Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Int J Mol Sci ; 19(2)2018 Feb 22.
Article in English | MEDLINE | ID: mdl-29470437

ABSTRACT

The function of the neuromuscular junction in children is amenable to electrophysiological testing. Of the two tests available, repetitive nerve stimulation is uncomfortable and has a reduced sensitivity compared with single-fibre methodology. The latter is the method of choice, recording the variability in neuromuscular transmission as a value called jitter. It can be performed by voluntary activation of the muscle being examined, which is not suitable in children, or by stimulation techniques. A modification of these techniques, called Stimulated Potential Analysis with Concentric needle Electrodes (SPACE), is well tolerated and can be performed while the child is awake. It has a high sensitivity (84%) for the diagnosis of neuromuscular transmission disorders, the majority of which are myasthenic syndromes, and a moderate specificity (70%). The latter can be improved by the exclusion of neurogenic causes and the determination of the degree of jitter abnormality. Minor jitter abnormalities, under 115% of the upper limit of normal, are usually caused by myopathies with an associated neuromuscular transmission disorder, whereas levels higher than this value are usually associated with one of the myasthenic conditions.


Subject(s)
Neuromuscular Junction/abnormalities , Neuropsychological Tests , Child , Electric Stimulation , Electromyography , Humans
2.
Proc Natl Acad Sci U S A ; 107(33): 14863-8, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20679195

ABSTRACT

The cellular basis of age-related behavioral decline remains obscure but alterations in synapses are likely candidates. Accordingly, the beneficial effects on neural function of caloric restriction and exercise, which are among the most effective anti-aging treatments known, might also be mediated by synapses. As a starting point in testing these ideas, we studied the skeletal neuromuscular junction (NMJ), a large, accessible peripheral synapse. Comparison of NMJs in young adult and aged mice revealed a variety of age-related structural alterations, including axonal swellings, sprouting, synaptic detachment, partial or complete withdrawal of axons from some postsynaptic sites, and fragmentation of the postsynaptic specialization. Alterations were significant by 18 mo of age and severe by 24 mo. A life-long calorie-restricted diet significantly decreased the incidence of pre- and postsynaptic abnormalities in 24-mo-old mice and attenuated age-related loss of motor neurons and turnover of muscle fibers. One month of exercise (wheel running) in 22-mo-old mice also reduced age-related synaptic changes but had no effect on motor neuron number or muscle fiber turnover. Time-lapse imaging in vivo revealed that exercise partially reversed synaptic alterations that had already occurred. These results demonstrate a critical effect of aging on synaptic structure and provide evidence that interventions capable of extending health span and lifespan can partially reverse these age-related synaptic changes.


Subject(s)
Aging/physiology , Caloric Restriction , Neuromuscular Junction/physiopathology , Physical Conditioning, Animal/physiology , Animals , Immunohistochemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Motor Neurons/metabolism , Motor Neurons/physiology , Muscle, Skeletal/abnormalities , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Neuromuscular Junction/abnormalities , Neuromuscular Junction/metabolism , Receptors, Cholinergic/metabolism , Synapses/metabolism
3.
Dig Dis ; 30(1): 19-23, 2012.
Article in English | MEDLINE | ID: mdl-22572680

ABSTRACT

The pathogenesis of diverticular disease is still poorly understood and considered to be multifactorial. Whereas classical pathogenetic concepts have focused on risk factors including increasing age, low-fiber diet and connective tissue disorders, novel concepts take into account that patients with diverticular disease exhibit disturbed intestinal motility patterns (that may result in functional obstruction and painful sensations) therefore postulating an underlying enteric neuro-/myopathy. Recent studies including quantitative evaluations of the enteric nervous system (ENS) in diverticular disease yielded hypoganglionic conditions of both myenteric and submucosal plexus as well as a nerve tissue remodeling in chronic diverticular disease. The disturbed neuromuscular communication was proven by demonstrating alterations in several enteric neurotransmitter systems, exemplified for the cholinergic, serotonergic, nitrergic system as well as for vasointestinal peptide, galanin and tachykinins. Novel lines of evidence have added the involvement of neurotrophic factors such as glial cell line-derived neurotrophic factor which is supposed to regulate ENS development and maintenance and which is downregulated in patients with diverticular disease. Consistent with the hypothesis of an enteric myopathy, deficits in smooth muscle integrity and composition such as hypertrophy, fibrotic transformation and gene expression deficits could be delineated. Taken together, the structural and functional findings on alterations of the ENS and the enteric musculature in diverticular disease provide evidence to strengthen the hypothesis that an enteric neuro-/myopathy may contribute to the development of colonic diverticula and the generation of symptoms in the course of the disease.


