Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Publication year range
1.
Am J Med Genet A ; 185(4): 1182-1186, 2021 04.
Article in English | MEDLINE | ID: mdl-33381903

ABSTRACT

The heterozygous deletion of 15q13.3 is a recurrently observed microdeletion syndrome associated with a relatively mild phenotype including learning disability and language impairment. In contrast, the homozygous deletion of 15q13.3 is extremely rare and is associated with a much severer phenotype that includes epileptic encephalopathy, profound intellectual disability, and hypotonia. Which of the genes within the deleted interval is responsible for the more severe features when biallelically deleted is currently unknown. Here, we report a patient with profound hypotonia, severe intellectual disability, and seizures who had biallelic loss-of-function variants in OTUD7A: a 15q13.3 deletion including the OTUD7A locus, and a frameshift OTUD7A variant c.1125del, p.(Glu375Aspfs*11). Unexpectedly, both aberrations occurred de novo. Our experiment using Caenorhabditis elegans showed that worms carrying a corresponding homozygous variant in the homolog OTUB-2 exhibited weakened muscle contraction suggestive of aberrant neuromuscular transmission. We concluded that the biallelic complete loss of OTUD7A in humans represents a presumably new autosomal recessive disorder characterized by profound hypotonia, severe intellectual disability, and seizures.


Subject(s)
Deubiquitinating Enzymes/genetics , Intellectual Disability/genetics , Muscle Hypotonia/genetics , Neuromuscular Junction Diseases/embryology , Animals , Caenorhabditis elegans/genetics , Child, Preschool , Frameshift Mutation/genetics , Homozygote , Humans , Intellectual Disability/complications , Intellectual Disability/physiopathology , Loss of Heterozygosity/genetics , Male , Muscle Contraction/genetics , Muscle Contraction/physiology , Muscle Hypotonia/physiopathology , Neuromuscular Junction Diseases/complications , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/physiopathology , Seizures/complications , Seizures/genetics , Seizures/physiopathology , Thiolester Hydrolases/genetics
2.
Int J Mol Sci ; 22(5)2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33671084

ABSTRACT

: Signal transduction at the neuromuscular junction (NMJ) is affected in many human diseases, including congenital myasthenic syndromes (CMS), myasthenia gravis, Lambert-Eaton myasthenic syndrome, Isaacs' syndrome, Schwartz-Jampel syndrome, Fukuyama-type congenital muscular dystrophy, amyotrophic lateral sclerosis, and sarcopenia. The NMJ is a prototypic cholinergic synapse between the motor neuron and the skeletal muscle. Synaptogenesis of the NMJ has been extensively studied, which has also been extrapolated to further understand synapse formation in the central nervous system. Studies of genetically engineered mice have disclosed crucial roles of secreted molecules in the development and maintenance of the NMJ. In this review, we focus on the secreted signaling molecules which regulate the clustering of acetylcholine receptors (AChRs) at the NMJ. We first discuss the signaling pathway comprised of neural agrin and its receptors, low-density lipoprotein receptor-related protein 4 (Lrp4) and muscle-specific receptor tyrosine kinase (MuSK). This pathway drives the clustering of acetylcholine receptors (AChRs) to ensure efficient signal transduction at the NMJ. We also discuss three secreted molecules (Rspo2, Fgf18, and connective tissue growth factor (Ctgf)) that we recently identified in the Wnt/ß-catenin and fibroblast growth factors (FGF) signaling pathways. The three secreted molecules facilitate the clustering of AChRs by enhancing the agrin-Lrp4-MuSK signaling pathway.


Subject(s)
Muscle Proteins/metabolism , Neuromuscular Agents/metabolism , Neuromuscular Junction Diseases/physiopathology , Neuromuscular Junction/physiology , Animals , Humans , Signal Transduction
3.
J Neurophysiol ; 123(5): 1864-1869, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32292122

ABSTRACT

Early-onset hypertonia is characteristic of developmental neuromotor disorders, including cerebral palsy (CP). The spa transgenic mouse displays early-onset spasticity, abnormal gait, and motor impairments that are remarkably similar to symptoms of human CP. Previously, we showed that spa mice have fewer motor neurons innervating the tibialis anterior (TA). An expanded innervation ratio may result in increased susceptibility to neuromuscular transmission failure (NMTF). We assessed NMTF in an ex vivo TA muscle nerve preparation from spa and wild-type (WT) mice by comparing forces elicited by nerve versus muscle stimulation. TA muscle innervation ratio was assessed by counting the number of muscle fibers and dividing by the number of TA motor neurons. Muscle fiber cross-sectional areas were also assessed in the TA muscle. We observed that NMTF was immediately present in spa mice, increased with repetitive stimulation, and associated with increased innervation ratio. These changes were concomitant with reduced TA muscle fiber cross-sectional area in spa mice compared with WT. Early-onset hypertonia is associated with increased innervation ratio and impaired neuromuscular transmission. These disturbances may exacerbate the underlying gait abnormalities present in individuals with hypertonia.NEW & NOTEWORTHY Nerve-muscle interaction is poorly understood in the context of early-onset spasticity and hypertonia. In an animal model of early-onset spasticity, spa mice, we found a marked impairment of tibialis anterior neuromuscular transmission. This impairment is associated with an increased innervation ratio (mean number of muscle fibers innervated by a single motor neuron). These disturbances may underlie weakness and gait disturbances observed in individual with developmental hypertonia and spasticity.


