuromuscular in vitro models, fostering future studies of neuromuscular mechanisms and disease.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Muscle Development , Neuromuscular Junction/cytology , Schwann Cells/cytology , Animals , Biomarkers/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Line , Cell Shape , Coculture Techniques , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Receptors, Cholinergic/metabolism , Signal Transduction
3.
Muscle Nerve ; 64(4): 388-403, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34328673

ABSTRACT

Recent development of novel therapies has improved mobility and quality of life for people suffering from inheritable neuromuscular disorders. Despite this progress, the majority of neuromuscular disorders are still incurable, in part due to a lack of predictive models of neuromuscular junction (NMJ) breakdown. Improvement of predictive models of a human NMJ would be transformative in terms of expanding our understanding of the mechanisms that underpin development, maintenance, and disease, and as a testbed with which to evaluate novel therapeutics. Induced pluripotent stem cells (iPSCs) are emerging as a clinically relevant and non-invasive cell source to create human NMJs to study synaptic development and maturation, as well as disease modeling and drug discovery. This review will highlight the recent advances and remaining challenges to generating an NMJ capable of eliciting contraction of stem cell-derived skeletal muscle in vitro. We explore the advantages and shortcomings of traditional NMJ culturing platforms, as well as the pioneering technologies and novel, biomimetic culturing systems currently in use to guide development and maturation of the neuromuscular synapse and extracellular microenvironment. Then, we will explore how this NMJ-in-a-dish can be used to study normal assembly and function of the efferent portion of the neuromuscular arc, and how neuromuscular disease-causing mutations disrupt structure, signaling, and function.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Neuromuscular Junction/physiology , Tissue Engineering/methods , Animals , Humans , Lab-On-A-Chip Devices , Motor Neurons/physiology , Neuromuscular Junction/cytology , Stem Cells/physiology
4.
JCI Insight ; 6(7)2021 04 08.
Article in English | MEDLINE | ID: mdl-33661767

ABSTRACT

Here, we report on the identification of Itga7-expressing muscle-resident glial cells activated by loss of neuromuscular junction (NMJ) integrity. Gene expression analysis at the bulk and single-cell level revealed that these cells are distinct from Itga7-expressing muscle satellite cells. We show that a selective activation and expansion of Itga7+ glial cells occur in response to muscle nerve lesion. Upon activation, muscle glial-derived progenies expressed neurotrophic genes, including nerve growth factor receptor, which enables their isolation by FACS. We show that activated muscle glial cells also expressed genes potentially implicated in extracellular matrix remodeling at NMJs. We found that tenascin C, which was highly expressed by muscle glial cells, activated upon nerve injury and preferentially localized to NMJ. Interestingly, we observed that the activation of muscle glial cells by acute nerve injury was reversible upon NMJ repair. By contrast, in a mouse model of ALS, in which NMJ degeneration is progressive, muscle glial cells steadily increased over the course of the disease. However, they exhibited an impaired neurotrophic activity, suggesting that pathogenic activation of glial cells may be implicated in ALS progression.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Muscle, Skeletal/cytology , Neuroglia/physiology , Spinal Cord Injuries/pathology , Animals , Antigens, CD/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Integrin alpha Chains/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Neuroglia/cytology , Neuromuscular Junction/cytology , Receptor, Nerve Growth Factor/genetics , Receptors, Cholinergic/metabolism , Sciatic Nerve/injuries , Single-Cell Analysis , Superoxide Dismutase-1/genetics
5.
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
6.
Proc Natl Acad Sci U S A ; 117(47): 29914-29924, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33168737

ABSTRACT

Neuropeptides are important for regulating numerous neural functions and behaviors. Release of neuropeptides requires long-lasting, high levels of cytosolic Ca2+ However, the molecular regulation of neuropeptide release remains to be clarified. Recently, Stac3 was identified as a key regulator of L-type Ca2+ channels (CaChs) and excitation-contraction coupling in vertebrate skeletal muscles. There is a small family of stac genes in vertebrates with other members expressed by subsets of neurons in the central nervous system. The function of neural Stac proteins, however, is poorly understood. Drosophila melanogaster contain a single stac gene, Dstac, which is expressed by muscles and a subset of neurons, including neuropeptide-expressing motor neurons. Here, genetic manipulations, coupled with immunolabeling, Ca2+ imaging, electrophysiology, and behavioral analysis, revealed that Dstac regulates L-type CaChs (Dmca1D) in Drosophila motor neurons and this, in turn, controls the release of neuropeptides.


