Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.169
Filter
1.
Cells ; 10(7)2021 06 28.
Article in English | MEDLINE | ID: mdl-34203264

ABSTRACT

Brachial plexus root avulsions cause debilitating upper limb paralysis. Short-term neuroprotective treatments have reported preservation of motor neurons and function in model animals while reports of long-term benefits of such treatments are scarce, especially the morphological sequelae. This morphological study investigated the long-term suppression of c-Jun- and neuronal nitric oxide synthase (nNOS) (neuroprotective treatments for one month) on the motor neuron survival, ultrastructural features of lower motor neurons, and forelimb function at six months after brachial plexus roots avulsion. Neuroprotective treatments reduced oxidative stress and preserved ventral horn motor neurons at the end of the 28-day treatment period relative to vehicle treated ones. Motor neuron sparing was associated with suppression of c-Jun, nNOS, and pro-apoptotic proteins Bim and caspases at this time point. Following 6 months of survival, neutral red staining revealed a significant loss of most of the motor neurons and ventral horn atrophy in the avulsed C6, 7, and 8 cervical segments among the vehicle-treated rats (n = 4). However, rats that received neuroprotective treatments c-Jun JNK inhibitor, SP600125 (n = 4) and a selective inhibitor of nNOS, 7-nitroindazole (n = 4), retained over half of their motor neurons in the ipsilateral avulsed side compared. Myelinated axons in the avulsed ventral horns of vehicle-treated rats were smaller but numerous compared to the intact contralateral ventral horns or neuroprotective-treated groups. In the neuroprotective treatment groups, there was the preservation of myelin thickness around large-caliber axons. Ultrastructural evaluation also confirmed the preservation of organelles including mitochondria and synapses in the two groups that received neuroprotective treatments compared with vehicle controls. Also, forelimb functional evaluation demonstrated that neuroprotective treatments improved functional abilities in the rats. In conclusion, neuroprotective treatments aimed at suppressing degenerative c-Jun and nNOS attenuated apoptosis, provided long-term preservation of motor neurons, their organelles, ventral horn size, and forelimb function.


Subject(s)
Brachial Plexus/physiopathology , Forelimb/physiopathology , Motor Neurons/metabolism , Motor Neurons/ultrastructure , Nitric Oxide Synthase Type I/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Radiculopathy/physiopathology , Spinal Nerve Roots/physiopathology , Animals , Anterior Horn Cells/drug effects , Anterior Horn Cells/pathology , Motor Neurons/drug effects , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Nitrosative Stress/drug effects , Oxidative Stress/drug effects , Radiculopathy/drug therapy , Rats, Sprague-Dawley , Recovery of Function/drug effects , Spinal Nerve Roots/drug effects
2.
Nat Commun ; 12(1): 4399, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34285221

ABSTRACT

The decline of neuronal synapses is an established feature of ageing accompanied by the diminishment of neuronal function, and in the motor system at least, a reduction of behavioural capacity. Here, we have investigated Drosophila motor neuron synaptic terminals during ageing. We observed cumulative fragmentation of presynaptic structures accompanied by diminishment of both evoked and miniature neurotransmission occurring in tandem with reduced motor ability. Through discrete manipulation of each neurotransmission modality, we find that miniature but not evoked neurotransmission is required to maintain presynaptic architecture and that increasing miniature events can both preserve synaptic structures and prolong motor ability during ageing. Our results establish that miniature neurotransmission, formerly viewed as an epiphenomenon, is necessary for the long-term stability of synaptic connections.


Subject(s)
Aging/physiology , Motor Neurons/physiology , Presynaptic Terminals/physiology , Synaptic Transmission/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Evoked Potentials, Motor/physiology , Male , Microscopy, Electron , Models, Animal , Motor Neurons/ultrastructure , Muscles/innervation , Muscles/physiology , Muscles/ultrastructure , Presynaptic Terminals/ultrastructure , Time Factors
3.
J Tissue Eng Regen Med ; 15(9): 763-775, 2021 09.
Article in English | MEDLINE | ID: mdl-34030216

