Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Am J Physiol Cell Physiol ; 318(5): C1030-C1041, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32293933

ABSTRACT

Na,K-ATPase is a membrane transporter that is critically important for skeletal muscle function. Mdx and Bla/J mice are the experimental models of Duchenne muscular dystrophy and dysferlinopathy that are known to differ in the molecular mechanism of the pathology. This study examines the function of α1- and α2-Na,K-ATPase isozymes in respiratory diaphragm and postural soleus muscles from mdx and Bla/J mice compared with control С57Bl/6 mice. In diaphragm muscles, the motor endplate structure was severely disturbed (manifested by defragmentation) in mdx mice only. The endplate membrane of both Bla/J and mdx mice was depolarized due to specific loss of the α2-Na,K-ATPase electrogenic activity and its decreased membrane abundance. Total FXYD1 subunit (modulates Na,K-ATPase activity) abundance was decreased in both mouse models. However, the α2-Na,K-ATPase protein content as well as mRNA expression were specifically and significantly reduced only in mdx mice. The endplate membrane cholesterol redistribution was most pronounced in mdx mice. Soleus muscles from Bla/J and mdx mice demonstrated reduction of the α2-Na,K-ATPase membrane abundance and mRNA expression similar to the diaphragm muscles. In contrast to diaphragm, the α2-Na,K-ATPase protein content was altered in both Bla/J and mdx mice; membrane cholesterol re-distribution was not observed. Thus, the α2-Na,K-ATPase is altered in both Bla/J and mdx mouse models of chronic muscle pathology. However, despite some similarities, the α2-Na,K-ATPase and cholesterol abnormalities are more pronounced in mdx mice.


Subject(s)
Membrane Proteins/genetics , Muscular Dystrophies/genetics , Phosphoproteins/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Cholesterol/genetics , Cholesterol/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred mdx , Motor Endplate/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Protein Isoforms/genetics , RNA, Messenger/genetics
2.
J Cell Physiol ; 233(10): 6329-6336, 2018 10.
Article in English | MEDLINE | ID: mdl-29719042

ABSTRACT

Motor endplates naturally undergo continual morphological changes that are altered in response to changes in neuromuscular activity. This study examines the consequences of acute (6-12 hr) disuse following hindlimb suspension on rat soleus muscle endplate structural stability. We identify early changes in several key signaling events including markers of protein kinase activation, AMPK phosphorylation and autophagy markers which may play a role in endplate remodeling. Acute disuse does not change endplate fragmentation, however, it decreases both the individual fragments and the total endplate area. This decrease was accompanied by an increase in the mean fluorescence intensity from the nicotinic acetylcholine receptors which compensate the endplate area loss. Muscle disuse decreased phosphorylation of AMPK and its substrate ACC, and stimulated mTOR controlled protein synthesis pathway and stimulated autophagy. Our findings provide evidence that changes in endplate stability are accompanied by reduced AMPK phosphorylation and an increase in autophagy markers, and these changes are evident within hours of onset of skeletal muscle disuse.


Subject(s)
Hindlimb Suspension/physiology , Motor Endplate/genetics , Protein Kinases/genetics , TOR Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Animals , Autophagy/physiology , Hindlimb/metabolism , Hindlimb/physiology , Motor Endplate/growth & development , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Phosphorylation , Protein Kinases/biosynthesis , Rats , Receptors, Nicotinic/genetics , Signal Transduction/genetics
3.
Neurosci Bull ; 34(3): 419-437, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29356943

ABSTRACT

A previous study has indicated that Krüppel-like factor 7 (KLF7), a transcription factor that stimulates Schwann cell (SC) proliferation and axonal regeneration after peripheral nerve injury, is a promising therapeutic transcription factor in nerve injury. We aimed to identify whether inhibition of microRNA-146b (miR-146b) affected SC proliferation, migration, and myelinated axon regeneration following sciatic nerve injury by regulating its direct target KLF7. SCs were transfected with miRNA lentivirus, miRNA inhibitor lentivirus, or KLF7 siRNA lentivirus in vitro. The expression of miR146b and KLF7, as well as SC proliferation and migration, were subsequently evaluated. In vivo, an acellular nerve allograft (ANA) followed by injection of GFP control vector or a lentiviral vector encoding an miR-146b inhibitor was used to assess the repair potential in a model of sciatic nerve gap. miR-146b directly targeted KLF7 by binding to the 3'-UTR, suppressing KLF7. Up-regulation of miR-146b and KLF7 knockdown significantly reduced the proliferation and migration of SCs, whereas silencing miR-146b resulted in increased proliferation and migration. KLF7 protein was localized in SCs in which miR-146b was expressed in vivo. Similarly, 4 weeks after the ANA, anti-miR-146b increased KLF7 and its target gene nerve growth factor cascade, promoting axonal outgrowth. Closer analysis revealed improved nerve conduction and sciatic function index score, and enhanced expression of neurofilaments, P0 (anti-peripheral myelin), and myelinated axon regeneration. Our findings provide new insight into the regulation of KLF7 by miR-146b during peripheral nerve regeneration and suggest a potential therapeutic strategy for peripheral nerve injury.