Subject(s)
Diverticulitis, Colonic/pathology , Neuromuscular Junction/abnormalities , Neuromuscular Junction/pathology , Diverticulitis, Colonic/etiology , Diverticulitis, Colonic/physiopathology , Gastrointestinal Motility , Humans , Neurotransmitter Agents/metabolism
4.
Dig Dis ; 30(1): 24-8, 2012.
Article in English | MEDLINE | ID: mdl-22572681

ABSTRACT

Although diverticular disease of the colon (diverticulosis) is a frequent finding in Western countries, its pathophysiologic grounds are still only partially understood. Traditionally considered as an age-related condition, colonic diverticulosis is probably the final result of several factors concurring together to determine the anatomo-functional abnormalities eventually causing outpouching of the viscus' mucosa. Among these factors, a relevant role seems to be played by an abnormal neuromuscular function of the large bowel, as shown by abnormal myoelectrical and motor function repeatedly described in these patients, as well as by altered visceral perception. These anomalies might be related to the recent demonstration of derangement of enteric innervation (especially involving interstitial cells of Cajal and enteric glial cells), mucosal neuropeptides, and mucosal inflammation. The latter may have a role of paramount importance in the development of visceral hypersensitivity, responsible for abdominal pain in a subset of patients.


Subject(s)
Diverticulitis, Colonic/physiopathology , Neuromuscular Junction/abnormalities , Neuromuscular Junction/physiopathology , Colon/pathology , Colon/physiopathology , Diverticulitis, Colonic/pathology , Gastrointestinal Motility/physiology , Humans , Motor Activity/physiology
5.
Nat Neurosci ; 11(3): 262-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18278041

ABSTRACT

Synapse formation requires proper interaction between pre- and postsynaptic cells. In anterograde signaling, neurons release factors to guide postsynaptic differentiation. However, less is known about how postsynaptic targets retrogradely regulate presynaptic differentiation or function. We found that muscle-specific conditional knockout of beta-catenin (Ctnnb1, also known as beta-cat) in mice caused both morphologic and functional defects in motoneuron terminals of neuromuscular junctions (NMJs). In the absence of muscle beta-catenin, acetylcholine receptor clusters were increased in size and distributed throughout a wider region. Primary nerve branches were mislocated, whereas secondary or intramuscular nerve branches were elongated and reduced in number. Both spontaneous and evoked neurotransmitter release was reduced at the mutant NMJs. Furthermore, short-term plasticity and calcium sensitivity of neurotransmitter release were compromised in beta-catenin-deficient muscle. In contrast, the NMJ was normal in morphology and function in motoneuron-specific beta-catenin-deficient mice. Taken together, these observations indicate a role for muscle beta-catenin in presynaptic differentiation and function, identifying a previously unknown retrograde signaling in the synapse formation and synaptic plasticity.


Subject(s)
Cell Differentiation/genetics , Motor Neurons/metabolism , Muscle, Skeletal/abnormalities , Muscle, Skeletal/innervation , Neuromuscular Junction/abnormalities , Receptors, Cholinergic/metabolism , beta Catenin/metabolism , Animals , Axonal Transport/genetics , Cell Communication/genetics , Growth Cones/metabolism , Growth Cones/ultrastructure , Mice , Mice, Knockout , Motor Neurons/cytology , Muscle, Skeletal/metabolism , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Neuronal Plasticity/genetics , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Receptor Aggregation/genetics , Signal Transduction/genetics , Synapses/metabolism , Synapses/ultrastructure
6.
Neuron ; 55(5): 741-55, 2007 Sep 06.
Article in English | MEDLINE | ID: mdl-17785181