Subject(s)
Motor Neurons/physiology , Muscle Hypertonia/physiopathology , Muscle, Skeletal/innervation , Muscle, Skeletal/physiopathology , Neuromuscular Junction/physiopathology , Synaptic Transmission/physiology , Animals , Cerebral Palsy/physiopathology , Disease Models, Animal , Female , Gait Disorders, Neurologic/physiopathology , Male , Mice, Knockout , Muscle Fibers, Skeletal/physiology , Neuromuscular Junction Diseases/physiopathology
4.
Am J Med Genet A ; 173(8): 2240-2245, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28544784

ABSTRACT

Defects in genes encoding the isoforms of the laminin alpha subunit have been linked to various phenotypic manifestations, including brain malformations, muscular dystrophy, ocular defects, cardiomyopathy, and skin abnormalities. We report here a severe defect of neuromuscular transmission in a consanguineous patient with a homozygous variant in the laminin alpha-5 subunit gene (LAMA5). The variant c.8046C>T (p.Arg2659Trp) is rare and has a predicted deleterious effect. The affected individual, who also carries a rare homozygous sequence variant in LAMA1, had muscle weakness, myopia, and facial tics. Magnetic resonance imaging of brain showed mild volume loss and periventricular T2 prolongation. Repetitive nerve stimulation revealed 50% decrement of compound muscle action potential amplitudes and 250% facilitation immediately after exercise, Endplate studies identified a profound reduction of the endplate potential quantal content and endplates with normal postsynaptic folding that were denuded or partially occupied by small nerve terminals. Expression studies revealed that p.Arg2659Trp caused decreased binding of laminin alpha-5 to SV2A and impaired laminin-521 cell-adhesion and cell projection support in primary neuronal cultures. In summary, this report describing severe neuromuscular transmission failure in a patient with a LAMA5 mutation expands the list of phenotypes associated with defects in genes encoding alpha-laminins.


Subject(s)
Laminin/genetics , Myasthenic Syndromes, Congenital/genetics , Neuromuscular Junction Diseases/genetics , Adult , Face/diagnostic imaging , Face/physiopathology , Female , Homozygote , Humans , Myasthenic Syndromes, Congenital/complications , Myasthenic Syndromes, Congenital/diagnostic imaging , Myasthenic Syndromes, Congenital/physiopathology , Myopia/complications , Myopia/diagnostic imaging , Myopia/genetics , Myopia/physiopathology , Neuromuscular Junction Diseases/complications , Neuromuscular Junction Diseases/diagnostic imaging , Neuromuscular Junction Diseases/physiopathology , Tics/complications , Tics/diagnostic imaging , Tics/genetics , Tics/physiopathology , Young Adult
5.
Proc Natl Acad Sci U S A ; 111(46): 16556-61, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368159

ABSTRACT

The motoneural control of skeletal muscle contraction requires the neuromuscular junction (NMJ), a midmuscle synapse between the motor nerve and myotube. The formation and maintenance of NMJs are orchestrated by the muscle-specific receptor tyrosine kinase (MuSK). Motor neuron-derived agrin activates MuSK via binding to MuSK's coreceptor Lrp4, and genetic defects in agrin underlie a congenital myasthenic syndrome (an NMJ disorder). However, MuSK-dependent postsynaptic differentiation of NMJs occurs in the absence of a motor neuron, indicating a need for nerve/agrin-independent MuSK activation. We previously identified the muscle protein Dok-7 as an essential activator of MuSK. Although NMJ formation requires agrin under physiological conditions, it is dispensable for NMJ formation experimentally in the absence of the neurotransmitter acetylcholine, which inhibits postsynaptic specialization. Thus, it was hypothesized that MuSK needs agrin together with Lrp4 and Dok-7 to achieve sufficient activation to surmount inhibition by acetylcholine. Here, we show that forced expression of Dok-7 in muscle enhanced MuSK activation in mice lacking agrin or Lrp4 and restored midmuscle NMJ formation in agrin-deficient mice, but not in Lrp4-deficient mice, probably due to the loss of Lrp4-dependent presynaptic differentiation. However, these NMJs in agrin-deficient mice rapidly disappeared after birth, and postsynaptic specializations emerged ectopically throughout myotubes whereas exogenous Dok-7-mediated MuSK activation was maintained. These findings demonstrate that the MuSK activator agrin plays another role essential for the postnatal maintenance, but not for embryonic formation, of NMJs and also for the postnatal, but not prenatal, midmuscle localization of postsynaptic specializations, providing physiological and pathophysiological insight into NMJ homeostasis.