Subject(s)
Gene Expression Regulation/physiology , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , Nerve Regeneration/physiology , Sciatic Neuropathy/therapy , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Disease Models, Animal , Female , Ganglia, Spinal/cytology , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/genetics , Male , MicroRNAs/genetics , Motor Endplate/genetics , Myelin P0 Protein/metabolism , Nerve Regeneration/genetics , Nerve Tissue Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/surgery
4.
J Neurosci ; 36(11): 3254-67, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26985035

ABSTRACT

Patients with Charcot-Marie-Tooth Type 2D (CMT2D), caused by dominant mutations in Glycl tRNA synthetase (GARS), present with progressive weakness, consistently in the hands, but often in the feet also. Electromyography shows denervation, and patients often report that early symptoms include cramps brought on by cold or exertion. Based on reported clinical observations, and studies of mouse models of CMT2D, we sought to determine whether weakened synaptic transmission at the neuromuscular junction (NMJ) is an aspect of CMT2D. Quantal analysis of NMJs in two different mouse models of CMT2D (Gars(P278KY), Gars(C201R)), found synaptic deficits that correlated with disease severity and progressed with age. Results of voltage-clamp studies revealed presynaptic defects characterized by: (1) decreased frequency of spontaneous release without any change in quantal amplitude (miniature endplate current), (2) reduced amplitude of evoked release (endplate current) and quantal content, (3) age-dependent changes in the extent of depression in response to repetitive stimulation, and (4) release failures at some NMJs with high-frequency, long-duration stimulation. Drugs that modify synaptic efficacy were tested to see whether neuromuscular performance improved. The presynaptic action of 3,4 diaminopyridine was not beneficial, whereas postsynaptic-acting physostigmine did improve performance. Smaller mutant NMJs with correspondingly fewer vesicles and partial denervation that eliminates some release sites also contribute to the reduction of release at a proportion of mutant NMJs. Together, these voltage-clamp data suggest that a number of release processes, while essentially intact, likely operate suboptimally at most NMJs of CMT2D mice. SIGNIFICANCE STATEMENT: We have uncovered a previously unrecognized aspect of axonal Charcot-Marie-Tooth disease in mouse models of CMT2D. Synaptic dysfunction contributes to impaired neuromuscular performance and disease progression. This suggests that drugs which improve synaptic efficacy at the NMJ could be considered in treating the pathophysiology of CMT2D patients.


Subject(s)
Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Glycine-tRNA Ligase/genetics , Mutation/genetics , Neuromuscular Junction/pathology , Synaptic Transmission/genetics , Age Factors , Aminopyridines/pharmacology , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/physiopathology , Electric Stimulation , Imaging, Three-Dimensional , Mice , Mice, Transgenic , Motor Endplate/genetics , Motor Endplate/physiopathology , Muscle Strength/genetics , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/drug effects , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Patch-Clamp Techniques , Receptors, Cholinergic/metabolism , Synaptic Potentials/drug effects , Synaptic Potentials/genetics , Synaptic Vesicles/pathology , Synaptic Vesicles/ultrastructure
5.
Hum Mol Genet ; 24(12): 3440-56, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25765661

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that primarily affects motoneurons in the brain and spinal cord. Astrocyte and microglia activation as well as skeletal muscle atrophy are also typical hallmarks of the disease. However, the functional relationship between astrocytes, microglia and skeletal muscle in the pathogenic process remains unclear. Here, we report that the tumor necrosis factor-like weak inducer of apoptosis (Tweak) and its receptor Fn14 are aberrantly expressed in spinal astrocytes and skeletal muscle of SOD1(G93A) mice. We show that Tweak induces motoneuron death, stimulates astrocytic interleukin-6 release and astrocytic proliferation in vitro. The genetic ablation of Tweak in SOD1(G93A) mice significantly reduces astrocytosis, microgliosis and ameliorates skeletal muscle atrophy. The peripheral neutralization of Tweak through antagonistic anti-Tweak antibody ameliorates muscle pathology and notably, decreases microglial activation in SOD1(G93A) mice. Unexpectedly, none of these approaches improved motor function, lifespan and motoneuron survival. Our work emphasizes the multi-systemic aspect of ALS, and suggests that a combinatorial therapy targeting multiple cell types will be instrumental to halt the neurodegenerative process.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Gliosis/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Tumor Necrosis Factors/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Cell Death , Cell Proliferation , Cytokine TWEAK , Disease Models, Animal , Gene Deletion , Gene Expression Regulation , Interleukin-6/biosynthesis , Life Expectancy , Mice , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Motor Endplate/genetics , Motor Endplate/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation , Receptors, Cell Surface/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , TWEAK Receptor , Tumor Necrosis Factors/metabolism , Up-Regulation
6.
Eur J Hum Genet ; 23(9): 1151-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25537362