Subject(s)
Calcium Channels/metabolism , Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Motor Neurons/metabolism , Neuromuscular Junction/metabolism , Neuropeptides/metabolism , Animals , Animals, Genetically Modified , Behavior Observation Techniques , Behavior, Animal , Drosophila melanogaster , Female , Intravital Microscopy , Larva , Male , Models, Animal , Motor Neurons/cytology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Neuromuscular Junction/cytology , Optical Imaging , Patch-Clamp Techniques , Presynaptic Terminals/metabolism
7.
Nat Commun ; 11(1): 5102, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037211

ABSTRACT

Skeletal muscle fibers are large syncytia but it is currently unknown whether gene expression is coordinately regulated in their numerous nuclei. Here we show by snRNA-seq and snATAC-seq that slow, fast, myotendinous and neuromuscular junction myonuclei each have different transcriptional programs, associated with distinct chromatin states and combinations of transcription factors. In adult mice, identified myofiber types predominantly express either a slow or one of the three fast isoforms of Myosin heavy chain (MYH) proteins, while a small number of hybrid fibers can express more than one MYH. By snRNA-seq and FISH, we show that the majority of myonuclei within a myofiber are synchronized, coordinately expressing only one fast Myh isoform with a preferential panel of muscle-specific genes. Importantly, this coordination of expression occurs early during post-natal development and depends on innervation. These findings highlight a previously undefined mechanism of coordination of gene expression in a syncytium.


Subject(s)
Cell Nucleus/genetics , Gene Expression Regulation , In Situ Hybridization, Fluorescence/methods , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , Sequence Analysis, RNA/methods , Animals , Female , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Myosin Heavy Chains/genetics , Neuromuscular Junction/cytology , Single-Cell Analysis , Tendons/cytology , Transcription, Genetic
8.
Stem Cell Reports ; 15(4): 941-954, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32888503

ABSTRACT

Satellite cells are main muscle stem cells that could provide myonuclei for myofiber growth and synaptic-specific gene expression during the early postnatal development. Here, we observed that splicing factor SRSF1 is highly expressed in myoblasts and its expression is closely related with satellite cell activation and proliferation. By genetic deletion of SRSF1 in myogenic progenitors, we found that SRSF1 is critical for satellite cell proliferation in vitro and in vivo. Most notably we also observed that SRSF1 is required for the functional neuromuscular junction (NMJ) formation, as SRSF1-deficient mice fail to form mature pretzel-like NMJs, which leads to muscle weakness and premature death in mice. Finally, we demonstrated that SRSF1 contributes to muscle innervation and muscle development likely by regulating a restricted set of tissue-specific alternative splicing events. Thus, our data define a unique role for SRSF1 in postnatal skeletal muscle growth and function in mice.


Subject(s)
Cell Differentiation , Neuromuscular Junction/cytology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Serine-Arginine Splicing Factors/metabolism , Alternative Splicing/genetics , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Proliferation , Gene Deletion , Mice, Inbred C57BL , Mice, Knockout , Muscular Atrophy/pathology , Serine-Arginine Splicing Factors/deficiency
9.
Neurobiol Aging ; 95: 1-8, 2020 11.
Article in English | MEDLINE | ID: mdl-32739557

ABSTRACT

The neuromuscular junction (NMJ) connects the motor neuron with myofibers allowing muscle contraction. Both aging and increased activity result in NMJ remodeling. Here, the effects of exercise were examined in young and aged soleus muscles. Using immunofluorescent staining procedures, cellular and active zone components of the NMJ were quantified following a treadmill running program. Immunofluorescence was employed to determine myofiber profiles (size and type). Two-way analysis of variance procedures with main effects of age and treatment showed that when analyzing NMJs at the cellular level, significant (p ≤ 0.05) effects were identified for age, but not treatment. However, when examining subcellular active zones, effects for exercise, but not for age, were detected. Myofiber cross-sectional area showed that aging elicited atrophy and that among younger muscles endurance exercise training yielded decrements in myofiber size. Conversely, among aged muscles training elicited whole muscle and myofiber trends (p < 0.10) toward hypertrophy. Thus, different components of the neuromuscular system harbor unique sensitivities to various stimuli enabling proper adaptations to attain optimal function under differing conditions.