ABSTRACT

Motoneuron transplantation into peripheral nerves undergoing Wallerian degeneration may have applications in treating diseases causing muscle paralysis. We investigated whether functional reinnervation of denervated muscle could be achieved by early or delayed transplantation after denervation. Adult rats were assigned to six groups with increasing denervation periods (0, 1, 4, 8, 12, and 24 weeks) before inoculation with culture medium containing (transplantation group) or lacking (surgical control group) dissociated embryonic motoneurons into the peroneal nerve. Electrophysiological and tissue analyses were performed 3 months after transplantation. Reinnervation of denervated muscles significantly increased relative muscle weight in the transplantation group compared with the surgical control group for denervation periods of 1 week (0.042% ± 0.0031% vs. 0.032% ± 0.0020%, respectively; p = 0.009), 4 weeks (0.044% ± 0.0069% vs. 0.026% ± 0.0045%, respectively; p = 0.0023), and 8 weeks (0.044% ± 0.0029% vs. 0.026% ± 0.0008%, respectively; p = 0.0023). The ratios of reinnervated muscle contractile forces to naïve muscle in the 0, 1, 4, 8, and 12 weeks transplantation groups were 3.79%, 18.99%, 8.05%, 6.30%, and 5.80%, respectively, indicating that these forces were sufficient for walking. The optimal implantation time for transplantation of motoneurons into the peripheral nerve was 1 week after nerve transection. However, the neurons transplanted 24 weeks after denervation survived and regenerated axons. These results indicated that there is time for preparing cells for transplantation in regenerative medicine and suggested that our method may be useful for paralysed muscles that are not expected to recover with current treatment.


Subject(s)
Denervation , Graft Survival , Motor Neurons/transplantation , Muscle, Skeletal/innervation , Peripheral Nerves/pathology , Wallerian Degeneration/therapy , Animals , Biomechanical Phenomena , Cell Survival , Electromyography , Motor Neurons/ultrastructure , Muscle Contraction/physiology , Muscle, Skeletal/diagnostic imaging , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Muscular Atrophy/prevention & control , Peripheral Nerves/physiopathology , Peripheral Nerves/ultrastructure , Rats, Inbred F344 , Wallerian Degeneration/physiopathology
4.
Cell Rep ; 34(12): 108889, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33761348

ABSTRACT

Spinal cord injury (SCI) often results in spasticity. There is currently no effective therapy for spasticity. Here, we describe a method to efficiently differentiate human pluripotent stem cells from spinal GABA neurons. After transplantation into the injured rat spinal cord, the DREADD (designer receptors exclusively activated by designer drug)-expressing spinal progenitors differentiate into GABA neurons, mitigating spasticity-like response of the rat hindlimbs and locomotion deficits in 3 months. Administering clozapine-N-oxide, which activates the grafted GABA neurons, further alleviates spasticity-like response, suggesting an integration of grafted GABA neurons into the local neural circuit. These results highlight the therapeutic potential of the spinal GABA neurons for SCI.


Subject(s)
GABAergic Neurons/pathology , Muscle Spasticity/pathology , Muscle Spasticity/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Spinal Cord/pathology , Action Potentials/physiology , Animals , Cell Differentiation , Cell Survival , Humans , Locomotion , Lumbar Vertebrae/pathology , Lumbar Vertebrae/physiopathology , Male , Motor Neurons/pathology , Motor Neurons/ultrastructure , Muscle Spasticity/complications , Neural Inhibition , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/transplantation , Rats, Sprague-Dawley , Spinal Cord/physiopathology , Spinal Cord Injuries/complications , Spinal Cord Injuries/therapy , Synapses/metabolism , Synapses/ultrastructure
5.
J Comp Neurol ; 529(11): 2842-2864, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33598920

ABSTRACT

The trigeminal blink reflex plays an important role in protecting the corneal surface from damage and preserving visual function in an unpredictable environment. The closing phase of the human reflex, produced by activation of the orbicularis oculi (ObOc) muscles, consists of an initial, small, ipsilateral R1 component, followed by a larger, bilateral R2 component. We investigated the circuitry that underlies this reflex in macaque (Macaca fascicularis and Macaca mulatta) monkeys by the use of single and dual tracer methods. Injection of retrograde tracer into the facial nucleus labeled neurons in the principal trigeminal nucleus, and in the spinal nucleus pars oralis and interpolaris, bilaterally, and in pars caudalis, ipsilaterally. Injection of anterograde tracer into the principal trigeminal nucleus labeled axons that directly terminated on ObOc motoneurons, with an ipsilateral predominance. Injection of anterograde tracer into pars caudalis of the spinal trigeminal nucleus labeled axons that directly terminated on ipsilateral ObOc motoneurons. The observed pattern of labeling indicates that the reticular formation ventromedial to the principal and spinal nuclei also contributes extensive bilateral input to ObOc motoneurons. Thus, much of the trigeminal sensory complex is in a position to supply a monosynaptic drive for lid closure, and the adjacent reticular formation can supply a disynaptic drive. These findings indicate that the assignment of the R1 and R2 components of the blink reflex to different parts of the trigeminal sensory complex cannot be exclusively based on subdivision connectional relationships with facial motoneurons. The characteristics of the R2 component may be due, instead, to other circuit properties.