ABSTRACT

Fetal akinesia deformation sequence (FADS) refers to a clinically and genetically heterogeneous group of disorders with congenital malformations related to impaired fetal movement. FADS can result from mutations in CHRNG, CHRNA1, CHRND, DOK7 and RAPSN; however, these genes only account for a minority of cases. Here we identify MUSK as a novel cause of lethal FADS. Fourteen affected fetuses from a Dutch genetic isolate were traced back to common ancestors 11 generations ago. Homozygosity mapping in two fetuses revealed MUSK as a candidate gene. All tested cases carried an identical homozygous variant c.1724T>C; p.(Ile575Thr) in the intracellular domain of MUSK. The carrier frequency in the genetic isolate was 8%, exclusively found in heterozygous carriers. Consistent with the established role of MUSK as a tyrosine kinase that orchestrates neuromuscular synaptogenesis, the fetal myopathy was accompanied by impaired acetylcholine receptor clustering and reduced tyrosine kinase activity at motor nerve endings. A functional assay in myocytes derived from human fetuses confirmed that the variant blocks MUSK-dependent motor endplate formation. Taken together, the results strongly support a causal role of this founder mutation in MUSK, further expanding the gene set associated with FADS and offering new opportunities for prenatal genetic testing.


Subject(s)
Arthrogryposis/genetics , Founder Effect , Motor Endplate/genetics , Mutation , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Cholinergic/genetics , Alleles , Amino Acid Sequence , Arthrogryposis/diagnosis , Arthrogryposis/pathology , Base Sequence , Female , Fetus , Gene Expression , Gene Frequency , Genes, Lethal , Genetic Testing , Homozygote , Humans , Male , Molecular Sequence Data , Motor Endplate/pathology , Muscle Cells/metabolism , Muscle Cells/pathology , Netherlands , Pedigree , Prenatal Diagnosis , Primary Cell Culture , Receptors, Cholinergic/chemistry
7.
PLoS One ; 8(11): e78342, 2013.
Article in English | MEDLINE | ID: mdl-24260111

ABSTRACT

In vertebrates, nerve muscle communication is mediated by the release of the neurotransmitter acetylcholine packed inside synaptic vesicles by a specific vesicular acetylcholine transporter (VAChT). Here we used a mouse model (VAChT KD(HOM)) with 70% reduction in the expression of VAChT to investigate the morphological and functional consequences of a decreased acetylcholine uptake and release in neuromuscular synapses. Upon hypertonic stimulation, VAChT KD(HOM) mice presented a reduction in the amplitude and frequency of miniature endplate potentials, FM 1-43 staining intensity, total number of synaptic vesicles and altered distribution of vesicles within the synaptic terminal. In contrast, under electrical stimulation or no stimulation, VAChT KD(HOM) neuromuscular junctions did not differ from WT on total number of vesicles but showed altered distribution. Additionally, motor nerve terminals in VAChT KD(HOM) exhibited small and flattened synaptic vesicles similar to that observed in WT mice treated with vesamicol that blocks acetylcholine uptake. Based on these results, we propose that decreased VAChT levels affect synaptic vesicle biogenesis and distribution whereas a lower ACh content affects vesicles shape.


Subject(s)
Acetylcholine/metabolism , Motor Endplate/metabolism , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism , Acetylcholine/genetics , Animals , Electric Stimulation , Mice , Mice, Knockout , Motor Endplate/genetics , Motor Endplate/ultrastructure , Synaptic Vesicles/genetics , Synaptic Vesicles/ultrastructure , Vesicular Acetylcholine Transport Proteins/genetics
8.
BMC Neurosci ; 14: 81, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23914941