Subject(s)
Aging/physiology , Muscle, Skeletal/pathology , Neuromuscular Junction/cytology , Neuromuscular Junction/physiology , Physical Conditioning, Animal/physiology , Adaptation, Physiological , Aging/pathology , Animals , Atrophy , Hypertrophy , Male , Muscle, Skeletal/cytology , Myofibrils/pathology , Neuronal Plasticity/physiology , Rats, Inbred F344
10.
J Neurosci ; 40(33): 6270-6288, 2020 08 12.
Article in English | MEDLINE | ID: mdl-32631939

ABSTRACT

Structural and functional plasticity induced by neuronal competition is a common feature of developing nervous systems. However, the rules governing how postsynaptic cells differentiate between presynaptic inputs are unclear. In this study, we characterized synaptic interactions following manipulations of tonic Ib or phasic Is glutamatergic motoneurons that coinnervate postsynaptic muscles of male or female Drosophila melanogaster larvae. After identifying drivers for each neuronal subtype, we performed ablation or genetic manipulations to alter neuronal activity and examined the effects on synaptic innervation and function at neuromuscular junctions. Ablation of either Ib or Is resulted in decreased muscle response, with some functional compensation occurring in the Ib input when Is was missing. In contrast, the Is terminal failed to show functional or structural changes following loss of the coinnervating Ib input. Decreasing the activity of the Ib or Is neuron with tetanus toxin light chain resulted in structural changes in muscle innervation. Decreased Ib activity resulted in reduced active zone (AZ) number and decreased postsynaptic subsynaptic reticulum volume, with the emergence of filopodial-like protrusions from synaptic boutons of the Ib input. Decreased Is activity did not induce structural changes at its own synapses, but the coinnervating Ib motoneuron increased the number of synaptic boutons and AZs it formed. These findings indicate that tonic Ib and phasic Is motoneurons respond independently to changes in activity, with either functional or structural alterations in the Ib neuron occurring following ablation or reduced activity of the coinnervating Is input, respectively.SIGNIFICANCE STATEMENT Both invertebrate and vertebrate nervous systems display synaptic plasticity in response to behavioral experiences, indicating that underlying mechanisms emerged early in evolution. How specific neuronal classes innervating the same postsynaptic target display distinct types of plasticity is unclear. Here, we examined whether Drosophila tonic Ib and phasic Is motoneurons display competitive or cooperative interactions during innervation of the same muscle, or compensatory changes when the output of one motoneuron is altered. We established a system to differentially manipulate the motoneurons and examined the effects of cell type-specific changes to one of the inputs. Our findings indicate Ib and Is motoneurons respond differently to activity mismatch or loss of the coinnervating input, with the Ib subclass responding robustly compared with Is motoneurons.


Subject(s)
Motor Neurons/cytology , Motor Neurons/physiology , Neuromuscular Junction/cytology , Neuromuscular Junction/physiology , Neuronal Plasticity , Synapses/physiology , Animals , Drosophila melanogaster , Female , Glutamic Acid/physiology , Male , Membrane Potentials , Presynaptic Terminals/physiology
11.
J Cell Biol ; 219(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32259198

ABSTRACT

Dscam2 is a cell surface protein required for neuronal development in Drosophila; it can promote neural wiring through homophilic recognition that leads to either adhesion or repulsion between neurites. Here, we report that Dscam2 also plays a post-developmental role in suppressing synaptic strength. This function is dependent on one of two distinct extracellular isoforms of the protein and is autonomous to motor neurons. We link the PI3K enhancer, Centaurin gamma 1A, to the Dscam2-dependent regulation of synaptic strength and show that changes in phosphoinositide levels correlate with changes in endosomal compartments that have previously been associated with synaptic strength. Using transmission electron microscopy, we find an increase in synaptic vesicles at Dscam2 mutant active zones, providing a rationale for the increase in synaptic strength. Our study provides the first evidence that Dscam2 can regulate synaptic physiology and highlights how diverse roles of alternative protein isoforms can contribute to unique aspects of brain development and function.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Endosomes/metabolism , GTPase-Activating Proteins/metabolism , Larva/growth & development , Motor Neurons/metabolism , Neural Cell Adhesion Molecules/metabolism , Neurogenesis/genetics , Phosphatidylinositol 3-Kinases/metabolism , Animals , Animals, Genetically Modified , Drosophila/growth & development , Drosophila Proteins/genetics , Endosomes/genetics , Endosomes/ultrastructure , Immunohistochemistry , Larva/genetics , Larva/physiology , Larva/ultrastructure , Microscopy, Electron, Transmission , Motor Neurons/physiology , Mutation , Neural Cell Adhesion Molecules/genetics , Neuromuscular Junction/cytology , Neuromuscular Junction/genetics , Peripheral Nervous System/metabolism , Phosphatidylinositols/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Protein Isoforms/metabolism , Synaptic Transmission/genetics , Synaptic Transmission/physiology
12.
Semin Cell Dev Biol ; 104: 31-38, 2020 08.
Article in English | MEDLINE | ID: mdl-32147379