ABSTRACT

Neurexins have been proposed to function as major mediators of the coordinated pre- and postsynaptic apposition. However, key evidence for this role in vivo has been lacking, particularly due to gene redundancy. Here, we have obtained null mutations in the single Drosophila neurexin gene (dnrx). dnrx loss of function prevents the normal proliferation of synaptic boutons at glutamatergic neuromuscular junctions, while dnrx gain of function in neurons has the opposite effect. DNRX mostly localizes to the active zone of presynaptic terminals. Conspicuously, dnrx null mutants display striking defects in synaptic ultrastructure, with the presence of detachments between pre- and postsynaptic membranes, abnormally long active zones, and increased number of T bars. These abnormalities result in corresponding alterations in synaptic transmission with reduced quantal content. Together, our results provide compelling evidence for an in vivo role of neurexins in the modulation of synaptic architecture and adhesive interactions between pre- and postsynaptic compartments.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Differentiation/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Neuromuscular Junction/abnormalities , Synapses/metabolism , Synaptic Transmission/genetics , Animals , Cell Adhesion/genetics , Cell Adhesion Molecules, Neuronal/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental/genetics , Molecular Sequence Data , Motor Neurons/metabolism , Motor Neurons/ultrastructure , Mutation/genetics , Neuromuscular Junction/growth & development , Neuromuscular Junction/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Sequence Homology, Amino Acid , Synapses/genetics , Synapses/ultrastructure , Synaptic Membranes/genetics , Synaptic Membranes/metabolism , Synaptic Membranes/ultrastructure
7.
J Cell Biol ; 173(1): 69-82, 2006 Apr 10.
Article in English | MEDLINE | ID: mdl-16606691

ABSTRACT

Scramblases are a family of single-pass plasma membrane proteins, identified by their purported ability to scramble phospholipids across the two layers of plasma membrane isolated from platelets and red blood cells. However, their true in vivo role has yet to be elucidated. We report the generation and isolation of null mutants of two Scramblases identified in Drosophila melanogaster. We demonstrate that flies lacking either or both of these Scramblases are not compromised in vivo in processes requiring scrambling of phospholipids. Instead, we show that D. melanogaster lacking both Scramblases have more vesicles and display enhanced recruitment from a reserve pool of vesicles and increased neurotransmitter secretion at the larval neuromuscular synapses. These defects are corrected by the introduction of a genomic copy of the Scramb 1 gene. The lack of phenotypes related to failure of scrambling and the neurophysiological analysis lead us to propose that Scramblases play a modulatory role in the process of neurotransmission.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Membrane Proteins/metabolism , Neuromuscular Junction/abnormalities , Neuromuscular Junction/enzymology , Phospholipid Transfer Proteins/physiology , Synaptic Transmission/physiology , Animals , Apoptosis/genetics , Cell Membrane/enzymology , Cell Membrane/genetics , Databases, Nucleic Acid , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Exocytosis/genetics , Gene Expression Regulation, Enzymologic/genetics , Immunity, Innate/genetics , Larva/enzymology , Larva/genetics , Larva/growth & development , Membrane Proteins/genetics , Membrane Proteins/physiology , Microscopy, Electron, Transmission , Molecular Sequence Data , Mutation/genetics , Neuromuscular Junction/genetics , Neurotransmitter Agents/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipids/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Synaptic Membranes/enzymology , Synaptic Membranes/metabolism , Synaptic Membranes/ultrastructure , Synaptic Transmission/genetics , Synaptic Vesicles/enzymology , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure
8.
Nat Neurosci ; 10(2): 177-85, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17220882

ABSTRACT

To understand the functions of NIPA1, mutated in the neurodegenerative disease hereditary spastic paraplegia, and of ichthyin, mutated in autosomal recessive congenital ichthyosis, we have studied their Drosophila melanogaster ortholog, spichthyin (Spict). Spict is found on early endosomes. Loss of Spict leads to upregulation of bone morphogenetic protein (BMP) signaling and expansion of the neuromuscular junction. BMP signaling is also necessary for a normal microtubule cytoskeleton and axonal transport; analysis of loss- and gain-of-function phenotypes indicate that Spict may antagonize this function of BMP signaling. Spict interacts with BMP receptors and promotes their internalization from the plasma membrane, implying that it inhibits BMP signaling by regulating BMP receptor traffic. This is the first demonstration of a role for a hereditary spastic paraplegia protein or ichthyin family member in a specific signaling pathway, and implies disease mechanisms for hereditary spastic paraplegia that involve dependence of the microtubule cytoskeleton on BMP signaling.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Nervous System Malformations/metabolism , Nervous System/embryology , Neuromuscular Junction/abnormalities , Presynaptic Terminals/metabolism , Receptors, Cell Surface/metabolism , Animals , Axonal Transport/genetics , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Expression Regulation, Developmental/genetics , Ichthyosis/genetics , Ichthyosis/metabolism , Ichthyosis/physiopathology , Membrane Proteins/genetics , Microtubules/genetics , Microtubules/metabolism , Microtubules/pathology , Molecular Sequence Data , Nervous System/cytology , Nervous System/metabolism , Nervous System Malformations/genetics , Nervous System Malformations/physiopathology , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Presynaptic Terminals/ultrastructure , Receptors, Cell Surface/genetics , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Signal Transduction/genetics , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Spastic Paraplegia, Hereditary/physiopathology
9.
J Cell Biol ; 150(6): 1385-98, 2000 Sep 18.
Article in English | MEDLINE | ID: mdl-10995443