ABSTRACT

BACKGROUND: Spinal cord injury (SCI) results in muscle atrophy and a shift of slow oxidative to fast glycolytic fibers. Electrical stimulation (ES) at least partially restores muscle mass and fiber type distribution. The objective of this study was to was to characterize the early molecular adaptations that occur in rat soleus muscle after initiating isometric resistance exercise by ES for one hour per day for 1, 3 or 7 days when ES was begun 16 weeks after SCI. Additionally, changes in mRNA levels after ES were compared with those induced in soleus at the same time points after gastrocnemius tenotomy (GA). RESULTS: ES increased expression of Hey1 and Pitx2 suggesting increased Notch and Wnt signaling, respectively, but did not normalize RCAN1.4, a measure of calcineurin/NFAT signaling, or PGC-1ß mRNA levels. ES increased PGC-1α expression but not that of slow myofibrillar genes. Microarray analysis showed that after ES, genes coding for calcium binding proteins and nicotinic acetylcholine receptors were increased, and the expression of genes involved in blood vessel formation and morphogenesis was altered. Of the 165 genes altered by ES only 16 were also differentially expressed after GA, of which 12 were altered in the same direction by ES and GA. In contrast to ES, GA induced expression of genes related to oxidative phosphorylation. CONCLUSIONS: Notch and Wnt signaling may be involved in ES-induced increases in the mass of paralyzed muscle. Molecular adaptations of paralyzed soleus to resistance exercise are delayed or defective compared to normally innervated muscle.


Subject(s)
Electric Stimulation Therapy , Gene Expression Regulation , Motor Endplate/genetics , Muscle, Skeletal/metabolism , Spinal Cord Injuries/genetics , Spinal Cord Injuries/therapy , Wnt Signaling Pathway , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium/metabolism , Female , Homeodomain Proteins/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Rats , Rats, Wistar , Repressor Proteins/metabolism , Transcription Factors/metabolism , Homeobox Protein PITX2
9.
J Mol Biol ; 414(4): 477-84, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22037585

ABSTRACT

While mutations in the myosin subfragment 1 motor domain can directly disrupt the generation and transmission of force along myofibrils and lead to myopathy, the mechanism whereby mutations in the myosin rod influences mechanical function is less clear. Here, we used a combination of various imaging techniques and molecular dynamics simulations to test the hypothesis that perturbations in the myosin rod can disturb normal sarcomeric uniformity and, like motor domain lesions, would influence force production and propagation. We show that disrupting the rod can alter its nanomechanical properties and, in vivo, can drive asymmetric myofilament and sarcomere formation. Our imaging results indicate that myosin rod mutations likely disturb production and/or propagation of contractile force. This provides a unifying theory where common pathological cascades accompany both myosin motor and specific rod domain mutations. Finally, we suggest that sarcomeric inhomogeneity, caused by asymmetric thick filaments, could be a useful index of myopathic dysfunction.


Subject(s)
Motor Endplate/physiology , Muscular Diseases/physiopathology , Myosin Subfragments/physiology , Sarcomeres/physiology , Humans , Models, Molecular , Motor Endplate/genetics , Muscle Contraction , Muscular Diseases/genetics , Muscular Diseases/pathology , Mutation , Myosin Subfragments/chemistry , Myosin Subfragments/genetics , Myosin Subfragments/ultrastructure , Sarcomeres/chemistry , Sarcomeres/genetics , Sarcomeres/ultrastructure
10.
J Bioenerg Biomembr ; 43(6): 581-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22089637

ABSTRACT

Mitochondria contribute to neuronal function not only via their ability to generate ATP, but also via their ability to buffer large Ca(2+) loads. This review summarizes evidence that mitochondrial Ca(2+) sequestration is especially important for sustaining the function of vertebrate motor nerve terminals during repetitive stimulation. Motor terminal mitochondria can sequester large amounts of Ca(2+) because they have mechanisms for limiting both the mitochondrial depolarization and the increase in matrix free [Ca(2+)] associated with Ca(2+) influx. In mice expressing mutations of human superoxide dismutase -1 (SOD1) that cause some cases of familial amyotrophic lateral sclerosis (fALS), motor terminals degenerate well before the death of motor neuron cell bodies. This review presents evidence for early and progressive mitochondrial dysfunction in motor terminals of mutant SOD1 mice (G93A, G85R). This dysfunction would impair mitochondrial ability to sequester stimulation-associated Ca(2+) loads, and thus likely contributes to the early degeneration of motor terminals.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Genetic Diseases, Inborn/enzymology , Mitochondria/enzymology , Motor Endplate/enzymology , Motor Neurons/enzymology , Superoxide Dismutase/metabolism , Amino Acid Substitution , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Calcium/metabolism , Disease Models, Animal , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Humans , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/pathology , Motor Endplate/genetics , Motor Endplate/pathology , Motor Neurons/pathology , Mutation , Mutation, Missense , Superoxide Dismutase/genetics , Superoxide Dismutase-1
11.
Muscle Nerve ; 44(5): 789-94, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21952943