Subject(s)
Agrin/physiology , Neuromuscular Junction/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Agrin/deficiency , Agrin/genetics , Alternative Splicing , Animals , Diaphragm/embryology , Diaphragm/growth & development , Enzyme Activation , Female , LDL-Receptor Related Proteins , Longevity/genetics , Male , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/physiology , Muscle Proteins/deficiency , Muscle Proteins/physiology , Neuromuscular Junction/embryology , Neuromuscular Junction/growth & development , Neuromuscular Junction Diseases/enzymology , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/physiopathology , Phosphorylation , Post-Synaptic Density/physiology , Protein Processing, Post-Translational , Receptors, Cholinergic/physiology , Receptors, LDL/deficiency , Receptors, LDL/physiology , Recombinant Fusion Proteins/metabolism , Rotarod Performance Test
6.
Anesth Analg ; 121(2): 366-72, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25902322

ABSTRACT

BACKGROUND: Postoperative residual neuromuscular blockade (NMB), defined as a train-of-four (TOF) ratio of <0.9, is an established risk factor for critical postoperative respiratory events and increased morbidity. At present, little is known about the occurrence of residual NMB in Canada. The RECITE (Residual Curarization and its Incidence at Tracheal Extubation) study was a prospective observational study at 8 hospitals in Canada investigating the incidence and severity of residual NMB. METHODS: Adult patients undergoing open or laparoscopic abdominal surgery expected to last <4 hours, ASA physical status I-III, and scheduled for general anesthesia with at least 1 dose of a nondepolarizing neuromuscular blocking agent for endotracheal intubation or maintenance of neuromuscular relaxation were enrolled in the study. Neuromuscular function was assessed using acceleromyography with the TOF-Watch SX. All reported TOF ratios were normalized to the baseline values. The attending anesthesiologist and all other observers were blinded to the TOF ratio (T4/T1) results. The primary and secondary objectives were to determine the incidence and severity of residual NMB (TOF ratio <0.9) just before tracheal extubation and at arrival at the postanesthesia care unit (PACU). RESULTS: Three hundred and two participants were enrolled. Data were available for 241 patients at tracheal extubation and for 207 patients at PACU arrival. Rocuronium was the NMB agent used in 99% of cases. Neostigmine was used for reversal of NMB in 73.9% and 72.0% of patients with TE and PACU data, respectively. The incidence of residual NMB was 63.5% (95% confidence interval, 57.4%-69.6%) at tracheal extubation and 56.5% (95% confidence interval, 49.8%-63.3%) at arrival at the PACU. In an exploratory analysis, no statistically significant differences were observed in the incidence of residual NMB according to gender, age, body mass index, ASA physical status, type of surgery, or comorbidities (all P > 0.13). CONCLUSIONS: Residual paralysis is common at tracheal extubation and PACU arrival, despite qualitative neuromuscular monitoring and the use of neostigmine. More effective detection and management of NMB is needed to reduce the risks associated with residual NMB.


Subject(s)
Androstanols/adverse effects , Neuromuscular Blockade/adverse effects , Neuromuscular Blocking Agents/adverse effects , Neuromuscular Junction Diseases/epidemiology , Neuromuscular Junction/drug effects , Paralysis/epidemiology , Abdomen/surgery , Adult , Airway Extubation , Anesthesia Recovery Period , Anesthesia, General , Antidotes/therapeutic use , Canada/epidemiology , Cholinesterase Inhibitors/therapeutic use , Female , Humans , Incidence , Laparoscopy , Male , Middle Aged , Neostigmine/therapeutic use , Neuromuscular Junction/physiopathology , Neuromuscular Junction Diseases/chemically induced , Neuromuscular Junction Diseases/diagnosis , Neuromuscular Junction Diseases/physiopathology , Neuromuscular Monitoring , Paralysis/chemically induced , Paralysis/diagnosis , Paralysis/physiopathology , Prospective Studies , Risk Factors , Rocuronium , Severity of Illness Index , Time Factors , Treatment Outcome
7.
Anesth Analg ; 121(2): 373-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25923435