ABSTRACT

The syntrophins are a family of structurally related proteins that contain multiple protein interaction motifs. Syntrophins associate directly with dystrophin, the product of the Duchenne muscular dystrophy locus, and its homologues. We have generated alpha-syntrophin null mice by targeted gene disruption to test the function of this association. The alpha-Syn(-/)- mice show no evidence of myopathy, despite reduced levels of alpha-dystrobrevin-2. Neuronal nitric oxide synthase, a component of the dystrophin protein complex, is absent from the sarcolemma of the alpha-Syn(-/)- mice, even where other syntrophin isoforms are present. alpha-Syn(-/)- neuromuscular junctions have undetectable levels of postsynaptic utrophin and reduced levels of acetylcholine receptor and acetylcholinesterase. The mutant junctions have shallow nerve gutters, abnormal distributions of acetylcholine receptors, and postjunctional folds that are generally less organized and have fewer openings to the synaptic cleft than controls. Thus, alpha-syntrophin has an important role in synapse formation and in the organization of utrophin, acetylcholine receptor, and acetylcholinesterase at the neuromuscular synapse.


Subject(s)
Cytoskeletal Proteins/deficiency , Dystrophin-Associated Proteins , Membrane Proteins/deficiency , Membrane Proteins/genetics , Muscle Proteins/genetics , Neuromuscular Junction/abnormalities , Synapses/metabolism , Acetylcholinesterase/metabolism , Animals , Blotting, Southern , Calcium-Binding Proteins , Dystrophin/metabolism , Fluorescent Antibody Technique , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Muscle Proteins/metabolism , Muscle, Skeletal/abnormalities , Muscle, Skeletal/enzymology , Neuromuscular Junction/chemistry , Neuromuscular Junction/ultrastructure , Neuropeptides/metabolism , Nitric Oxide Synthase/metabolism , Receptors, Cholinergic/analysis , Receptors, Cholinergic/metabolism , Sarcolemma/metabolism , Synapses/chemistry , Utrophin
10.
Neuron ; 36(4): 635-48, 2002 Nov 14.
Article in English | MEDLINE | ID: mdl-12441053

ABSTRACT

Activity-dependent and -independent signals collaborate to regulate synaptogenesis, but their relative contributions are unclear. Here, we describe the formation of neuromuscular synapses at which neurotransmission is completely and specifically blocked by mutation of the neurotransmitter-synthesizing enzyme choline acetyltransferase. Nerve terminals differentiate extensively in the absence of neurotransmitter, but neurotransmission plays multiple roles in synaptic differentiation. These include influences on the numbers of pre- and postsynaptic partners, the distribution of synapses in the target field, the number of synaptic sites per target cell, and the number of axons per synaptic site. Neurotransmission also regulates the formation or stability of transient acetylcholine receptor-rich processes (myopodia) that may initiate nerve-muscle contact. At subsequent stages, neurotransmission delays some steps in synaptic maturation but accelerates others. Thus, neurotransmission affects synaptogenesis from early stages and coordinates rather than drives synaptic maturation.


Subject(s)
Acetylcholine/deficiency , Cell Differentiation/genetics , Choline O-Acetyltransferase/deficiency , Neuromuscular Junction/abnormalities , Presynaptic Terminals/metabolism , Synaptic Transmission/genetics , Acetylcholine/biosynthesis , Animals , Choline O-Acetyltransferase/genetics , Diaphragm/abnormalities , Diaphragm/innervation , Diaphragm/ultrastructure , Fetus , Gene Deletion , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Electron , Motor Neurons/metabolism , Motor Neurons/ultrastructure , Mutation/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/ultrastructure , Phrenic Nerve/abnormalities , Phrenic Nerve/ultrastructure , Presynaptic Terminals/ultrastructure
11.
Neuron ; 41(4): 521-34, 2004 Feb 19.
Article in English | MEDLINE | ID: mdl-14980202