ABSTRACT

INTRODUCTION: Congenital myasthenic syndromes (CMS) are disabling but treatable disorders. Anticholinesterase therapy is effective in most of them, but is contraindicated in endplate (EP) acetylcholinesterase (AChE) deficiency, the slow-channel syndrome, Dok-7 myasthenia, and ß(2) -laminin deficiency, and is not useful in CMS due to defects in muscle-specific kinase (MuSK), agrin, and plectin. EP AChE, Dok-7, and ß(2)-laminin deficiencies respond favorably to ephedrine, but ephedrine can no longer be prescribed in the USA. METHODS: We used albuterol, another sympathomimetic agent, to treat 3 patients with EP AChE deficiency and 15 with Dok-7 myasthenia. Response to therapy was evaluated by a 9-point questionnaire pertaining to activities of daily life. RESULTS: Comparison of the pre- and posttreatment responses indicated a beneficial response to albuterol (P < 0.001) in both patient groups. The adverse effects of therapy were like those of ephedrine. CONCLUSION: Our observations should spur controlled, prospective clinical trials of albuterol in these as well as other CMS.


Subject(s)
Acetylcholinesterase/deficiency , Albuterol/therapeutic use , Motor Endplate/enzymology , Muscle Proteins , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/enzymology , Adolescent , Adult , Albuterol/pharmacology , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Motor Endplate/drug effects , Motor Endplate/genetics , Muscle Proteins/genetics , Myasthenic Syndromes, Congenital/genetics , Surveys and Questionnaires , Young Adult
12.
Cell Mol Neurobiol ; 31(1): 155-61, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21061159

ABSTRACT

The distribution of calcitonin gene-related peptide (CGRP) was examined in skeletal muscles of fore and hind limb as well as in oral and cranio-facial regions of the degenerating muscle (dmu) mouse, which harbours a null mutation in the voltage-gated sodium channel gene Scn8a. In limb, oral and cranio-facial muscles of wild type mice, only a few motor endplates contained CGRP-immunoreactivity. However, many CGRP-immunoreactive motor endplates appeared in the triceps brachii muscle, the biceps brachii muscle, the brachialis muscle, and the gastrocnemius muscle of dmu mice. CGRP-immunoreactive density of motor endplates in the skeletal muscles was also elevated by the mutation. In these muscles, the atrophy of muscle fibers could be detected and the density of cell nuclei in the musculature increased. In the flexor digitorum profundus muscle, the flexor digitorum superficialis muscle, and the soleus muscle as well as in oral and craniofacial muscles, however, the distribution of CGRP-immunoreactivity was barely affected by the mutation. The morphology of muscle fibers and the distribution of cell nuclei within them were also similar in wild type and dmu mice. In the lumbar spinal cord of dmu mice, CGRP-immunoreactive density of spinal motoneurons increased. These findings suggest that the atrophic degeneration in some fore and hind limb muscles of dmu mice may increase CGRP expression in their motoneurons.


Subject(s)
Calcitonin Gene-Related Peptide/genetics , Forelimb/metabolism , Hindlimb/metabolism , Motor Endplate/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Animals , Calcitonin Gene-Related Peptide/metabolism , Forelimb/pathology , Gene Expression , Hindlimb/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Motor Endplate/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Sodium Channels/genetics , Up-Regulation
13.
J Biol Chem ; 285(45): 34589-96, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20813841

ABSTRACT

The lifetime of nicotinic acetylcholine receptors (AChRs) in neuromuscular junctions (NMJs) is increased from <1 day to >1 week during early postnatal development. However, the exact timing of AChR stabilization is not known, and its correlation to the concurrent embryonic to adult AChR channel conversion, NMJ remodeling, and neuromuscular diseases is unclear. Using a novel time lapse in vivo imaging technology we show that replacement of the entire receptor population of an individual NMJ occurs end plate-specifically within hours. This makes it possible to follow directly in live animals changing stabilities of end plate receptors. In three different, genetically modified mouse models we demonstrate that the metabolic half-life values of synaptic AChRs increase from a few hours to several days after postnatal day 6. Developmental stabilization is independent of receptor subtype and apparently regulated by an intrinsic muscle-specific maturation program. Myosin Va, an F-actin-dependent motor protein, is also accumulated synaptically during postnatal development and thus could mediate the stabilization of end plate AChR.


Subject(s)
Aging/physiology , Motor Endplate/metabolism , Muscle Development/physiology , Receptors, Nicotinic/metabolism , Synapses/metabolism , Actins/genetics , Actins/metabolism , Animals , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Mice , Mice, Knockout , Motor Endplate/genetics , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Type V/genetics , Myosin Type V/metabolism , Neuromuscular Diseases/genetics , Neuromuscular Diseases/metabolism , Receptors, Nicotinic/genetics , Synapses/genetics
14.
Hum Mol Genet ; 17(24): 4022-35, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18806275

ABSTRACT

In humans and great apes, CHRNA1 encoding the muscle nicotinic acetylcholine receptor alpha subunit carries an inframe exon P3A, the inclusion of which yields a nonfunctional alpha subunit. In muscle, the P3A(-) and P3A(+) transcripts are generated in a 1:1 ratio but the functional significance and regulation of the alternative splicing remain elusive. An intronic mutation (IVS3-8G>A), identified in a patient with congenital myasthenic syndrome, disrupts an intronic splicing silencer (ISS) and results in exclusive inclusion of the downstream P3A exon. We found that the ISS-binding splicing trans-factor was heterogeneous nuclear ribonucleoprotein (hnRNP) H and the mutation attenuated the affinity of hnRNP for the ISS approximately 100-fold. We next showed that direct placement of hnRNP H to the 3' end of intron 3 silences, and siRNA-mediated downregulation of hnRNP H enhances recognition of exon P3A. Analysis of the human genome suggested that the hnRNPH-binding UGGG motif is overrepresented close to the 3' ends of introns. Pursuing this clue, we showed that alternative exons of GRIP1, FAS, VPS13C and NRCAM are downregulated by hnRNP H. Our findings imply that the presence of the hnRNP H-binding motif close to the 3' end of an intron is an essential but underestimated splicing regulator of the downstream exon.


Subject(s)
Exons/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/genetics , Myasthenic Syndromes, Congenital/genetics , Point Mutation/genetics , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , 3' Flanking Region/genetics , Adolescent , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Child, Preschool , Chlorocebus aethiops , Female , HeLa Cells , Humans , Introns/genetics , Male , Molecular Sequence Data , Motor Endplate/genetics , Motor Endplate/pathology , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/pathology , Pedigree , Protein Binding/genetics , RNA Splicing/genetics
15.
J Neurophysiol ; 100(4): 2361-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18650312

ABSTRACT

In many neuron types, the axon initial segment (AIS) has the lowest threshold for action potential generation. Its active properties are determined by the targeted expression of specific voltage-gated channel subunits. We show that the Na+ channel NaV1.6 displays a striking aggregation at the AIS of cortical neurons. To assess the functional role of this subunit, we used Scn8amed mice that are deficient for NaV1.6 subunits but still display prominent Na+ channel aggregation at the AIS. In CA1 pyramidal cells from Scn8amed mice, we found a depolarizing shift in the voltage dependence of activation of the transient Na+ current (INaT), indicating that NaV1.6 subunits activate at more negative voltages than other NaV subunits. Additionally, persistent and resurgent Na+ currents were significantly reduced. Current-clamp recordings revealed a significant elevation of spike threshold in Scn8amed mice as well as a shortening of the estimated delay between spike initiation at the AIS and its arrival at the soma. In combination with simulations using a realistic computer model of a CA1 pyramidal cell, our results imply that a hyperpolarized voltage dependence of activation of AIS NaV1.6 channels is important both in determining spike threshold and localizing spike initiation to the AIS. In addition to altered spike initiation, Scn8amed mice also showed a strongly reduced spike gain as expected with combined changes in persistent and resurgent currents and spike threshold. These results suggest that NaV1.6 subunits at the AIS contribute significantly to its role as spike trigger zone and shape repetitive discharge properties of CA1 neurons.


Subject(s)
Axons/physiology , Cerebral Cortex/physiology , Nerve Tissue Proteins/physiology , Pyramidal Cells/physiology , Sodium Channels/physiology , Action Potentials/physiology , Algorithms , Animals , Calcium Channels/physiology , Calcium Signaling/physiology , Cerebral Cortex/cytology , Computer Simulation , Electrophysiology , Immunohistochemistry , Membrane Potentials/physiology , Mice , Mice, Knockout , Models, Neurological , Models, Statistical , Motor Endplate/genetics , Motor Endplate/physiology , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Potassium Channels/physiology , Pyramidal Cells/ultrastructure , Sodium Channels/genetics , Temperature
16.
FEBS J ; 275(9): 2042-54, 2008 May.
Article in English | MEDLINE | ID: mdl-18384381