ABSTRACT

BACKGROUND: Pipecuronium is a steroidal neuromuscular blocking agent. Sugammadex, a relaxant binding γ-cyclodextrin derivative, reverses the effect of rocuronium, vecuronium, and pancuronium. We investigated whether sugammadex reverses moderate pipecuronium-induced neuromuscular blockade (NMB) and the doses required to achieve reversal. METHODS: This single-center, randomized, double-blind, 5-group parallel-arm study comprised 50 patients undergoing general anesthesia with propofol, sevoflurane, fentanyl, and pipecuronium. Neuromuscular monitoring was performed with acceleromyography (TOF-Watch SX) according to international standards. When the NMB recovered spontaneously to train-of-four count 2, patients randomly received 1.0, 2.0, 3.0, or 4.0 mg/kg of sugammadex or placebo. Recovery time from sugammadex injection to normalized train-of-four (TOF) ratio 0.9 was the primary outcome variable. The recovery time from the sugammadex injection to final T1 was the secondary end point. Postoperative neuromuscular functions were also assessed. RESULTS: Each patient who received sugammadex recovered to a normalized TOF ratio of 0.9 within 5.0 minutes (95% lower confidence interval for the lowest dose 70.1%; for all doses 90.8%) and 79% of these patients reached a normalized TOF ratio 0.9 within 2.0 minutes (95% lower confidence interval for the lowest dose 26.7%; for all doses 63.7%). T1 recovered several minutes after the TOF ratio. No residual postoperative NMB was observed. CONCLUSIONS: Sugammadex adequately and rapidly reverses pipecuronium-induced moderate NMB during sevoflurane anesthesia. Once the train-of-four count has spontaneously returned to 2 responses following pipecuronium administration, a dose of 2.0 mg/kg of sugammadex is sufficient to reverse the NMB.


Subject(s)
Anesthesia, General/methods , Anesthetics, Inhalation , Antidotes/therapeutic use , Methyl Ethers , Neuromuscular Blockade/methods , Neuromuscular Blocking Agents/therapeutic use , Neuromuscular Junction Diseases/prevention & control , Neuromuscular Junction/drug effects , Pipecuronium/therapeutic use , gamma-Cyclodextrins/therapeutic use , Adult , Aged , Anesthesia Recovery Period , Double-Blind Method , Female , Humans , Hungary , Male , Middle Aged , Neuromuscular Blockade/adverse effects , Neuromuscular Blocking Agents/adverse effects , Neuromuscular Junction/physiopathology , Neuromuscular Junction Diseases/chemically induced , Neuromuscular Junction Diseases/diagnosis , Neuromuscular Junction Diseases/physiopathology , Neuromuscular Monitoring , Pipecuronium/adverse effects , Recovery of Function , Sevoflurane , Sugammadex , Time Factors , Treatment Outcome
8.
Muscle Nerve ; 50(5): 854-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24862941

ABSTRACT

INTRODUCTION: Small-amplitude, short-duration motor unit action potentials are non-specific findings seen in myopathies and neuromuscular junction (NMJ) disorders. NMJ studies (repetitive nerve stimulation and single-fiber electromyography) can determine if such findings are related to NMJ abnormalities but are not considered routinely in atypical cases. METHODS: Medical records of 338 patients with confirmed NMJ disorders were reviewed to identify cases with a clinical or electrodiagnostic impression of myopathy during initial evaluation. A history of muscle biopsy with findings that did not support a myopathic process was required for inclusion. RESULTS: Four patients met the inclusion criteria. NMJ studies were abnormal in all cases. One patient had elevated acetylcholine receptor antibodies. Three patients were antibody negative: 2 demonstrated immunotherapy responsiveness, and 1 had a Rapsyn mutation. CONCLUSIONS: NMJ disorders may mimic myopathies, and NMJ studies should be performed to clarify so-called "myopathic" electromyographic findings to avoid unnecessary testing and delayed diagnosis.


Subject(s)
Muscular Diseases/diagnosis , Muscular Diseases/physiopathology , Neuromuscular Junction Diseases/physiopathology , Adolescent , Antibodies/blood , Biopsy , Creatine Kinase/blood , Electric Stimulation , Electromyography , Female , Humans , Muscle, Skeletal/physiopathology , Receptors, Cholinergic/immunology , Retrospective Studies
9.
J Neurosci ; 32(15): 5074-84, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22496553

ABSTRACT

The actin-binding protein plastin 3 (PLS3) has been identified as a modifier of the human motoneuron disease spinal muscular atrophy (SMA). SMA is caused by decreased levels of the survival motor neuron protein (SMN) and in its most severe form causes death in infants and young children. To understand the mechanism of PLS3 in SMA, we have analyzed pls3 RNA and protein in zebrafish smn mutants. We show that Pls3 protein levels are severely decreased in smn(-/-) mutants without a reduction in pls3 mRNA levels. Moreover, we show that both pls3 mRNA and protein stability are unaffected when Smn is reduced. This indicates that SMN affects PLS3 protein production. We had previously shown that, in smn mutants, the presynaptic protein SV2 is decreased at neuromuscular junctions. Transgenically driving human PLS3 in motoneurons rescues the decrease in SV2 expression. To determine whether PLS3 could also rescue function, we performed behavioral analysis on smn mutants and found that they had a significant decrease in spontaneous swimming and turning. Driving PLS3 transgenically in motoneurons rescued both of these defects. These data show that PLS3 protein levels are dependent on SMN and that PLS3 is able to rescue the neuromuscular defects and corresponding movement phenotypes caused by low levels of Smn suggesting that decreased PLS3 contributes to SMA motor phenotypes.