Subject(s)
Blinking/physiology , Motor Neurons/physiology , Nerve Net/physiology , Trigeminal Nucleus, Spinal/physiology , Animals , Female , Macaca fascicularis , Macaca mulatta , Male , Motor Neurons/chemistry , Motor Neurons/ultrastructure , Nerve Net/chemistry , Nerve Net/ultrastructure , Trigeminal Nucleus, Spinal/chemistry , Trigeminal Nucleus, Spinal/ultrastructure
6.
Brain Res ; 1758: 147331, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33539796

ABSTRACT

During spaceflight and immediately after it, adaptive neuroplastic changes occur in the sensorimotor structures of the central nervous system, which are associated with changes of mainly vestibular and visual signals. It is known that the movement of the eyeball in the vertical direction is carried out by muscles that are innervated by the trochlear nerve (CN IV) and the oculomotor nerve (CN III). To elucidate the cellular processes underlying the atypical vertical nystagmus that occurs under microgravity conditions, it seems necessary to study the state of these nuclei in animals in more detail after prolonged space flights. We carried out a qualitative and quantitative light-optical and ultrastructural analysis of the nuclei of the trochlear nerve in mice after a 30-day flight on the Bion-M1 biosatellite. As a result, it was shown that the dendrites of motoneurons in the nucleus of the trochlear nerve significantly reorganized their geometry and orientation under microgravity conditions. The number of dendritic branches was increased, possibly in order to amplify the reduced signal flow. To ensure such plastic changes, the number and size of mitochondria in the soma of motoneurons and in axons coming from the vestibular structures increased. Thus, the main role in the adaptation of the trochlear nucleus to microgravity conditions, apparently, belongs to the dendrites of motoneurons, which rearrange their structure and function to enhance the flow of sensory information. These results complement our knowledge of the causes of atypical nystagmus in microgravity.


Subject(s)
Adaptation, Physiological/physiology , Motor Neurons/ultrastructure , Space Flight , Trochlear Nerve/ultrastructure , Weightlessness/adverse effects , Animals , Male , Mice , Mice, Inbred C57BL
7.
J Neurochem ; 157(3): 520-531, 2021 05.
Article in English | MEDLINE | ID: mdl-33480022

ABSTRACT

Progranulin (PGRN) is a secreted glycoprotein with multiple biological functions in early embryogenesis, anti-inflammation, and neurodegeneration. A good model for the functional study of PGRN is the zebrafish with knockdown or knockout of grn, the gene encoding PGRN. Morpholino oligonucleotides (MOs) and zinc finger nucleases have been used to generate zebrafish grn models, yet they have shown inconsistent phenotypes due to either the neurotoxicity of the MOs or possible genetic compensation responses during gene editing. In this study, we generated stable grna (one of the major grn homologues of zebrafish) knockout zebrafish by using CRISPR/Cas9-mediated genome editing. A grna sgRNA was designed to target the similar repeated sequence shared by exon 13, exon 15, and exon 19 in zebrafish. The F1 generation with the frameshift mutation of + 4 bp (the addition of 4 bp to exon15), which causes a premature termination, was obtained and subjected to morphological and behavioral evaluation. The grna knockout zebrafish showed neurodevelopmental defects, including spinal motor neurons with shorter axons, decreased sensory hair cells, thinning of the outer nuclear layer and thickening of the inner nuclear layer of the retina, decreased expression of rhodopsin in the cone cells, and motor behavior changes. Moreover, the phenotypes of grna knockout zebrafish could be rescued with the Tol2 system carrying the grna gene. The grna knockout zebrafish model generated in this study provides a useful tool to study PGRN function and has potential for high-throughput drug screening for disease therapy.