ABSTRACT

Neuromuscular junctions (NMJs) are specialized synapses in the peripheral nervous system that allow the transmission of neuronal impulses to skeletal muscles for their contraction. Due to its size and accessibility, the NMJ is a commonly used model for studying basic principles of synapse organization and function. Similar to synapses in the central nervous system, NMJs are composed of presynaptic axonal terminals, the postsynaptic machinery formed at the membrane of the muscle fibers, and the synapse-associated glial cells. The special glial cells at the NMJs are called terminal Schwann cells or perisynaptic Schwann cells (PSCs). Decades of studies on the NMJ, as well as the most recent discoveries, have revealed multiple functions for PSCs at different stages of synaptic formation, maintenance, and disassembly. This review summarizes major observations in the field.


Subject(s)
Neuromuscular Junction/metabolism , Presynaptic Terminals/metabolism , Schwann Cells/metabolism , Animals , Models, Biological , Neuromuscular Junction/cytology , Schwann Cells/cytology
13.
Nat Commun ; 11(1): 1025, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32094341

ABSTRACT

A bioengineered skeletal muscle construct that mimics structural and functional characteristics of native skeletal muscle is a promising therapeutic option to treat extensive muscle defect injuries. We previously showed that bioprinted human skeletal muscle constructs were able to form multi-layered bundles with aligned myofibers. In this study, we investigate the effects of neural cell integration into the bioprinted skeletal muscle construct to accelerate functional muscle regeneration in vivo. Neural input into this bioprinted skeletal muscle construct shows the improvement of myofiber formation, long-term survival, and neuromuscular junction formation in vitro. More importantly, the bioprinted constructs with neural cell integration facilitate rapid innervation and mature into organized muscle tissue that restores normal muscle weight and function in a rodent model of muscle defect injury. These results suggest that the 3D bioprinted human neural-skeletal muscle constructs can be rapidly integrated with the host neural network, resulting in accelerated muscle function restoration.


Subject(s)
Bioprinting/methods , Guided Tissue Regeneration/methods , Muscular Diseases/therapy , Myoblasts, Skeletal/physiology , Neurons/physiology , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/therapeutic use , Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Disease Models, Animal , Feasibility Studies , Humans , Hydrogels/chemistry , Male , Muscle, Skeletal/cytology , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Muscular Diseases/physiopathology , Nerve Net/physiology , Neuromuscular Junction/cytology , Neuromuscular Junction/physiology , Printing, Three-Dimensional , Rats , Time Factors
14.
Nat Commun ; 11(1): 1092, 2020 02 27.
Article in English | MEDLINE | ID: mdl-32107390

ABSTRACT

Micro(mi)RNA-based post-transcriptional regulatory mechanisms have been broadly implicated in the assembly and modulation of synaptic connections required to shape neural circuits, however, relatively few specific miRNAs have been identified that control synapse formation. Using a conditional transgenic toolkit for competitive inhibition of miRNA function in Drosophila, we performed an unbiased screen for novel regulators of synapse morphogenesis at the larval neuromuscular junction (NMJ). From a set of ten new validated regulators of NMJ growth, we discovered that miR-34 mutants display synaptic phenotypes and cell type-specific functions suggesting distinct downstream mechanisms in the presynaptic and postsynaptic compartments. A search for conserved downstream targets for miR-34 identified the junctional receptor CNTNAP4/Neurexin-IV (Nrx-IV) and the membrane cytoskeletal effector Adducin/Hu-li tai shao (Hts) as proteins whose synaptic expression is restricted by miR-34. Manipulation of miR-34, Nrx-IV or Hts-M function in motor neurons or muscle supports a model where presynaptic miR-34 inhibits Nrx-IV to influence active zone formation, whereas, postsynaptic miR-34 inhibits Hts to regulate the initiation of bouton formation from presynaptic terminals.