Subject(s)
Cell Survival/physiology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Motor Neurons/physiology , Movement Disorders/genetics , Movement Disorders/physiopathology , Animals , Animals, Genetically Modified , Blotting, Western , Cell Line , DNA/biosynthesis , DNA/genetics , DNA, Antisense/pharmacology , Down-Regulation/physiology , Fluorescent Antibody Technique , Half-Life , Locomotion/physiology , Microscopy, Confocal , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/physiopathology , Polymerase Chain Reaction , Protein Processing, Post-Translational , RNA/biosynthesis , RNA/genetics , Terminology as Topic , Zebrafish
10.
Mol Ther ; 20(7): 1384-92, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22371845

ABSTRACT

Acetylcholinesterase (AChE) at the neuromuscular junction (NMJ) is anchored to the synaptic basal lamina via a triple helical collagen Q (ColQ). Congenital defects of ColQ cause endplate AChE deficiency and myasthenic syndrome. A single intravenous administration of adeno-associated virus serotype 8 (AAV8)-COLQ to Colq(-/-) mice recovered motor functions, synaptic transmission, as well as the morphology of the NMJ. ColQ-tailed AChE was specifically anchored to NMJ and its amount was restored to 89% of the wild type. We next characterized the molecular basis of this efficient recovery. We first confirmed that ColQ-tailed AChE can be specifically targeted to NMJ by an in vitro overlay assay in Colq(-/-) mice muscle sections. We then injected AAV1-COLQ-IRES-EGFP into the left tibialis anterior and detected AChE in noninjected limbs. Furthermore, the in vivo injection of recombinant ColQ-tailed AChE protein complex into the gluteus maximus muscle of Colq(-/-) mice led to accumulation of AChE in noninjected forelimbs. We demonstrated for the first time in vivo that the ColQ protein contains a tissue-targeting signal that is sufficient for anchoring itself to the NMJ. We propose that the protein-anchoring strategy is potentially applicable to a broad spectrum of diseases affecting extracellular matrix molecules.


Subject(s)
Acetylcholinesterase/metabolism , Collagen/genetics , Collagen/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neuromuscular Junction Diseases/therapy , Neuromuscular Junction/metabolism , Acetylcholinesterase/genetics , Animals , Dependovirus/genetics , Genetic Therapy , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/physiopathology , Synaptic Transmission
11.
Am J Physiol Endocrinol Metab ; 303(4): E551-61, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22739110

ABSTRACT

Diabetic neuropathy is associated with functional and morphological changes of the neuromuscular junction (NMJ) associated with muscle weakness. This study examines the effect of type 1 diabetes on NMJ function. Swiss Webster mice were made diabetic with three interdaily ip injections of streptozotocin (STZ). Mice were severely hyperglycemic within 7 days after the STZ treatment began. Whereas performance of mice on a rotating rod remained normal, the twitch tension response of the isolated extensor digitorum longus to nerve stimulation was reduced significantly at 4 wk after the onset of STZ-induced hyperglycemia. This mechanical alteration was associated with increased amplitude and prolonged duration of miniature end-plate currents (mEPCs). Prolongation of mEPCs was not due to expression of the embryonic acetylcholine receptor but to reduced muscle expression of acetylcholine esterase (AChE). Greater sensitivity of mEPC decay time to the selective butyrylcholinesterase (BChE) inhibitor PEC suggests that muscle attempts to compensate for reduced AChE levels by increasing expression of BChE. These alterations of AChE are attributed to STZ-induced hyperglycemia since similar mEPC prolongation and reduced AChE expression were found for db/db mice. The reduction of muscle end-plate AChE activity early during the onset of STZ-induced hyperglycemia may contribute to endplate pathology and subsequent muscle weakness during diabetes.