Subject(s)
CRISPR-Cas Systems , Gene Knockout Techniques/methods , Intercellular Signaling Peptides and Proteins/genetics , Motor Activity/genetics , Neurodevelopmental Disorders/genetics , RNA, Guide, Kinetoplastida/genetics , Zebrafish Proteins/genetics , Zebrafish , Animals , Axons/pathology , Axons/ultrastructure , Behavior, Animal , Body Weight/genetics , CRISPR-Associated Protein 9 , Exons/genetics , Frameshift Mutation , Genotype , High-Throughput Screening Assays/methods , Mice, Transgenic , Motor Neurons/pathology , Motor Neurons/ultrastructure , Neurodevelopmental Disorders/physiopathology , Neurodevelopmental Disorders/psychology
8.
Cell ; 184(3): 759-774.e18, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33400916

ABSTRACT

To investigate circuit mechanisms underlying locomotor behavior, we used serial-section electron microscopy (EM) to acquire a synapse-resolution dataset containing the ventral nerve cord (VNC) of an adult female Drosophila melanogaster. To generate this dataset, we developed GridTape, a technology that combines automated serial-section collection with automated high-throughput transmission EM. Using this dataset, we studied neuronal networks that control leg and wing movements by reconstructing all 507 motor neurons that control the limbs. We show that a specific class of leg sensory neurons synapses directly onto motor neurons with the largest-caliber axons on both sides of the body, representing a unique pathway for fast limb control. We provide open access to the dataset and reconstructions registered to a standard atlas to permit matching of cells between EM and light microscopy data. We also provide GridTape instrumentation designs and software to make large-scale EM more accessible and affordable to the scientific community.


Subject(s)
Aging/physiology , Drosophila melanogaster/ultrastructure , Microscopy, Electron, Transmission , Motor Neurons/ultrastructure , Sensory Receptor Cells/ultrastructure , Animals , Automation , Connectome , Extremities/innervation , Peripheral Nerves/ultrastructure , Synapses/ultrastructure
9.
Brain Struct Funct ; 226(3): 889-900, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33475854

ABSTRACT

Detailed information about the development of excitatory and inhibitory synapses on the genioglossal (GG) motoneuron may help to understand the mechanism of fine control of GG motoneuron firing and the coordinated tongue movement during postnatal development. For this, we investigated the development of γ-aminobutyric acid (GABA)-immunopositive (GABA +), glycine + (Gly +), and glutamate + (Glut +) axon terminals (boutons) on the somata of rat GG motoneurons at a postnatal day 2 (P2), P6 and P18 by retrograde labeling of GG motoneurons with horseradish peroxidase, electron microscopic postembedding immunogold staining with GABA, Gly, and Glut antisera, and quantitative analysis. The number of boutons per GG motoneuron somata and the mean length of bouton apposition, measures of bouton size and synaptic covering percentage, were significantly increased from P2/P6 to P18. The number and fraction of GABA + only boutons of all boutons decreased significantly, whereas those of Gly + only boutons increased significantly from P2/P6 to P18, suggesting developmental switch from GABAergic to glycinergic synaptic transmission. The fraction of mixed GABA +/Gly + boutons of all boutons was the highest among inhibitory bouton types throughout the postnatal development. The fractions of excitatory and inhibitory boutons of all boutons remained unchanged during postnatal development. These findings reveal a distinct developmental pattern of inhibitory synapses on the GG motoneurons different from that on spinal or trigeminal motoneurons, which may have an important role in the regulation of the precise and coordinated movements of the tongue during the maturation of the oral motor system.


Subject(s)
Dendrites/ultrastructure , Glutamic Acid/metabolism , Motor Neurons/ultrastructure , Presynaptic Terminals/ultrastructure , Animals , Male , Microscopy, Electron/methods , Motor Neurons/physiology , Neural Inhibition/physiology , Rats, Sprague-Dawley , Synapses/physiology , Trigeminal Nuclei/ultrastructure , gamma-Aminobutyric Acid/metabolism
10.
Muscle Nerve ; 63(3): 413-420, 2021 03.
Article in English | MEDLINE | ID: mdl-33269488

ABSTRACT

INTRODUCTION: We recently developed an inducible model of dysphagia using intralingual injection of cholera toxin B conjugated to saporin (CTB-SAP) to cause death of hypoglossal neurons. In this study we aimed to evaluate tongue morphology and ultrastructural changes in hypoglossal neurons and nerve fibers in this model. METHODS: Tissues were collected from 20 rats (10 control and 10 CTB-SAP animals) on day 9 post-injection. Tongues were weighed, measured, and analyzed for microscopic changes using laminin immunohistochemistry. Hypoglossal neurons and axons were examined using transmission electron microscopy. RESULTS: The cross-sectional area of myofibers in the posterior genioglossus was decreased in CTB-SAP-injected rats. Degenerative changes were observed in both the cell bodies and distal axons of hypoglossal neurons. DISCUSSION: Preliminary results indicate this model may have translational application to a variety of neurodegenerative diseases resulting in tongue dysfunction and associated dysphagia.