ABSTRACT

We describe the isolation and characterization of nwk (nervous wreck), a temperature-sensitive paralytic mutant that causes excessive growth of larval neuromuscular junctions (NMJs), resulting in increased synaptic bouton number and branch formation. Ultrastructurally, mutant boutons have reduced size and fewer active zones, associated with a reduction in synaptic transmission. nwk encodes an FCH and SH3 domain-containing adaptor protein that localizes to the periactive zone of presynaptic terminals and binds to the Drosophila ortholog of Wasp (Wsp), a key regulator of actin polymerization. wsp null mutants display synaptic overgrowth similar to nwk and enhance the nwk morphological phenotype in a dose-dependent manner. Evolutionarily, Nwk belongs to a previously undescribed family of adaptor proteins that includes the human srGAPs, which regulate Rho activity downstream of Robo receptors. We propose that Nwk controls synapse morphology by regulating actin dynamics downstream of growth signals in presynaptic terminals.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Drosophila Proteins/metabolism , Drosophila/growth & development , Microfilament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/abnormalities , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/isolation & purification , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , Cell Differentiation/genetics , Chromosome Mapping , DNA, Complementary/analysis , DNA, Complementary/genetics , Drosophila/cytology , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/isolation & purification , Evolution, Molecular , Microscopy, Electron , Molecular Sequence Data , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Neuromuscular Junction/physiology , Neuromuscular Junction/ultrastructure , Phylogeny , Protein Structure, Tertiary/genetics , Sequence Homology, Amino Acid , Synaptic Transmission/genetics , Transgenes/genetics , Wiskott-Aldrich Syndrome Protein , src Homology Domains/genetics
12.
Neuron ; 33(4): 529-43, 2002 Feb 14.
Article in English | MEDLINE | ID: mdl-11856528

ABSTRACT

Proper synaptic development is critical for establishing all aspects of neural function including learning, memory, and locomotion. Here, we describe the phenotypic consequences of mutations in the wishful thinking (wit) gene, the Drosophila homolog of the vertebrate BMP type II receptor. Mutations in wit result in pharate lethality that can be rescued by expression of a wit transgene in motor neurons but not in muscles. Mutant larvae exhibit small synapses, severe defects in evoked junctional potentials, a lower frequency of spontaneous vesicle release, and an alteration in the ultrastructure of synaptic active zones. These results reveal a novel role for BMP signaling in regulating Drosophila neuromuscular junction synapse assembly and activity and may indicate that similar pathways could govern vertebrate synapse development.


Subject(s)
Central Nervous System/abnormalities , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental/physiology , Mutation/physiology , Neuromuscular Junction/abnormalities , Protein Serine-Threonine Kinases/genetics , Receptors, Cell Surface/genetics , Transcription Factors , Animals , Body Patterning/genetics , Bone Morphogenetic Protein Receptors, Type II , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/genetics , Central Nervous System/growth & development , Central Nervous System/ultrastructure , DNA, Complementary/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/isolation & purification , Drosophila melanogaster/growth & development , Drosophila melanogaster/ultrastructure , Female , Genes, Lethal/genetics , Immunohistochemistry , Male , Molecular Sequence Data , Motor Neurons/metabolism , Motor Neurons/ultrastructure , Neuromuscular Junction/growth & development , Neuromuscular Junction/ultrastructure , Neuronal Plasticity/genetics , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Phenotype , Protein Serine-Threonine Kinases/isolation & purification , Receptors, Cell Surface/isolation & purification , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Signal Transduction/genetics
13.
Neuron ; 33(4): 545-58, 2002 Feb 14.
Article in English | MEDLINE | ID: mdl-11856529

ABSTRACT

We conducted a large-scale screen for Drosophila mutants that have structural abnormalities of the larval neuromuscular junction (NMJ). We recovered mutations in wishful thinking (wit), a gene that positively regulates synaptic growth. wit encodes a BMP type II receptor. In wit mutant larvae, the size of the NMJs is greatly reduced relative to the size of the muscles. wit NMJs have reduced evoked excitatory junctional potentials, decreased levels of the synaptic cell adhesion molecule Fasciclin II, and synaptic membrane detachment at active zones. Wit is expressed by a subset of neurons, including motoneurons. The NMJ phenotype is specifically rescued by transgenic expression of Wit only in motoneurons. Thus, Wit appears to function as a presynaptic receptor that regulates synaptic size at the Drosophila NMJ.


Subject(s)
Body Patterning/genetics , Central Nervous System/abnormalities , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental/physiology , Mutation/genetics , Neuromuscular Junction/abnormalities , Protein Serine-Threonine Kinases/genetics , Animals , Animals, Genetically Modified/abnormalities , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Bone Morphogenetic Protein Receptors, Type II , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Adhesion/genetics , Central Nervous System/growth & development , Central Nervous System/ultrastructure , Down-Regulation/genetics , Drosophila Proteins/isolation & purification , Drosophila melanogaster/growth & development , Drosophila melanogaster/ultrastructure , Elapid Venoms/metabolism , Female , Genetic Testing , Male , Molecular Sequence Data , Neuromuscular Junction/growth & development , Neuromuscular Junction/ultrastructure , Neuronal Plasticity/genetics , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Protein Serine-Threonine Kinases/isolation & purification , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Signal Transduction/genetics , Synaptic Membranes/genetics , Synaptic Membranes/metabolism , Synaptic Membranes/ultrastructure
14.
Neuron ; 36(3): 403-16, 2002 Oct 24.
Article in English | MEDLINE | ID: mdl-12408844

ABSTRACT

In a genetic screen for genes that control synapse development, we have identified spinster (spin), which encodes a multipass transmembrane protein. spin mutant synapses reveal a 200% increase in bouton number and a deficit in presynaptic release. We demonstrate that spin is expressed in both nerve and muscle and is required both pre- and postsynaptically for normal synaptic growth. We have localized Spin to a late endosomal compartment and present evidence for altered endosomal/lysosomal function in spin. We also present evidence that synaptic overgrowth in spin is caused by enhanced/misregulated TGF-beta signaling. TGF-beta receptor mutants show dose-dependent suppression of synaptic overgrowth in spin. Furthermore, mutations in Dad, an inhibitory Smad, cause synapse overgrowth. We present a model for synaptic growth control with implications for the etiology of lysosomal storage and neurodegenerative disease.


Subject(s)
Cell Differentiation/genetics , Drosophila Proteins/deficiency , Drosophila melanogaster/growth & development , Endosomes/metabolism , Membrane Proteins/deficiency , Nervous System/growth & development , Presynaptic Terminals/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Compartmentation/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Female , Gene Expression Regulation, Developmental/genetics , HeLa Cells , Humans , Hypertrophy/genetics , Hypertrophy/metabolism , Hypertrophy/physiopathology , Lysosomes/metabolism , Male , Membrane Proteins/genetics , Motor Neurons/cytology , Motor Neurons/metabolism , Muscle, Skeletal/abnormalities , Muscle, Skeletal/growth & development , Muscle, Skeletal/innervation , Mutation/genetics , Nervous System/cytology , Nervous System/metabolism , Neuromuscular Junction/abnormalities , Neuromuscular Junction/growth & development , Neuromuscular Junction/pathology , Presynaptic Terminals/pathology , Presynaptic Terminals/ultrastructure , Synaptic Transmission/genetics , Transforming Growth Factor beta/genetics
15.
Mol Cell Biol ; 25(14): 5973-84, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15988013

ABSTRACT

In cultured hippocampal neurons, synaptogenesis is largely independent of synaptic transmission, while several accounts in the literature indicate that synaptogenesis at cholinergic neuromuscular junctions in mammals appears to partially depend on synaptic activity. To systematically examine the role of synaptic activity in synaptogenesis at the neuromuscular junction, we investigated neuromuscular synaptogenesis and neurotransmitter release of mice lacking all synaptic vesicle priming proteins of the Munc13 family. Munc13-deficient mice are completely paralyzed at birth and die immediately, but form specialized neuromuscular endplates that display typical synaptic features. However, the distribution, number, size, and shape of these synapses, as well as the number of motor neurons they originate from and the maturation state of muscle cells, are profoundly altered. Surprisingly, Munc13-deficient synapses exhibit significantly increased spontaneous quantal acetylcholine release, although fewer fusion-competent synaptic vesicles are present and nerve stimulation-evoked secretion is hardly elicitable and strongly reduced in magnitude. We conclude that the residual transmitter release in Munc13-deficient mice is not sufficient to sustain normal synaptogenesis at the neuromuscular junction, essentially causing morphological aberrations that are also seen upon total blockade of neuromuscular transmission in other genetic models. Our data confirm the importance of Munc13 proteins in synaptic vesicle priming at the neuromuscular junction but indicate also that priming at this synapse may differ from priming at glutamatergic and gamma-aminobutyric acid-ergic synapses and is partly Munc13 independent. Thus, non-Munc13 priming proteins exist at this synapse or vesicle priming occurs in part spontaneously: i.e., without dedicated priming proteins in the release machinery.