Subject(s)
Acetylcholinesterase/deficiency , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 1/enzymology , Diabetic Neuropathies/enzymology , Neuromuscular Junction Diseases/enzymology , Acetylcholinesterase/biosynthesis , Animals , Butyrylcholinesterase/biosynthesis , Cholinesterase Inhibitors/pharmacology , Diabetic Neuropathies/physiopathology , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/deficiency , Hyperglycemia/enzymology , Hyperglycemia/physiopathology , Male , Mice , Motor Endplate/enzymology , Motor Endplate/physiopathology , Muscle Weakness/enzymology , Muscle Weakness/physiopathology , Neuromuscular Junction Diseases/physiopathology , Physostigmine/analogs & derivatives , Physostigmine/pharmacology
12.
Ann N Y Acad Sci ; 1488(1): 3-15, 2021 03.
Article in English | MEDLINE | ID: mdl-33040338

ABSTRACT

The neuromuscular junction (NMJ) is a specialized structure that works as an interface to translate the action potential of the presynaptic motor neuron (MN) in the contraction of the postsynaptic myofiber. The design of appropriate experimental models is essential to have efficient and reliable approaches to study NMJ development and function, but also to generate conditions that recapitulate distinct features of diseases. Initial studies relied on the use of tissue slices maintained under the same environment and in which single motor axons were difficult to trace. Later, MNs and muscle cells were obtained from primary cultures or differentiation of progenitors and cocultured as monolayers; however, the tissue architecture was lost. Current approaches include self-assembling 3D structures or the incorporation of biomaterials with cells to generate engineered tissues, although the incorporation of Schwann cells remains a challenge. Thus, numerous investigations have established different NMJ models, some of which are quite complex and challenging. Our review summarizes the in vitro models that have emerged in recent years to coculture MNs and skeletal muscle, trying to mimic the healthy and diseased NMJ. We expect our review may serve as a reference for choosing the appropriate experimental model for the required purposes of investigation.


Subject(s)
Action Potentials/physiology , Motor Neurons/physiology , Neuromuscular Junction Diseases/physiopathology , Neuromuscular Junction/physiology , Neuromuscular Junction/physiopathology , Schwann Cells/physiology , Animals , Humans , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Neuromuscular Junction Diseases/diagnosis
13.
Neuron ; 51(5): 601-12, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16950158

ABSTRACT

An important step for cholinergic transmission involves the vesicular storage of acetylcholine (ACh), a process mediated by the vesicular acetylcholine transporter (VAChT). In order to understand the physiological roles of the VAChT, we developed a genetically altered strain of mice with reduced expression of this transporter. Heterozygous and homozygous VAChT knockdown mice have a 45% and 65% decrease in VAChT protein expression, respectively. VAChT deficiency alters synaptic vesicle filling and affects ACh release. Whereas VAChT homozygous mutant mice demonstrate major neuromuscular deficits, VAChT heterozygous mice appear normal in that respect and could be used for analysis of central cholinergic function. Behavioral analyses revealed that aversive learning and memory are not altered in mutant mice; however, performance in cognitive tasks involving object and social recognition is severely impaired. These observations suggest a critical role of VAChT in the regulation of ACh release and physiological functions in the peripheral and central nervous system.


Subject(s)
Brain/metabolism , Neuromuscular Junction Diseases/etiology , Neuromuscular Junction/metabolism , Recognition, Psychology/physiology , Vesicular Acetylcholine Transport Proteins/deficiency , Acetylcholine/analysis , Acetylcholine/metabolism , Animals , Blotting, Northern , Blotting, Southern , Brain/pathology , Brain/physiopathology , Brain Chemistry , Chromatography, High Pressure Liquid , Female , Male , Membrane Potentials/physiology , Mice , Mice, Transgenic , Microdialysis , Motor Activity/physiology , Neuromuscular Junction/pathology , Neuromuscular Junction/physiopathology , Neuromuscular Junction Diseases/pathology , Neuromuscular Junction Diseases/physiopathology , Polymerase Chain Reaction , RNA, Messenger/analysis , Synaptic Transmission/physiology , Vesicular Acetylcholine Transport Proteins/genetics
14.
Neurobiol Dis ; 38(1): 125-35, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20085811

ABSTRACT

Spinal muscular atrophy (SMA) is caused by insufficient levels of the survival motor neuron (SMN) protein leading to muscle paralysis and respiratory failure. In mouse, introducing the human SMN2 gene partially rescues Smn(-)(/)(-) embryonic lethality. However current models were either too severe or nearly unaffected precluding convenient drug testing for SMA. We report here new SMN2;Smn(-/-) lines carrying one to four copies of the human SMN2 gene. Mice carrying three SMN2 copies exhibited an intermediate phenotype with delayed appearance of motor defects and developmental breathing disorders reminiscent of those found in severe SMA patients. Although normal at birth, at 7 days of age respiratory rate was decreased and apnea frequency was increased in SMA mice in parallel with the appearance of neuromuscular junction defects in the diaphragm. With median survival of 15 days and postnatal onset of neurodegeneration, these mice could be an important tool for evaluating new therapeutics.