Subject(s)
Cholera Toxin/pharmacology , Deglutition Disorders , Disease Models, Animal , Hypoglossal Nerve/drug effects , Motor Neurons/drug effects , Muscle Fibers, Skeletal/drug effects , Rats , Saporins/pharmacology , Tongue/drug effects , Animals , Axons/drug effects , Axons/ultrastructure , Hypoglossal Nerve/ultrastructure , Immunohistochemistry , Injections, Intramuscular , Laminin , Motor Neurons/ultrastructure , Muscle Fibers, Skeletal/pathology , Neurons/drug effects , Neurons/ultrastructure , Organ Size , Tongue/pathology
11.
Cell Death Differ ; 28(4): 1379-1397, 2021 04.
Article in English | MEDLINE | ID: mdl-33184465

ABSTRACT

Motor neurons (MNs) are highly energetic cells and recent studies suggest that altered energy metabolism precede MN loss in amyotrophic lateral sclerosis (ALS), an age-onset neurodegenerative disease. However, clear mechanistic insights linking altered metabolism and MN death are still missing. In this study, induced pluripotent stem cells from healthy controls, familial ALS, and sporadic ALS patients were differentiated toward spinal MNs, cortical neurons, and cardiomyocytes. Metabolic flux analyses reveal an MN-specific deficiency in mitochondrial respiration in ALS. Intriguingly, all forms of familial and sporadic ALS MNs tested in our study exhibited similar defective metabolic profiles, which were attributed to hyper-acetylation of mitochondrial proteins. In the mitochondria, Sirtuin-3 (SIRT3) functions as a mitochondrial deacetylase to maintain mitochondrial function and integrity. We found that activating SIRT3 using nicotinamide or a small molecule activator reversed the defective metabolic profiles in all our ALS MNs, as well as correct a constellation of ALS-associated phenotypes.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Motor Neurons/metabolism , Motor Neurons/pathology , Sirtuin 3/genetics , Animals , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mitochondria/metabolism , Mitochondria/ultrastructure , Motor Neurons/ultrastructure , Sirtuin 3/metabolism
12.
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
13.
Nat Neurosci ; 23(12): 1637-1643, 2020 12.
Article in English | MEDLINE | ID: mdl-32929244

ABSTRACT

Imaging neuronal networks provides a foundation for understanding the nervous system, but resolving dense nanometer-scale structures over large volumes remains challenging for light microscopy (LM) and electron microscopy (EM). Here we show that X-ray holographic nano-tomography (XNH) can image millimeter-scale volumes with sub-100-nm resolution, enabling reconstruction of dense wiring in Drosophila melanogaster and mouse nervous tissue. We performed correlative XNH and EM to reconstruct hundreds of cortical pyramidal cells and show that more superficial cells receive stronger synaptic inhibition on their apical dendrites. By combining multiple XNH scans, we imaged an adult Drosophila leg with sufficient resolution to comprehensively catalog mechanosensory neurons and trace individual motor axons from muscles to the central nervous system. To accelerate neuronal reconstructions, we trained a convolutional neural network to automatically segment neurons from XNH volumes. Thus, XNH bridges a key gap between LM and EM, providing a new avenue for neural circuit discovery.


Subject(s)
Image Processing, Computer-Assisted/methods , Neurons/ultrastructure , Animals , Axons/physiology , Axons/ultrastructure , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Cerebral Cortex/ultrastructure , Dendrites/physiology , Dendrites/ultrastructure , Drosophila melanogaster , Female , Holography , Imaging, Three-Dimensional , Machine Learning , Male , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Motor Neurons/ultrastructure , Muscle, Skeletal/innervation , Muscle, Skeletal/ultrastructure , Nanotechnology , Neural Networks, Computer , Pyramidal Cells/ultrastructure , Sensory Receptor Cells/physiology , Sensory Receptor Cells/ultrastructure , Tomography
14.
J Neurogenet ; 34(3-4): 307-322, 2020.
Article in English | MEDLINE | ID: mdl-32808848