ABSTRACT

AChRepsilon(-/-) mice lack epsilon-subunits of the acetylcholine receptor and thus fail to express adult-type receptors. The expression of fetal-type receptors throughout postnatal life alters postsynaptic signal transduction and causes a fast-to-slow fiber type transition, both in slow-twitch soleus muscle and in fast-twitch extensor digitorum longus muscle. In comparison to wild-type muscle, the proportion of type 1 slow fibers is significantly increased (6%), whereas the proportion of fast fibers is reduced (in soleus, type 2A by 12%, and in extensor digitorum longus, type 2B/2D by 10%). The increased levels of troponin I(slow) transcripts clearly support a fast-to-slow fiber type transition. Shifts of protein and transcript levels are not restricted to 'myogenic' genes but also affect 'synaptogenic' genes. Clear increases are observed for acetylcholine receptor alpha-subunits and the postsynaptically located utrophin. Although the fast-to-slow fiber type transition appears to occur in a coordinated manner in both muscle types, muscle-specific differences are retained. Most prominently, the differential expression level of the synaptic regulator MuSK is significantly lower in extensor digitorum muscle than in soleus muscle. The results show a new quality in muscle plasticity, in that changes in the functional properties of endplate receptors modulate the contractile properties of skeletal muscles. Muscle thus represents a self-matching system that adjusts contractile properties and synaptic function to variable functional demands.


Subject(s)
Muscle Fibers, Fast-Twitch/chemistry , Muscle Fibers, Slow-Twitch/chemistry , Receptors, Cholinergic/metabolism , Animals , Gene Expression/physiology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Endplate/genetics , Motor Endplate/metabolism , Muscle Contraction/physiology , Muscle Fibers, Fast-Twitch/cytology , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/cytology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/physiology , Myosin Heavy Chains/analysis , Myosin Heavy Chains/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptors, Cholinergic/genetics
17.
Am J Pathol ; 170(2): 644-57, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17255332

ABSTRACT

The primary autoantigen in myasthenia gravis, the acetylcholine receptor (AChR), is clustered and anchored in the postsynaptic membrane of the neuromuscular junction by rapsyn. Previously, we found that overexpression of rapsyn by cDNA transfection protects AChRs in rat muscles from antibody-mediated loss in passive transfer experimental autoimmune myasthenia gravis (EAMG). Here, we determined whether rapsyn overexpression can reduce or even reverse AChR loss in muscles that are already damaged by chronic EAMG, which mimics the human disease. Active immunization against purified AChR was performed in female Lewis rats. Rapsyn overexpression resulted in an increase in total muscle membrane AChR levels, with some AChR at neuromuscular junctions but much of it in extrasynaptic membrane regions. At the ultrastructural level, most endplates in rapsyn-treated chronic EAMG muscles showed increased damage to the postsynaptic membrane. Although rapsyn overexpression stabilized AChRs in intact or mildly damaged endplates, the rapsyn-induced increase of membrane AChR enhanced autoantibody binding and membrane damage in severe ongoing disease. Thus, these results show the complexity of synaptic stabilization of AChR during the autoantibody attack. They also indicate that the expression of receptor-associated proteins may determine the severity of autoimmune diseases caused by anti-receptor antibodies.


Subject(s)
Gene Expression , Motor Endplate/metabolism , Muscle Proteins/biosynthesis , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Receptors, Cholinergic/metabolism , Synaptic Membranes/metabolism , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , Chronic Disease , Female , Humans , Motor Endplate/genetics , Motor Endplate/immunology , Motor Endplate/ultrastructure , Muscle Proteins/genetics , Muscle Proteins/immunology , Myasthenia Gravis, Autoimmune, Experimental/genetics , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/pathology , Rats , Rats, Inbred Lew , Receptors, Cholinergic/immunology , Synaptic Membranes/immunology
18.
Neurology ; 66(8): 1223-9, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16525123

ABSTRACT

BACKGROUND: Most congenital myasthenic syndromes are caused by defects in postsynaptic or synaptic basal lamina-associated proteins; congenital myasthenic syndromes (CMSs) associated with presynaptic defects are uncommon. Here, the authors describe clinical, electrophysiologic, and morphologic features of two novel and highly disabling CMSs, one determined by presynaptic and the other determined by combined presynaptic and postsynaptic defects. METHODS: Microelectrode, single channel patch clamp, immunocytochemical, [(125)I]alpha-bungarotoxin binding, and quantitative electron microscopy studies of endplates were performed. Candidate genes were directly sequenced. RESULTS: Patient 1, a 7-year-old boy, had severe myasthenic symptoms since infancy. Patient 2, a 48-year-old man, had delayed motor milestones and became progressively weaker after age 2 years. Both used wheelchairs and had a 30-50% EMG decrement on 2-Hz stimulation. Evoked quantal release was reduced to approximately 25% of normal in both. In Patient 2, the synaptic response to acetylcholine was further compromised by degeneration of the junctional folds with concomitant loss of the acetylcholine receptor (AChR). A search for mutations in components of the synaptic vesicle release complex and in other candidate proteins failed to identify the molecular basis of the two syndromes. CONCLUSIONS: Combined clinical, morphologic, and in vitro electrophysiologic findings define two novel congenital myasthenic syndromes. The molecular basis of these syndromes awaits discovery.