Subject(s)
Acetylcholine/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Nerve Tissue Proteins/deficiency , Neuromuscular Junction/abnormalities , Neuromuscular Junction/embryology , Neurotransmitter Agents/metabolism , Animals , Diaphragm/abnormalities , Diaphragm/innervation , Electrophysiology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Mutant Strains , Mutation , Nerve Tissue Proteins/genetics , Neuromuscular Junction/ultrastructure , Phrenic Nerve/abnormalities , Spinal Cord/abnormalities , Synaptic Vesicles/physiology
16.
Neurotoxicol Teratol ; 30(5): 440-7, 2008.
Article in English | MEDLINE | ID: mdl-18508234

ABSTRACT

Though caffeine is broadly distributed in many plants and foods, little is known about the teratogenic effects of caffeine during early embryonic development. Here, we used zebrafish as a model to test toxicity and teratogenicity since they have transparent eggs, making the organogenesis of zebrafish embryos easier to observe. When the exposure doses of caffeine were less than 150 ppm (17.5, 35, 50, 100 and 150 ppm), the zebrafish embryos exhibited no significant differences in survival rates after comparison with vehicle-control (0 ppm) group. As the exposure dosages increased, the survival rates decreased. No embryos survived after treatment with 300 ppm caffeine or higher dosages. The most evident change in embryos treated with caffeine was a shorter body length (vehicle-control: 3.26+/-0.01 mm, n=49; vs 150 ppm of caffeine: 2.67+/-0.03 mm, n=50). In addition, caffeine-treated embryos exhibited significantly reduced tactile sensitivity frequencies of touch-induced movement (vehicle-control: 9.93+/-0.77 vs 17.5-150 ppm caffeine: 5.37+/-0.52-0.10+/-0.06). Subtle changes are easily observed by staining with specific monoclonal antibodies F59, Znp1 and Zn5 to detect morphological changes in muscle fibers, primary motor axons and secondary motor axon projections, respectively. Our data show that the treatment of caffeine leads to misalignment of muscle fibers and motor neuron defects, especially secondary motor neuron axonal growth defects.


Subject(s)
Caffeine/toxicity , Dyskinesia, Drug-Induced/physiopathology , Embryo, Nonmammalian/drug effects , Motor Neurons/drug effects , Muscle, Skeletal/drug effects , Spinal Cord/drug effects , Animals , Antibodies, Monoclonal , Disease Models, Animal , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/pathology , Embryo, Nonmammalian/abnormalities , Embryonic Development/drug effects , Embryonic Development/physiology , Motor Neurons/pathology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/abnormalities , Muscle, Skeletal/innervation , Neuromuscular Junction/abnormalities , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiopathology , Phosphodiesterase Inhibitors/toxicity , Sensation Disorders/chemically induced , Sensation Disorders/pathology , Sensation Disorders/physiopathology , Spinal Cord/abnormalities , Survival Rate , Time Factors , Toxicity Tests , Zebrafish
17.
Neuroscience ; 148(3): 700-11, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17706883

ABSTRACT

ATP is co-released in significant quantities with acetylcholine from motor neurons at skeletal neuromuscular junctions (NMJ). However, the role of this neurotransmitter in muscle function remains unclear. The P2X2 ion channel receptor subunit is expressed during development of the skeletal NMJ, but not in adult muscle fibers, although it is re-expressed during muscle fiber regeneration. Using mice deficient for the P2X2 receptor subunit for ATP (P2X2(-/-)), we demonstrate a role for purinergic signaling in NMJ development. Whereas control NMJs were characterized by precise apposition of pre-synaptic motor nerve terminals and post-synaptic junctional folds rich in acetylcholine receptors (AChRs), NMJs in P2X2(-/-) mice were disorganized: misapposition of nerve terminals and post-synaptic AChR expression localization was common; the density of post-synaptic junctional folds was reduced; and there was increased end-plate fragmentation. These changes in NMJ structure were associated with muscle fiber atrophy. In addition there was an increase in the proportion of fast type muscle fibers. These findings demonstrate a role for P2X2 receptor-mediated signaling in NMJ formation and suggest that purinergic signaling may play an as yet largely unrecognized part in synapse formation.


Subject(s)
Adenosine Triphosphate/metabolism , Muscle, Skeletal/abnormalities , Muscle, Skeletal/physiopathology , Neuromuscular Junction/abnormalities , Neuromuscular Junction/physiopathology , Receptors, Purinergic P2/genetics , Acetylcholine/metabolism , Animals , Cell Differentiation/genetics , Female , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle, Skeletal/innervation , Neuromuscular Junction/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Receptors, Purinergic P2X2 , Signal Transduction/genetics , Synapses/metabolism , Synapses/pathology , Synaptic Membranes/metabolism , Synaptic Membranes/pathology , Synaptic Transmission/genetics
18.
Neurotoxicol Teratol ; 29(5): 562-9, 2007.
Article in English | MEDLINE | ID: mdl-17644306