Subject(s)
Muscular Atrophy, Spinal/physiopathology , Neuromuscular Junction Diseases/physiopathology , Respiratory Paralysis/physiopathology , Animals , Diaphragm/innervation , Diaphragm/physiopathology , Disease Models, Animal , Disease Progression , Genes, Lethal/physiology , Genetic Predisposition to Disease/genetics , Humans , Mice , Mice, Transgenic , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/metabolism , Respiratory Insufficiency/genetics , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/physiopathology , Respiratory Paralysis/genetics , Respiratory Paralysis/metabolism , Survival of Motor Neuron 2 Protein/genetics
15.
Curr Opin Neurol ; 23(5): 496-501, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20664347

ABSTRACT

PURPOSE OF REVIEW: The objective of this review is to summarize recent advances in the treatment of various neuromuscular disorders including neuropathies, neuromuscular junction disorders, and myopathies. RECENT FINDINGS: Immunotherapy with sophisticated agents for myasthenia gravis and inflammatory myopathies, neuroprotection with vitamin E for chemotherapy-induced neuropathy, and promising gene transfer and exon-skipping therapies for muscular dystrophy are among the most exciting recent developments in the treatment of neuromuscular disorders. SUMMARY: In spite of significant advances, therapy in many neuromuscular diseases remains far from satisfactory. Better understanding of the underlying molecular and pathophysiologic processes for both hereditary and acquired disorders should lead to more refined and successful therapeutic approaches, reducing physical and other types of disability while posing fewer side effects.


Subject(s)
Muscular Diseases/therapy , Neuromuscular Diseases/therapy , Neuromuscular Junction Diseases/therapy , Humans , Muscular Diseases/genetics , Muscular Diseases/physiopathology , Neuromuscular Diseases/genetics , Neuromuscular Diseases/physiopathology , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/physiopathology
16.
Curr Opin Neurol ; 23(5): 489-95, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20651592

ABSTRACT

PURPOSE OF REVIEW: This review summarizes the recent advances on pathogenesis of antibody-mediated disorders of the neuromuscular junction, and results of studies on clinical assessment and treatments. RECENT FINDINGS: The incidence of myasthenia gravis, particularly in patients older than 50 years, is rising, and this is not solely due to improved disease recognition. It is uncertain how muscle specific tyrosine kinase (MuSK) antibody positive myasthenia gravis results in neuromuscular transmission failure since MuSK antibodies alter neuromuscular junction morphology without altering acetylcholine receptor numbers or turnover. Clinical tools have been developed that allow rapid and reliable disease assessment. The myasthenia gravis composite score addresses items commonly affected in myasthenia gravis, is sensitive to detect clinical change and helps guide the physician in therapy prescription. Immunosuppression remains the mainstay of myasthenia gravis treatment. Other therapies, such as rituximab, are increasingly prescribed for refractory myasthenia gravis, and drugs that inhibit complement are being explored in myasthenia gravis and Guillain-Barré syndrome (GBS). In Lambert-Eaton myasthenic syndrome (LEMS), SOX antibodies help distinguish between tumour and nontumour LEMS. Ganglioside complexes in GBS and Miller-Fisher syndrome are frequently present and are more pathogenic. SUMMARY: Developments in serological assays, particularly of cell-based assays, are continuing to improve the diagnosis and investigation of these conditions. Learning more on pathogenicity has helped us to apply newer therapies.


Subject(s)
Autoimmune Diseases , Neuromuscular Junction Diseases , Autoantibodies/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Autoimmune Diseases/therapy , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/physiopathology , Guillain-Barre Syndrome/therapy , Humans , Isaacs Syndrome/immunology , Isaacs Syndrome/physiopathology , Isaacs Syndrome/therapy , Lambert-Eaton Myasthenic Syndrome/immunology , Lambert-Eaton Myasthenic Syndrome/physiopathology , Lambert-Eaton Myasthenic Syndrome/therapy , Miller Fisher Syndrome/immunology , Miller Fisher Syndrome/physiopathology , Miller Fisher Syndrome/therapy , Myasthenia Gravis/pathology , Myasthenia Gravis/physiopathology , Myasthenia Gravis/therapy , Neuromuscular Junction Diseases/immunology , Neuromuscular Junction Diseases/physiopathology , Neuromuscular Junction Diseases/therapy , Thymectomy/adverse effects
17.
Am J Physiol Regul Integr Comp Physiol ; 298(3): R784-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20089713