Subject(s)
Acetylcholinesterase/deficiency , Evoked Potentials , Myasthenic Syndromes, Congenital/physiopathology , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Receptors, Cholinergic/deficiency , Acetylcholinesterase/chemistry , Acetylcholinesterase/genetics , Child , Evoked Potentials/genetics , Humans , Male , Middle Aged , Motor Endplate/genetics , Motor Endplate/physiopathology , Motor Endplate/ultrastructure , Mutation , Myasthenic Syndromes, Congenital/enzymology , Myasthenic Syndromes, Congenital/genetics , Nerve Degeneration/enzymology , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Presynaptic Terminals/enzymology , Presynaptic Terminals/ultrastructure , Protein Conformation , Receptors, Cholinergic/chemistry , Receptors, Cholinergic/genetics , Synaptic Vesicles/enzymology , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure
19.
Neuron ; 41(5): 701-10, 2004 Mar 04.
Article in English | MEDLINE | ID: mdl-15003170

ABSTRACT

Migraine is a common, disabling, multifactorial, episodic neurovascular disorder of unknown etiology. Familial hemiplegic migraine type 1 (FHM-1) is a Mendelian subtype of migraine with aura that is caused by missense mutations in the CACNA1A gene that encodes the alpha(1) subunit of neuronal Ca(v)2.1 Ca(2+) channels. We generated a knockin mouse model carrying the human pure FHM-1 R192Q mutation and found multiple gain-of-function effects. These include increased Ca(v)2.1 current density in cerebellar neurons, enhanced neurotransmission at the neuromuscular junction, and, in the intact animal, a reduced threshold and increased velocity of cortical spreading depression (CSD; the likely mechanism for the migraine aura). Our data show that the increased susceptibility for CSD and aura in migraine may be due to cortical hyperexcitability. The R192Q FHM-1 mouse is a promising animal model to study migraine mechanisms and treatments.


Subject(s)
Calcium Channels/genetics , Cortical Spreading Depression/genetics , Disease Models, Animal , Genetic Predisposition to Disease , Migraine with Aura/genetics , Recombination, Genetic , Animals , Calcium Channels/biosynthesis , Calcium Channels, N-Type , Calcium Channels, P-Type , Calcium Channels, Q-Type , Cells, Cultured , Female , Humans , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Migraine with Aura/metabolism , Motor Endplate/genetics , Motor Endplate/metabolism , Mutation
20.
Exp Neurol ; 185(2): 290-6, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14736510

ABSTRACT

Uncertainties from the literature concerning the role of apolipoprotein E (apoE) in central cholinergic function prompted us to investigate what effect apoE may have on transmission at the neuromuscular junction. Both spontaneous and evoked release were measured in isolated extensor digitorum longus (edl) and soleus muscles from both wild-type and apoE-deficient mice. Miniature endplate and nerve-evoked endplate potentials (MEPPs and EPPs, respectively) were indistinguishable in edl muscles in both groups of mice; however, MEPP amplitudes in soleus muscles were significantly larger (by an average of 23%) in apoE-deficient mice compared with 5- to 7-week-old age-matched wild-type mice. The EPP amplitudes were also larger in soleus muscles in the mutant mice, but this was a reflection of the larger quantal size in this muscle because quantal content, determined from the ratio of the average EPP amplitude to average MEPP amplitude, was unchanged from normal in the mutant mice. The MEPP frequency and the percent of nerve stimulations failing to produce an EPP were unchanged from normal in both muscle types in the mutant mice. The difference in quantal size in soleus muscle transmission between mutant and wild-type mice was abolished in the presence of neostigmine, an acetylcholinesterase inhibitor. The results suggest that apoE normally associates with acetylcholinesterase in the synaptic cleft of slow muscles, modulating the activity of the enzyme and therefore quantal size.


Subject(s)
Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Motor Endplate/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Acetylcholinesterase/metabolism , Animals , Evoked Potentials, Motor/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Endplate/enzymology , Motor Endplate/genetics , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Neuromuscular Junction/enzymology , Neuromuscular Junction/metabolism , Synapses/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...