Subject(s)
Calmodulin-Binding Proteins/genetics , Cell Adhesion Molecules, Neuronal/genetics , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Presynaptic Terminals/physiology , Animals , Animals, Genetically Modified , Calmodulin-Binding Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Larva/growth & development , Morphogenesis/genetics , Mutation , Neuromuscular Junction/cytology , Neuromuscular Junction/growth & development
15.
J Biosci Bioeng ; 129(4): 486-493, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31678066

ABSTRACT

Motor neurons differentiated from induced pluripotent stem (iPS) cells have attracted attention for use in the construction of drug screening systems for neuronal diseases, such as amyotrophic lateral sclerosis. However, conventional drug screening systems using 2-dimensional (2D) cultures of iPS cell-derived motor neurons often evaluate the cell survival rate, morphological changes in the cells and/or gene expression analysis, and these parameters do not always reflect the actual functions of motor neurons, i.e., the induction of muscle contractions. In the present study, we developed a neuromuscular junction model comprising motor neurons and myotubes, which were differentiated from iPS cells and C2C12 myoblasts, respectively. Using this model, the contractile activity and force generation of the myotubes via the neuromuscular junction were successfully measured in both two- and three-dimensional (3D) cell culture systems. The results suggested that this neuromuscular junction model can be used to construct a drug candidate screening system for neuronal diseases.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Models, Biological , Motor Neurons/cytology , Motor Neurons/physiology , Muscle Fibers, Skeletal/cytology , Neuromuscular Junction/physiology , Tissue Engineering/methods , Animals , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Coculture Techniques/methods , Embryo, Mammalian , Female , Mice , Mice, Inbred BALB C , Muscle Contraction , Neuromuscular Junction/cytology , Pregnancy , Tissue Scaffolds
16.
Wiley Interdiscip Rev Syst Biol Med ; 12(1): e1462, 2020 01.
Article in English | MEDLINE | ID: mdl-31407867

ABSTRACT

Decades of research in skeletal muscle physiology have provided multiscale insights into the structural and functional complexity of this important anatomical tissue, designed to accomplish the task of generating contraction, force and movement. Skeletal muscle can be viewed as a biomechanical device with various interacting components including the autonomic nerves for impulse transmission, vasculature for efficient oxygenation, and embedded regulatory and metabolic machinery for maintaining cellular homeostasis. The "omics" revolution has propelled a new era in muscle research, allowing us to discern minute details of molecular cross-talk required for effective coordination between the myriad interacting components for efficient muscle function. The objective of this review is to provide a systems-level, comprehensive mapping the molecular mechanisms underlying skeletal muscle structure and function, in health and disease. We begin this review with a focus on molecular mechanisms underlying muscle tissue development (myogenesis), with an emphasis on satellite cells and muscle regeneration. We next review the molecular structure and mechanisms underlying the many structural components of the muscle: neuromuscular junction, sarcomere, cytoskeleton, extracellular matrix, and vasculature surrounding muscle. We highlight aberrant molecular mechanisms and their possible clinical or pathophysiological relevance. We particularly emphasize the impact of environmental stressors (inflammation and oxidative stress) in contributing to muscle pathophysiology including atrophy, hypertrophy, and fibrosis. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Developmental Biology > Developmental Processes in Health and Disease Models of Systems Properties and Processes > Cellular Models.


Subject(s)
Models, Biological , Muscle, Skeletal , Muscular Diseases , Animals , Biophysical Phenomena , Extracellular Matrix/physiology , Humans , Muscle Contraction/physiology , Muscle, Skeletal/anatomy & histology , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/physiology , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Neuromuscular Junction/cytology , Neuromuscular Junction/physiology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/physiology , Synapses/metabolism , Synapses/physiology
17.
Neurosci Lett ; 715: 134644, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31765730

ABSTRACT

Super-resolution microscopy techniques offer subdiffraction limited resolution that is two- to ten-fold improved compared to that offered by conventional confocal microscopy. This breakthrough in resolution for light microscopy has contributed to new findings in neuroscience and synapse biology. This review will focus on the Structured Illumination Microscopy (SIM), Stimulated emission depletion (STED) microscopy, and Stochastic optical reconstruction microscopy (STORM) / Single molecule localization microscopy (SMLM) techniques and compare them for the better understanding of their differences and their suitability for the analysis of synapse biology. In addition, we will discuss a few practical aspects of these microscopic techniques, including resolution, image acquisition speed, multicolor capability, and other advantages and disadvantages. Tips for the improvement of microscopy will be introduced; for example, information resources for recommended dyes, the limitations of multicolor analysis, and capabilities for live imaging. In addition, we will summarize how super-resolution microscopy has been used for analyses of neuromuscular junctions and synapses.