ABSTRACT

Sodium benzoate (SB) is a commonly used food preservative and anti-microbial agent in many foods from soup to cereals. However, little is known about the SB-induced toxicity and teratogenicity during early embryonic development. Here, we used zebrafish as a model to test the toxicity and teratogenicity because of their transparent eggs; therefore, the organogenesis of zebrafish embryos is easy to observe. After low dosages of SB (1-1000 ppm) treatment, the zebrafish embryos exhibited a 100% survival rate. As the exposure dosages increased, the survival rates decreased. No embryos survived after treatment with 2000 ppm SB. The 50% lethal dose (LD(50)) of zebrafish is found to be in the range of 1400-1500 ppm. Gut abnormalities, malformation of pronephros, defective hatching gland and edema in pericardial sac were observed after treatment with SB. Compared to untreated littermates (vehicle-treated control), SB-treated embryos exhibited significantly reduced tactile sensitivity frequencies of touch-induced movement (vehicle-treated control: 27.60+/-1.98 v.s. 1000 ppm SB: 7.89+/-5.28; N=30). Subtle changes are easily observed by staining with specific monoclonal antibodies F59, Znp1 and alpha6F to detect morphology changes in muscle fibers, motor axons and pronephros, respectively. Our data showed that the treatment of SB led to misalignment of muscle fibers, motor neuron innervations, excess acetyl-choline receptor cluster and defective pronephric tubes. On the basis of these observations, we suggest that sodium benzoate is able to induce neurotoxicity and nephrotoxicity of zebrafish larvae.


Subject(s)
Abnormalities, Drug-Induced/pathology , Abnormalities, Drug-Induced/psychology , Larva/anatomy & histology , Sodium Benzoate/toxicity , Teratogens , Zebrafish/physiology , Animals , Antibodies, Monoclonal , Dose-Response Relationship, Drug , Embryo, Nonmammalian/drug effects , Gastrointestinal Tract/abnormalities , Kidney/abnormalities , Kidney/growth & development , Motor Activity/drug effects , Motor Neurons/drug effects , Motor Neurons/pathology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Neuromuscular Junction/abnormalities , Neuromuscular Junction/pathology , Phenotype , Physical Stimulation , Receptors, Cholinergic/drug effects , Receptors, Cholinergic/genetics , Time Factors
19.
J Neurosci ; 24(46): 10302-9, 2004 Nov 17.
Article in English | MEDLINE | ID: mdl-15548643

ABSTRACT

The syntrophins are modular adapter proteins that function by recruiting signaling molecules to the cytoskeleton via their direct association with proteins of the dystrophin protein family. We investigated the physiological function of beta2-syntrophin by generating a line of mice lacking this syntrophin isoform. The beta2-syntrophin null mice show no overt phenotype, or muscular dystrophy, and form structurally normal neuromuscular junctions (NMJs). To determine whether physiological consequences caused by the lack of beta2-syntrophin were masked by compensation from the alpha-syntrophin isoform, we crossed these mice with our previously described alpha-syntrophin null mice to produce mice lacking both isoforms. The alpha/beta2-syntrophin null mice have NMJs that are structurally more aberrant than those lacking only alpha-syntrophin. The NMJs of the alpha/beta2-syntrophin null mice have fewer junctional folds than either parent strain, and the remaining folds are abnormally shaped with few openings to the synaptic space. The levels of acetylcholine receptors are reduced to 23% of wild type in mice lacking both syntrophin isoforms. Furthermore, the alpha/beta2-syntrophin null mice ran significantly shorter distances on voluntary exercise wheels despite having normal neuromuscular junction transmission as determined by micro-electrode recording of endplate potentials. We conclude that both alpha-syntrophin and beta2-syntrophin play distinct roles in forming and maintaining NMJ structure and that each syntrophin can partially compensate for the loss of the other.


Subject(s)
Membrane Proteins/physiology , Muscle Proteins/physiology , Neuromuscular Junction/physiology , Animals , Calcium-Binding Proteins , Crosses, Genetic , Diaphragm/physiology , Dystrophin-Associated Proteins , Membrane Potentials , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Microscopy, Fluorescence , Motor Activity/physiology , Motor Endplate/physiology , Muscle Proteins/genetics , Neuromuscular Junction/abnormalities , Neuromuscular Junction/ultrastructure , Phenotype , Protein Isoforms/genetics , Protein Isoforms/physiology , Receptors, Cholinergic/metabolism
20.
Ann N Y Acad Sci ; 998: 114-24, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14592868
SELECTION OF CITATIONS
SEARCH DETAIL