ABSTRACT

Sydney Brenner's choice of Caenorhabditis elegans as a model organism for understanding the nervous system has accelerated discoveries of gene function in neural circuit development and behavior. In this review, we discuss a striking example of synaptic remodeling in the C. elegans motor circuit in which DD class motor neurons effectively reverse polarity as presynaptic and postsynaptic domains at opposite ends of the DD neurite switch locations. Originally revealed by EM reconstruction conducted over 40 years ago, DD remodeling has since been investigated by live cell imaging methods that exploit the power of C. elegans genetics to reveal key effectors of synaptic plasticity. Although synapses are also extensively rewired in developing mammalian circuits, the underlying remodeling mechanisms are largely unknown. Here, we highlight the possibility that studies in C. elegans can reveal pathways that orchestrate synaptic remodeling in more complex organisms. Specifically, we describe (1) transcription factors that regulate DD remodeling, (2) the cellular and molecular cascades that drive synaptic remodeling and (3) examples of circuit modifications in vertebrate neurons that share some similarities with synaptic remodeling in C. elegans DD neurons.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Cyclic AMP/physiology , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Gene Expression Regulation , Genes, Reporter , Intravital Microscopy , Ion Channels/genetics , Ion Channels/physiology , Larva , Mammals/physiology , Microscopy, Electron , Microtubules/ultrastructure , Motor Neurons/physiology , Motor Neurons/ultrastructure , Nerve Tissue Proteins/physiology , Neurons/ultrastructure , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Recombinant Proteins/metabolism , Strigiformes/physiology , Transcription Factors/physiology , gamma-Aminobutyric Acid/physiology
15.
J Cell Biol ; 219(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32369543

ABSTRACT

The maintenance of axons for the lifetime of an organism requires an axonal cytoskeleton that is robust but also flexible to adapt to mechanical challenges and to support plastic changes of axon morphology. Furthermore, cytoskeletal organization has to adapt to axons of dramatically different dimensions, and to their compartment-specific requirements in the axon initial segment, in the axon shaft, at synapses or in growth cones. To understand how the cytoskeleton caters to these different demands, this review summarizes five decades of electron microscopic studies. It focuses on the organization of microtubules and neurofilaments in axon shafts in both vertebrate and invertebrate neurons, as well as the axon initial segments of vertebrate motor- and interneurons. Findings from these ultrastructural studies are being interpreted here on the basis of our contemporary molecular understanding. They strongly suggest that axon architecture in animals as diverse as arthropods and vertebrates is dependent on loosely cross-linked bundles of microtubules running all along axons, with only minor roles played by neurofilaments.


Subject(s)
Axons/ultrastructure , Cytoskeleton/ultrastructure , Intermediate Filaments/ultrastructure , Microtubules/ultrastructure , Sensory Receptor Cells/ultrastructure , Animals , Axons/physiology , Cytoskeleton/physiology , Intermediate Filaments/physiology , Interneurons/physiology , Interneurons/ultrastructure , Invertebrates/anatomy & histology , Invertebrates/physiology , Microtubules/physiology , Motor Neurons/physiology , Motor Neurons/ultrastructure , Neuronal Plasticity/physiology , Sensory Receptor Cells/physiology , Vertebrates/anatomy & histology , Vertebrates/physiology
16.
Brain Res ; 1741: 146875, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32389588

ABSTRACT

Diazoxide (DZX), an anti-hypertonic and anti-hypoglycemic drug, was shown to have anti-inflammatory effects in several injured cell types outside the central nervous system. In the brain, the neuroprotective potential of DZX is well described, however, its anticipated anti-inflammatory effect after acute injury has not been systematically analyzed. To disclose the anti-inflammatory effect of DZX in the central nervous system, an injury was induced in the hypoglossal and facial nuclei and in the oculomotor nucleus by unilateral axonal transection and unilateral target deprivation (enucleation), respectively. On the fourth day after surgery, microglial analysis was performed on tissue in which microglia were DAB-labeled and motoneurons were labeled with immunofluorescence. DZX treatment was given either prophylactically, starting 7 days prior to the injury and continuing until the animals were sacrificed, or postoperatively only, with daily intraperitoneal injections (1.25 mg/kg; in 10 mg/ml dimethyl sulfoxide in distilled water). Prophylactically + postoperatively applied DZX completely eliminated the microglial reaction in each motor nuclei. If DZX was applied only postoperatively, some microglial activation could be detected, but its magnitude was still significantly smaller than the non-DZX-treated controls. The effect of DZX could also be demonstrated through an extended period, as tested in the hypoglossal nucleus on day 7 after the operation. Neuronal counts, determined at day 4 after the operation in the hypoglossal nucleus, demonstrated no loss of motor neurons, however, an increased Feret's diameter of mitochondria could be measured, suggesting increased oxidative stress in the injured cells. The increase of mitochondrial Feret's diameter could also be prevented with DZX treatment.


Subject(s)
Brain Stem/drug effects , Diazoxide/administration & dosage , Gliosis/drug therapy , Microglia/drug effects , Motor Neurons/drug effects , Vasodilator Agents/administration & dosage , Animals , Brain Stem/metabolism , Brain Stem/ultrastructure , Drug Administration Schedule , Facial Nucleus/drug effects , Facial Nucleus/metabolism , Facial Nucleus/ultrastructure , Gliosis/metabolism , Gliosis/pathology , Male , Mice , Mice, Inbred BALB C , Microglia/metabolism , Microglia/ultrastructure , Motor Neurons/metabolism , Motor Neurons/ultrastructure , Oculomotor Nuclear Complex/drug effects , Oculomotor Nuclear Complex/metabolism , Oculomotor Nuclear Complex/ultrastructure , Oxidative Stress/drug effects , Oxidative Stress/physiology
17.
J Comp Neurol ; 528(16): 2767-2784, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32352566

ABSTRACT

Detailed anatomical maps of individual organs and entire animals have served as invaluable entry points for ensuing dissection of their evolution, development, and function. The pharynx of the nematode Caenorhabditis elegans is a simple neuromuscular organ with a self-contained, autonomously acting nervous system, composed of 20 neurons that fall into 14 anatomically distinct types. Using serial electron micrograph (EM) reconstruction, we re-evaluate here the connectome of the pharyngeal nervous system, providing a novel and more detailed view of its structure and predicted function. Contrasting the previous classification of pharyngeal neurons into distinct inter- and motor neuron classes, we provide evidence that most pharyngeal neurons are also likely sensory neurons and most, if not all, pharyngeal neurons also classify as motor neurons. Together with the extensive cross-connectivity among pharyngeal neurons, which is more widespread than previously realized, the sensory-motor characteristics of most neurons define a shallow network architecture of the pharyngeal connectome. Network analysis reveals that the patterns of neuronal connections are organized into putative computational modules that reflect the known functional domains of the pharynx. Compared with the somatic nervous system, pharyngeal neurons both physically associate with a larger fraction of their neighbors and create synapses with a greater proportion of their neighbors. We speculate that the overall architecture of the pharyngeal nervous system may be reminiscent of the architecture of ancestral, primitive nervous systems.


Subject(s)
Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans/physiology , Connectome , Pharynx/innervation , Pharynx/physiology , Animals , Feeding Behavior/physiology , Motor Neurons/physiology , Motor Neurons/ultrastructure , Sensory Receptor Cells/physiology , Sensory Receptor Cells/ultrastructure , Synapses/physiology , Synapses/ultrastructure
18.
Cell Rep ; 30(10): 3506-3519.e6, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32160553

ABSTRACT

Genetic variations in TMEM106B, coding for a lysosomal membrane protein, affect frontotemporal lobar degeneration (FTLD) in GRN- (coding for progranulin) and C9orf72-expansion carriers and might play a role in aging. To determine the physiological function of TMEM106B, we generated TMEM106B-deficient mice. These mice develop proximal axonal swellings caused by drastically enlarged LAMP1-positive vacuoles, increased retrograde axonal transport of lysosomes, and accumulation of lipofuscin and autophagosomes. Giant vacuoles specifically accumulate at the distal end and within the axon initial segment, but not in peripheral nerves or at axon terminals, resulting in an impaired facial-nerve-dependent motor performance. These data implicate TMEM106B in mediating the axonal transport of LAMP1-positive organelles in motoneurons and axonal sorting at the initial segment. Our data provide mechanistic insight into how TMEM106B affects lysosomal proteolysis and degradative capacity in neurons.


Subject(s)
Axon Initial Segment/metabolism , Frontotemporal Lobar Degeneration/genetics , Genetic Predisposition to Disease , Lysosomes/metabolism , Membrane Proteins/genetics , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Axon Initial Segment/ultrastructure , Axonal Transport , Brain Stem/pathology , Cell Nucleus/metabolism , Facial Nerve/pathology , Lysosomes/ultrastructure , Membrane Proteins/deficiency , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/ultrastructure , Muscles/innervation , Nerve Tissue Proteins/deficiency , Risk Factors
19.
Ecotoxicol Environ Saf ; 191: 110159, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-31962214

ABSTRACT

Mercury chloride (HgCl2) is a chemical pollutant widely found in the environment. This form of mercury is able to promote several damages to the Central Nervous System (CNS), however the effects of HgCl2 on the spinal cord, an important pathway for the communication between the CNS and the periphery, are still poorly understood. The aim of this work was to investigate the effects of HgCl2 exposure on spinal cord of adult rats. For this, animals were exposed to a dose of 0.375 mg/kg/day, for 45 days. Then, they were euthanized, the spinal cord collected and we investigated the mercury concentrations in medullary parenchyma and the effects on oxidative biochemistry, proteomic profile and tissue structures. Our results showed that exposure to this metal promoted increased levels of Hg in the spinal cord, impaired oxidative biochemistry by triggering oxidative stress, mudulated antioxidant system proteins, energy metabolism and myelin structure; as well as caused disruption in the myelin sheath and reduction in neuronal density. Despite the low dose, we conclude that prolonged exposure to HgCl2 triggers biochemical changes and modulates the expression of several proteins, resulting in damage to the myelin sheath and reduced neuronal density in the spinal cord.


Subject(s)
Environmental Pollutants/toxicity , Mercuric Chloride/toxicity , Motor Neurons/drug effects , Neurodegenerative Diseases/chemically induced , Proteome/metabolism , Spinal Cord/drug effects , Animals , Antioxidants/metabolism , Axons/drug effects , Axons/ultrastructure , Male , Motor Neurons/metabolism , Motor Neurons/ultrastructure , Myelin Sheath/ultrastructure , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oxidation-Reduction , Oxidative Stress/drug effects , Proteomics , Rats , Rats, Wistar , Spinal Cord/metabolism , Spinal Cord/ultrastructure
20.
Brain Struct Funct ; 225(1): 403-425, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31875262

ABSTRACT

The motor outflow for the pupillary light reflex originates in the preganglionic motoneuron subdivision of the Edinger-Westphal nucleus (EWpg), which also mediates lens accommodation. Despite their importance for vision, the morphology, ultrastructure and luminance-related inputs of these motoneurons have not been fully described in primates. In macaque monkeys, we labeled EWpg motoneurons from ciliary ganglion and orbital injections. Both approaches indicated preganglionic motoneurons occupy an EWpg organized as a unitary, ipsilateral cell column. When tracers were placed in the pretectal complex, labeled terminals targeted the ipsilateral EWpg and reached contralateral EWpg by crossing both above and below the cerebral aqueduct. They also terminated in the lateral visceral column, a ventrolateral periaqueductal gray region containing neurons projecting to the contralateral pretectum. Combining olivary pretectal and ciliary ganglion injections to determine whether a direct pupillary light reflex projection is present revealed a labeled motoneuron subpopulation that displayed close associations with labeled pretectal terminal boutons. Ultrastructurally, this subpopulation received synaptic contacts from labeled pretectal terminals that contained numerous clear spherical vesicles, suggesting excitation, and scattered dense-core vesicles, suggesting peptidergic co-transmitters. A variety of axon terminal classes, some of which may serve the near response, synapsed on preganglionic motoneurons. Quantitative analysis indicated that pupillary motoneurons receive more inhibitory inputs than lens motoneurons. To summarize, the pupillary light reflex circuit utilizes a monosynaptic, excitatory, bilateral pretectal projection to a distinct subpopulation of EWpg motoneurons. Furthermore, the interconnections between the lateral visceral column and olivary pretectal nucleus may provide pretectal cells with bilateral retinal fields.


Subject(s)
Edinger-Westphal Nucleus/ultrastructure , Motor Neurons/ultrastructure , Reflex, Pupillary/physiology , Synapses/ultrastructure , Animals , Female , Macaca fascicularis , Macaca mulatta , Male , Neural Pathways/ultrastructure , Neuroanatomical Tract-Tracing Techniques , Periaqueductal Gray/ultrastructure , Presynaptic Terminals/ultrastructure , Pretectal Region/ultrastructure , Synaptic Membranes/ultrastructure , Synaptic Vesicles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...