Subject(s)
Microscopy, Fluorescence/methods , Neuromuscular Junction/cytology , Synapses , Animals , Humans
18.
Nat Commun ; 10(1): 5575, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31811118

ABSTRACT

Synapses are highly specialized for neurotransmitter signaling, yet activity-dependent growth factor release also plays critical roles at synapses. While efficient neurotransmitter signaling relies on precise apposition of release sites and neurotransmitter receptors, molecular mechanisms enabling high-fidelity growth factor signaling within the synaptic microenvironment remain obscure. Here we show that the auxiliary calcium channel subunit α2δ-3 promotes the function of an activity-dependent autocrine Bone Morphogenetic Protein (BMP) signaling pathway at the Drosophila neuromuscular junction (NMJ). α2δ proteins have conserved synaptogenic activity, although how they execute this function has remained elusive. We find that α2δ-3 provides an extracellular scaffold for an autocrine BMP signal, suggesting a mechanistic framework for understanding α2δ's conserved role in synapse organization. We further establish a transcriptional requirement for activity-dependent, autocrine BMP signaling in determining synapse density, structure, and function. We propose that activity-dependent, autocrine signals provide neurons with continuous feedback on their activity state for modulating both synapse structure and function.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Calcium Channels, L-Type/metabolism , Drosophila melanogaster/metabolism , Neuromuscular Junction/metabolism , Signal Transduction/physiology , Synapses/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Calcium/metabolism , Calcium Channels, L-Type/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/metabolism , Male , Neurogenesis/genetics , Neurogenesis/physiology , Neuromuscular Junction/cytology , Phenotype , Synapses/genetics , Synaptic Transmission/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
Sci Rep ; 9(1): 18450, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804576

ABSTRACT

Proper formation of neuromuscular synapses requires the reciprocal communication between motor neurons and muscle cells. Several anterograde and retrograde signals involved in neuromuscular junction formation are known. However the postsynaptic mechanisms regulating presynaptic differentiation are still incompletely understood. Here we report that the skeletal muscle calcium channel (CaV1.1) is required for motor nerve differentiation and that the mechanism by which CaV1.1 controls presynaptic differentiation utilizes activity-dependent calcium signaling in muscle. In mice lacking CaV1.1 or CaV1.1-driven calcium signaling motor nerves are ectopically located and aberrantly defasciculated. Axons fail to recognize their postsynaptic target structures and synaptic vesicles and active zones fail to correctly accumulate at the nerve terminals opposite AChR clusters. These presynaptic defects are independent of aberrant AChR patterning and more sensitive to deficient calcium signals. Thus, our results identify CaV1.1-driven calcium signaling in muscle as a major regulator coordinating multiple aspects of presynaptic differentiation at the neuromuscular synapse.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Motor Neurons/physiology , Muscle, Skeletal/innervation , Neuromuscular Junction/growth & development , Animals , Calcium/metabolism , Calcium Channels, L-Type/genetics , Cell Differentiation/physiology , Mice , Mice, Knockout , Models, Animal , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Neuromuscular Junction/cytology , Neuromuscular Junction/metabolism , Presynaptic Terminals/physiology
20.
Elife ; 82019 09 10.
Article in English | MEDLINE | ID: mdl-31502540

ABSTRACT

How circuits assemble starting from stem cells is a fundamental question in developmental neurobiology. We test the hypothesis that, in neuronal stem cells, temporal transcription factors predictably control neuronal terminal features and circuit assembly. Using the Drosophila motor system, we manipulate expression of the classic temporal transcription factor Hunchback (Hb) specifically in the NB7-1 stem cell, which produces U motor neurons (MNs), and then we monitor dendrite morphology and neuromuscular synaptic partnerships. We find that prolonged expression of Hb leads to transient specification of U MN identity, and that embryonic molecular markers do not accurately predict U MN terminal features. Nonetheless, our data show Hb acts as a potent regulator of neuromuscular wiring decisions. These data introduce important refinements to current models, show that molecular information acts early in neurogenesis as a switch to control motor circuit wiring, and provide novel insight into the relationship between stem cell and circuit.


Subject(s)
DNA-Binding Proteins/biosynthesis , Drosophila Proteins/biosynthesis , Gene Expression , Motor Neurons/physiology , Neural Pathways/embryology , Neuromuscular Junction/physiology , Stem Cells/physiology , Transcription Factors/biosynthesis , Animals , Drosophila , Motor Neurons/cytology , Neuromuscular Junction/cytology , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...