ABSTRACT

Cardiopulmonary bypass (CPB) is associated with significant postoperative morbidity, but its effects on the neuromuscular system are unclear. Recent studies indicate that even relatively short periods of mechanical ventilation result in significant neuromuscular effects. Carbon monoxide (CO) has gained recent attention as therapy to reduce the deleterious effects of CPB. We hypothesized that 1) CPB results in impaired neuromuscular transmission and reduced diaphragm force generation; and 2) CO treatment during CPB will mitigate these effects. In adult male Sprague-Dawley rats, diaphragm muscle-specific force and neuromuscular transmission properties were measured 90 min after weaning from normothermic CPB (1 h). During CPB, either low-dose inhaled CO (250 ppm) or air was administered. The short period of mechanical ventilation used in the present study ( approximately 3 h) did not adversely affect diaphragm muscle contractile properties or neuromuscular transmission. CPB elicited a significant decrease in isometric diaphragm muscle-specific force compared with time-matched, mechanically ventilated rats ( approximately 25% decline in both twitch and tetanic force). Diaphragm muscle fatigability to 40-Hz repetitive stimulation did not change significantly. Neuromuscular transmission failure during repetitive activation was 60 +/- 2% in CPB animals compared with 76 +/- 4% in mechanically ventilated rats (P < 0.05). CO treatment during CPB abrogated the neuromuscular effects of CPB, such that diaphragm isometric twitch force and neuromuscular transmission were no longer significantly different from mechanically ventilated rats. Thus, CPB has important detrimental effects on diaphragm muscle contractility and neuromuscular transmission that are largely mitigated by CO treatment. Further studies are needed to ascertain the underlying mechanisms of CPB-induced neuromuscular dysfunction and to establish the potential role of CO therapy.


Subject(s)
Carbon Monoxide/pharmacology , Cardiopulmonary Bypass/adverse effects , Diaphragm , Muscle Fatigue/drug effects , Neuromuscular Junction Diseases , Administration, Inhalation , Animals , Antimetabolites/pharmacology , Body Temperature , Carbon Dioxide/blood , Diaphragm/drug effects , Diaphragm/innervation , Diaphragm/physiopathology , Dose-Response Relationship, Drug , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Fatigue/physiology , Neuromuscular Junction Diseases/drug therapy , Neuromuscular Junction Diseases/etiology , Neuromuscular Junction Diseases/physiopathology , Oxygen/blood , Rats , Rats, Sprague-Dawley , Respiration, Artificial , Ventilator Weaning
19.
Rinsho Shinkeigaku ; 60(2): 152-156, 2020 Feb 27.
Article in Japanese | MEDLINE | ID: mdl-31956197

ABSTRACT

A 27 year-old Canadian man suffered from fluctuating muscle weakness in the past several years. The patient had a past history of intestinal bleeding, bifid uvula and hypothyroidism in his childhood. Repetitive nerve stimulation tests showed a decrement pattern in the left deltoid muscle. The single fiber electromyography of the left extensor digitorum muscle showed an increment of jitter. Both findings were improved by the edrophonium test. He was diagnosed as having phosphoglucomutase 1 (PGM1) deficiency, as the compound heterozygote mutation of the PGM1 gene was recognized in the whole-exome sequencing and the enzyme activity of PGM1 was defective in the biopsied muscle. Treatment with the galactose lead to improvement of the fluctuating muscle weakness and decremental pattern in the repetitive stimulation test. PGM1 deficiency should be listed in the differential diagnosis of the neuromuscular junction disorder, when the patient is seronegative for antibodies related with myasthenia gravis and shows symptoms or signs consistent with PGM1 deficiency.


Subject(s)
Electrophysiology , Glycogen Storage Disease/complications , Glycogen Storage Disease/diagnosis , Muscle Weakness/etiology , Neuromuscular Junction Diseases/complications , Neuromuscular Junction Diseases/diagnosis , Adult , Humans , Male , Neuromuscular Junction Diseases/physiopathology
20.
Neurol Clin ; 38(3): 481-491, 2020 08.
Article in English | MEDLINE | ID: mdl-32703462

ABSTRACT

Skeletal muscle channelopathies are rare genetic neuromuscular conditions that include the nondystrophic myotonias and periodic paralyses. They cause disabling muscle symptoms and can limit educational potential, work opportunities, socialization, and quality of life. Effective therapy is available, making it essential to recognize and treat this group of disorders. Here, the authors highlight important aspects regarding diagnosis and management using illustrative case reports.


Subject(s)
Andersen Syndrome/diagnosis , Andersen Syndrome/genetics , Channelopathies/diagnosis , Channelopathies/genetics , Hypokalemic Periodic Paralysis/diagnosis , Hypokalemic Periodic Paralysis/genetics , Adolescent , Andersen Syndrome/physiopathology , Channelopathies/physiopathology , Humans , Hypokalemic Periodic Paralysis/physiopathology , Male , Muscle, Skeletal/physiopathology , Mutation/genetics , Myotonic Disorders/diagnosis , Myotonic Disorders/genetics , Myotonic Disorders/physiopathology , Neuromuscular Junction Diseases/diagnosis , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL