Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 246
Filtrar
1.
Channels (Austin) ; 18(1): 2349823, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38720415

RESUMEN

Myotonia congenita (MC) is a rare hereditary muscle disease caused by variants in the CLCN1 gene. Currently, the correlation of phenotype-genotype is still uncertain between dominant-type Thomsen (TMC) and recessive-type Becker (BMC). The clinical data and auxiliary examinations of MC patients in our clinic were retrospectively collected. Electromyography was performed in 11 patients and available family members. Whole exome sequencing was conducted in all patients. The clinical and laboratory data of Chinese MC patients reported from June 2004 to December 2022 were reviewed. A total of 11 MC patients were included in the study, with a mean onset age of 12.64 ± 2.73 years. The main symptom was muscle stiffness of limbs. Warm-up phenomenon and percussion myotonia were found in all patients. Electromyogram revealed significant myotonic charges in all patients and two asymptomatic carriers, while muscle MRI and biopsy showed normal or nonspecific changes. Fourteen genetic variants including 6 novel variants were found in CLCN1. Ninety-eight Chinese patients were re-analyzed and re-summarized in this study. There were no significant differences in the demographic data, clinical characteristics, and laboratory findings between 52 TMC and 46 BMC patients. Among the 145 variants in CLCN1, some variants, including the most common variant c.892 G>A, could cause TMC in some families and BMC in others. This study expanded the clinical and genetic spectrum of Chinese patients with MC. It was difficult to distinguish between TMC and BMC only based on the clinical, laboratory, and genetic characteristics.


Asunto(s)
Pueblo Asiatico , Canales de Cloruro , Miotonía Congénita , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Adulto Joven , Pueblo Asiatico/genética , China , Canales de Cloruro/genética , Pueblos del Este de Asia , Electromiografía , Mutación , Miotonía Congénita/genética , Miotonía Congénita/fisiopatología , Estudios Retrospectivos
2.
Int J Mol Sci ; 22(17)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34502093

RESUMEN

The troponin complex is a key regulator of muscle contraction. Multiple variants in skeletal troponin encoding genes result in congenital myopathies. TNNC2 has been implicated in a novel congenital myopathy, TNNI2 and TNNT3 in distal arthrogryposis (DA), and TNNT1 and TNNT3 in nemaline myopathy (NEM). Variants in skeletal troponin encoding genes compromise sarcomere function, e.g., by altering the Ca2+ sensitivity of force or by inducing atrophy. Several potential therapeutic strategies are available to counter the effects of variants, such as troponin activators, introduction of wild-type protein through AAV gene therapy, and myosin modulation to improve muscle contraction. The mechanisms underlying the pathophysiological effects of the variants in skeletal troponin encoding genes are incompletely understood. Furthermore, limited knowledge is available on the structure of skeletal troponin. This review focusses on the physiology of slow and fast skeletal troponin and the pathophysiology of reported variants in skeletal troponin encoding genes. A better understanding of the pathophysiological effects of these variants, together with enhanced knowledge regarding the structure of slow and fast skeletal troponin, will direct the development of treatment strategies.


Asunto(s)
Miotonía Congénita/metabolismo , Troponina/metabolismo , Animales , Humanos , Contracción Muscular , Miotonía Congénita/genética , Miotonía Congénita/fisiopatología , Sarcómeros/metabolismo , Troponina/química , Troponina/genética
4.
Elife ; 102021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33904400

RESUMEN

In addition to the hallmark muscle stiffness, patients with recessive myotonia congenita (Becker disease) experience debilitating bouts of transient weakness that remain poorly understood despite years of study. We performed intracellular recordings from muscle of both genetic and pharmacologic mouse models of Becker disease to identify the mechanism underlying transient weakness. Our recordings reveal transient depolarizations (plateau potentials) of the membrane potential to -25 to -35 mV in the genetic and pharmacologic models of Becker disease. Both Na+ and Ca2+ currents contribute to plateau potentials. Na+ persistent inward current (NaPIC) through NaV1.4 channels is the key trigger of plateau potentials and current through CaV1.1 Ca2+ channels contributes to the duration of the plateau. Inhibiting NaPIC with ranolazine prevents the development of plateau potentials and eliminates transient weakness in vivo. These data suggest that targeting NaPIC may be an effective treatment to prevent transient weakness in myotonia congenita.


Myotonia is a neuromuscular condition that causes problems with the relaxation of muscles following voluntary movements. One type of myotonia is Becker disease, also called recessive myotonia congenita. This is a genetic condition that causes muscle stiffness as a result of involuntary muscle activity. Patients may also suffer transient weakness for a few seconds or as long as several minutes after initiating a movement. The cause of these bouts of temporary weakness is still unclear, but there are hints that it could be linked to the muscle losing its excitability, the ability to respond to the stimuli that make it contract. However, this is at odds with findings that show that muscles in Becker disease are hyperexcitable. Muscle excitability depends on the presence of different concentrations of charged ions (positively charged sodium, calcium and potassium ions and negatively charged chloride ions) inside and outside of each muscle cells. These different concentrations of ions create an electric potential across the cell membrane, also called the 'membrane potential'. When a muscle cell gets stimulated, proteins on the cell membrane known as ion channels open. This allows the flow of ions between the inside and the outside of the cell, which causes an electrical current that triggers muscle contraction. To better understand the causes behind this muscle weakness, Myers et al. used mice that had either been genetically manipulated or given drugs to mimic Becker disease. By measuring both muscle force and the electrical currents that drive contraction, Myers et al. found that the mechanism underlying post-movement weakness involved a transient change in the concentrations of positively charged ions inside and outside the cells. Further experiments showed that proteins that regulate the passage of both sodium and calcium in and out of the cell ­ called sodium and calcium channels ­ contributed to this change in concentration. In addition, Myers et al. discovered that using a drug called ranolazine to stop sodium ions from entering the cell eliminated transient weakness in live mice. These findings suggest that in Becker disease, muscles cycle rapidly between being hyperexcited or not able to be excited, and that targeting the flow of sodium ions into the cell could be an effective treatment to prevent transient weakness in myotonia congenita. This study paves the way towards the development of new therapies to treat Becker disease as well as other muscle ion channel diseases with transient weakness such as periodic paralysis.


Asunto(s)
Potenciales de la Membrana/fisiología , Miotonía Congénita/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Miotonía Congénita/diagnóstico , Miotonía Congénita/genética , Sodio/fisiología
5.
Muscle Nerve ; 63(3): E21-E24, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33314145

Asunto(s)
Inhibidores de Puntos de Control Inmunológico/efectos adversos , Melanoma/tratamiento farmacológico , Enfermedades Musculares/inducido químicamente , Miositis/inducido químicamente , Miotonía Congénita/complicaciones , Distrofia Miotónica/complicaciones , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Neoplasias Cutáneas/tratamiento farmacológico , Anciano , Anticuerpos Monoclonales Humanizados/efectos adversos , Cardiomiopatía Dilatada , Canales de Cloruro/genética , Conectina/genética , Trastornos de Deglución/inducido químicamente , Trastornos de Deglución/complicaciones , Trastornos de Deglución/diagnóstico , Trastornos de Deglución/fisiopatología , Electrodiagnóstico , Electromiografía , Humanos , Ipilimumab/efectos adversos , Imagen por Resonancia Magnética , Masculino , Melanoma/secundario , Enfermedades Musculares/complicaciones , Enfermedades Musculares/genética , Enfermedades Musculares/fisiopatología , Miositis/complicaciones , Miositis/diagnóstico , Miositis/fisiopatología , Miotonía Congénita/diagnóstico , Miotonía Congénita/genética , Miotonía Congénita/fisiopatología , Distrofia Miotónica/diagnóstico , Distrofia Miotónica/fisiopatología , Conducción Nerviosa , Nivolumab/efectos adversos , Parestesia/inducido químicamente , Parestesia/complicaciones , Parestesia/fisiopatología , Enfermedades del Sistema Nervioso Periférico/complicaciones , Enfermedades del Sistema Nervioso Periférico/diagnóstico , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Proteínas de Unión al ARN/genética , Neoplasias Cutáneas/patología , Neoplasias de la Columna Vertebral/tratamiento farmacológico , Neoplasias de la Columna Vertebral/secundario
6.
Pflugers Arch ; 472(10): 1481-1494, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32748018

RESUMEN

In myotonia, reduced Cl- conductance of the mutated ClC-1 channels causes hindered muscle relaxation after forceful voluntary contraction due to muscle membrane hyperexcitability. Repetitive contraction temporarily decreases myotonia, a phenomena called "warm up." The underlying mechanism for the reduction of hyperexcitability in warm-up is currently unknown. Since potassium displacement is known to reduce excitability in, for example, muscle fatigue, we characterized the role of potassium in native myotonia congenita (MC) muscle. Muscle specimens of ADR mice (an animal model for low gCl- conductance myotonia) were exposed to increasing K+ concentrations. To characterize functional effects of potassium ion current, the muscle of ADR mice was exposed to agonists and antagonists of the big conductance Ca2+-activated K+ channel (BK) and the voltage-gated Kv7 channel. Effects were monitored by functional force and membrane potential measurements. By increasing [K+]0 to 5 mM, the warm-up phenomena started earlier and at [K+]0 7 mM only weak myotonia was detected. The increase of [K+]0 caused a sustained membrane depolarization accompanied with a reduction of myotonic bursts in ADR mice. Retigabine, a Kv7.2-Kv7.5 activator, dose-dependently reduced relaxation deficit of ADR myotonic muscle contraction and promoted the warm-up phenomena. In vitro results of this study suggest that increasing potassium conductivity via activation of voltage-gated potassium channels enhanced the warm-up phenomena, thereby offering a potential therapeutic treatment option for myotonia congenita.


Asunto(s)
Canales de Cloruro/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Miotonía Congénita/metabolismo , Potasio/metabolismo , Animales , Cloruros/metabolismo , Canales de Potasio KCNQ/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Masculino , Potenciales de la Membrana , Ratones , Contracción Muscular , Mutación , Miotonía Congénita/genética , Miotonía Congénita/fisiopatología , Bloqueadores de los Canales de Potasio/farmacología
7.
Muscle Nerve ; 62(4): 430-444, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32270509

RESUMEN

The nondystrophic myotonias are rare muscle hyperexcitability disorders caused by gain-of-function mutations in the SCN4A gene or loss-of-function mutations in the CLCN1 gene. Clinically, they are characterized by myotonia, defined as delayed muscle relaxation after voluntary contraction, which leads to symptoms of muscle stiffness, pain, fatigue, and weakness. Diagnosis is based on history and examination findings, the presence of electrical myotonia on electromyography, and genetic confirmation. In the absence of genetic confirmation, the diagnosis is supported by detailed electrophysiological testing, exclusion of other related disorders, and analysis of a variant of uncertain significance if present. Symptomatic treatment with a sodium channel blocker, such as mexiletine, is usually the first step in management, as well as educating patients about potential anesthetic complications.


Asunto(s)
Fatiga/fisiopatología , Debilidad Muscular/fisiopatología , Músculo Esquelético/fisiopatología , Mialgia/fisiopatología , Trastornos Miotónicos/fisiopatología , Acetazolamida/uso terapéutico , Edad de Inicio , Inhibidores de Anhidrasa Carbónica/uso terapéutico , Canales de Cloruro/genética , Electrodiagnóstico , Electromiografía , Pruebas Genéticas , Humanos , Lamotrigina/uso terapéutico , Mexiletine/uso terapéutico , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/genética , Miotonía Congénita/fisiopatología , Trastornos Miotónicos/genética , Canal de Sodio Activado por Voltaje NAV1.4/genética , Guías de Práctica Clínica como Asunto , Ranolazina/uso terapéutico , Bloqueadores de los Canales de Sodio/uso terapéutico , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico
8.
Int J Mol Sci ; 21(7)2020 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-32276507

RESUMEN

Myotonia congenita (MC) is a rare disorder characterized by stiffness and weakness of the limb and trunk muscles. Mutations in the SCN4A gene encoding the alpha-subunit of the voltage-gated sodium channel Nav1.4 have been reported to be responsible for sodium channel myotonia (SCM). The Nav1.4 channel is expressed in skeletal muscles, and its related channelopathies affect skeletal muscle excitability, which can manifest as SCM, paramyotonia and periodic paralysis. In this study, the missense mutation p.V445M was identified in two individual families with MC. To determine the functional consequences of having a mutated Nav1.4 channel, whole-cell patch-clamp recording of transfected Chinese hamster ovary cells was performed. Evaluation of the transient Na+ current found that a hyperpolarizing shift occurs at both the activation and inactivation curves with an increase of the window currents in the mutant channels. The Nav1.4 channel's co-expression with the Navß4 peptide can generate resurgent Na+ currents at repolarization following a depolarization. The magnitude of the resurgent currents is higher in the mutant than in the wild-type (WT) channel. Although the decay kinetics are comparable between the mutant and WT channels, the time to the peak of resurgent Na+ currents in the mutant channel is significantly protracted compared with that in the WT channel. These findings suggest that the p.V445M mutation in the Nav1.4 channel results in an increase of both sustained and resurgent Na+ currents, which may contribute to hyperexcitability with repetitive firing and is likely to facilitate recurrent myotonia in SCM patients.


Asunto(s)
Mutación Missense , Miotonía Congénita/genética , Miotonía Congénita/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.4/fisiología , Secuencia de Aminoácidos , Animales , Pueblo Asiatico , Células CHO , Canalopatías/genética , Canalopatías/metabolismo , Canalopatías/fisiopatología , Cricetulus , Femenino , Humanos , Masculino , Miotonía Congénita/metabolismo , Canal de Sodio Activado por Voltaje NAV1.4/química , Canal de Sodio Activado por Voltaje NAV1.4/genética , Canal de Sodio Activado por Voltaje NAV1.4/metabolismo , Técnicas de Placa-Clamp , Linaje
9.
Muscle Nerve ; 61(4): 491-495, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31944327

RESUMEN

INTRODUCTION: Brody myopathy (BM) is a recessive condition caused by mutations in the ATP2A1 gene and usually induces impaired muscle relaxation during and after exercise. Diagnosis relies on needle electromyography showing electrical silence, muscle biopsy with decreased sarcoplasmic reticulum calcium adenosine triphosphatase activity, and genetic analysis. Electrodiagnostic functional analyses are useful in the diagnosis of channelopathies, and thus may be impaired in BM. METHODS: We performed exercise tests and repetitive nerve stimulation (RNS; 10 supramaximal stimuli at 3 Hz) in 10 patients with BM. RESULTS: All participants showed incremental responses on RNS. Compound muscle action potential amplitude was increased and duration was decreased, especially in the ulnar nerve (+30.2 ± 7.1% and - 30.3 ± 2.8%, respectively; both P < .001). DISCUSSION: Easily accessible, this sign, referred to as the Arzel sign, could prove to be a very useful tool in BM diagnosis and in broadening its phenotype.


Asunto(s)
Potenciales de Acción/fisiología , Electrodiagnóstico/métodos , Músculo Esquelético/fisiopatología , Miotonía Congénita/diagnóstico , Nervio Cubital/fisiopatología , Estimulación Eléctrica , Electromiografía , Prueba de Esfuerzo , Femenino , Humanos , Masculino , Mutación , Miotonía Congénita/genética , Miotonía Congénita/fisiopatología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética
10.
Neurosci Lett ; 714: 134579, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31669315

RESUMEN

Nondystrophic myotonias are disorders of Na+ (Nav1.4 or SCN4A) and Cl- (CLCN1) channels in skeletal muscles, and frequently show phenotype heterogeneity. The molecular mechanism underlying their pathophysiology and phenotype heterogeneity remains unclear. As zebrafish models have been recently exploited for studies of the pathophysiology and phenotype heterogeneity of various human genetic diseases, a zebrafish model may be useful for delineating nondystrophic myotonias. Here, we generated transgenic zebrafish expressing a human mutant allele of SCN4A, referred to as Tg(mylpfa:N440K), and needle electromyography revealed increased number of myotonic discharges and positive sharp waves in the muscles of Tg(mylpfa:N440K) than in controls. In addition, forced exercise test at a water temperature of 24 °C showed a decrease in the distance moved, time spent in and number of visits to the zone with stronger swimming resistance. Finally, a forced exercise test at a water temperature of 18 °C exhibited a higher number of dive-bombing periods and drifting-down behavior than in controls. These findings indicate that Tg(mylpfa:N440K) is a good vertebrate model of exercise- and cold-induced human nondystrophic myotonias. This zebrafish model may contribute to provide insight into the pathophysiology of myotonia in sodium channelopathy and could be used to explore a new therapeutic avenue.


Asunto(s)
Frío , Modelos Animales de Enfermedad , Músculo Esquelético/fisiopatología , Miotonía Congénita/genética , Canal de Sodio Activado por Voltaje NAV1.4/genética , Esfuerzo Físico , Pez Cebra , Animales , Animales Modificados Genéticamente , Electromiografía , Mutación Missense , Miotonía/genética , Miotonía/fisiopatología , Miotonía Congénita/fisiopatología , Trastornos Miotónicos/genética , Trastornos Miotónicos/fisiopatología , Parálisis Periódica Hiperpotasémica/genética , Parálisis Periódica Hiperpotasémica/fisiopatología
11.
Neuromuscul Disord ; 29(11): 907-909, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31732390

RESUMEN

Variants of the skeletal muscle sodium channel gene SCN4A are associated with different neuromuscular disorders including sodium channel myotonia. Here, we report an infant with a de novo variant in SCN4A presenting with neonatal onset of severe muscle stiffness with involvement of facial and eyelid muscles, and life-threatening events with respiratory failure due to severe apnoea and thorax rigidity. The boy dramatically improved in both respiratory and motor function under carbamazepine therapy.


Asunto(s)
Variación Genética , Enfermedades Musculares/genética , Miotonía Congénita/genética , Canal de Sodio Activado por Voltaje NAV1.4/genética , Insuficiencia Respiratoria/genética , Elasticidad , Humanos , Lactante , Recién Nacido , Masculino , Enfermedades Musculares/fisiopatología , Enfermedades Musculares/terapia , Miotonía Congénita/fisiopatología , Miotonía Congénita/terapia , Fenotipo , Insuficiencia Respiratoria/fisiopatología , Insuficiencia Respiratoria/terapia
12.
J Electromyogr Kinesiol ; 49: 102362, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31610484

RESUMEN

INTRODUCTION: Myotonia congenita (MC) is caused by pathogenic variants in the CLCN1 gene coding the chloride channel protein. METHODS: To test the hypothesis that needle EMG could be helpful in distinguishing between the recessive and dominant MC, we performed EMG examination in 36 patients (23 men) aged 4-61 years with genetically proven MC: in 30 patients with autosomal recessive MC (Becker MC) and in 6 with autosomal dominant MC (Thomsen MC). RESULTS: Myotonic discharges were recorded in 95.8% of examined muscles. For the whole MC group we observed a significant positive correlation between parameters of motor unit activity potentials (MUAPs) in vastus lateralis and tibialis anterior muscles and the duration of the disease. Similar correlation for biceps brachii also was found in Becker MC subgroup only. DISCUSSION: EMG could still be helpful in diagnosis of MC and together with provocative tests might be useful in differentiation between recessive and autosomal MC.


Asunto(s)
Electromiografía/métodos , Potenciales Evocados Motores , Mutación , Miotonía Congénita/fisiopatología , Adolescente , Adulto , Niño , Preescolar , Diagnóstico Diferencial , Femenino , Genes Dominantes , Genes Recesivos , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/fisiopatología , Miotonía Congénita/diagnóstico , Miotonía Congénita/genética
13.
Skelet Muscle ; 9(1): 14, 2019 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-31133047

RESUMEN

BACKGROUND: Myopalladin (MYPN) is a component of the sarcomere that tethers nebulin in skeletal muscle and nebulette in cardiac muscle to alpha-actinin at the Z lines. Autosomal dominant MYPN mutations cause hypertrophic, dilated, or restrictive cardiomyopathy. Autosomal recessive MYPN mutations have been reported in only six families showing a mildly progressive nemaline or cap myopathy with cardiomyopathy in some patients. CASE PRESENTATION: A consanguineous family with congenital to adult-onset muscle weakness and hanging big toe was reported. Muscle biopsy showed minimal changes with internal nuclei, type 1 fiber predominance, and ultrastructural defects of Z line. Muscle CT imaging showed marked hypodensity of the sartorius bilaterally and MRI scattered abnormal high-intensity areas in the internal tongue muscle and in the posterior cervical muscles. Cardiac involvement was demonstrated by magnetic resonance imaging and late gadolinium enhancement. Whole exome sequencing analysis identified a homozygous loss of function single nucleotide deletion in the exon 11 of the MYPN gene in two siblings. Full-length MYPN protein was undetectable on immunoblotting, and on immunofluorescence, its localization at the Z line was missed. CONCLUSIONS: This report extends the phenotypic spectrum of recessive MYPN-related myopathies showing: (1) the two patients had hanging big toe and the oldest one developed spine and hand contractures, none of these signs observed in the previously reported patients, (2) specific ultrastructural changes consisting in Z line fragmentation, but (3) no nemaline or caps on muscle pathology.


Asunto(s)
Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Miotonía Congénita/genética , Adulto , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Consanguinidad , Exones , Femenino , Genes Recesivos , Homocigoto , Humanos , Mutación con Pérdida de Función , Masculino , Microscopía Electrónica de Transmisión , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , Miotonía Congénita/diagnóstico por imagen , Miotonía Congénita/fisiopatología , Linaje , Eliminación de Secuencia , Dedos del Pie/diagnóstico por imagen
14.
J Physiol ; 597(1): 225-235, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30284249

RESUMEN

KEY POINTS: During myotonia congenita, reduced chloride (Cl- ) conductance results in impaired muscle relaxation and increased muscle stiffness after forceful voluntary contraction. Repetitive contraction of myotonic muscle decreases or even abolishes myotonic muscle stiffness, a phenomenon called 'warm up'. Pharmacological inhibition of low Cl- channels by anthracene-9-carboxylic acid in muscle from mice and ADR ('arrested development of righting response') muscle from mice showed a relaxation deficit under physiological conditions compared to wild-type muscle. At increased osmolarity up to 400 mosmol L-1 , the relaxation deficit of myotonic muscle almost reached that of control muscle. These effects were mediated by the cation and anion cotransporter, NKCC1, and anti-myotonic effects of hypertonicity were at least partly antagonized by the application of bumetanide. ABSTRACT: Low chloride-conductance myotonia is caused by mutations in the skeletal muscle chloride (Cl- ) channel gene type 1 (CLCN1). Reduced Cl- conductance of the mutated channels results in impaired muscle relaxation and increased muscle stiffness after forceful voluntary contraction. Exercise decreases muscle stiffness, a phenomena called 'warm up'. To gain further insight into the patho-mechanism of impaired muscle stiffness and the warm-up phenomenon, we characterized the effects of increased osmolarity on myotonic function. Functional force and membrane potential measurements were performed on muscle specimens of ADR ('arrested development of righting response') mice (an animal model for low gCl- conductance myotonia) and pharmacologically-induced myotonia. Specimens were exposed to solutions of increasing osmolarity at the same time as force and membrane potentials were monitored. In the second set of experiments, ADR muscle and pharmacologically-induced myotonic muscle were exposed to an antagonist of NKCC1. Upon osmotic stress, ADR muscle was depolarized to a lesser extent than control wild-type muscle. High osmolarity diminished myotonia and facilitated the warm-up phenomenon as depicted by a faster muscle relaxation time (T90/10 ). Osmotic stress primarily resulted in the activation of the NKCC1. The inhibition of NKCC1 with bumetanide prevented the depolarization and reversed the anti-myotonic effect of high osmolarity. Increased osmolarity decreased signs of myotonia and facilitated the warm-up phenomenon in different in vitro models of myotonia. Activation of NKCC1 activity promotes warm-up and reduces the number of contractions required to achieve normal relaxation kinetics.


Asunto(s)
Miotonía Congénita/fisiopatología , Concentración Osmolar , Animales , Bumetanida/farmacología , Modelos Animales de Enfermedad , Femenino , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Miembro 2 de la Familia de Transportadores de Soluto 12/fisiología
15.
Hum Mol Genet ; 27(24): 4263-4272, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30215711

RESUMEN

Congenital myopathies are typically characterised by early onset hypotonia, weakness and hallmark features on biopsy. Despite the rapid pace of gene discovery, ∼50% of patients with a congenital myopathy remain without a genetic diagnosis following screening of known disease genes. We performed exome sequencing on two consanguineous probands diagnosed with a congenital myopathy and muscle biopsy showing selective atrophy/hypotrophy or absence of type II myofibres. We identified variants in the gene (MYL1) encoding the skeletal muscle fast-twitch specific myosin essential light chain (ELC) in both probands. A homozygous essential splice acceptor variant (c.479-2A > G, predicted to result in skipping of exon 5 was identified in Proband 1, and a homozygous missense substitution (c.488T>G, p.(Met163Arg)) was identified in Proband 2. Protein modelling of the p.(Met163Arg) substitution predicted it might impede intermolecular interactions that facilitate binding to the IQ domain of myosin heavy chain, thus likely impacting on the structure and functioning of the myosin motor. MYL1 was markedly reduced in skeletal muscle from both probands, suggesting that the missense substitution likely results in an unstable protein. Knock down of myl1 in zebrafish resulted in abnormal morphology, disrupted muscle structure and impaired touch-evoked escape responses, thus confirming that skeletal muscle fast-twitch specific myosin ELC is critical for myofibre development and function. Our data implicate MYL1 as a crucial protein for adequate skeletal muscle function and that MYL1 deficiency is associated with severe congenital myopathy.


Asunto(s)
Músculo Esquelético/fisiopatología , Cadenas Ligeras de Miosina/genética , Miotonía Congénita/genética , Alelos , Animales , Consanguinidad , Modelos Animales de Enfermedad , Exoma/genética , Homocigoto , Humanos , Masculino , Músculo Esquelético/metabolismo , Mutación , Cadenas Pesadas de Miosina/genética , Miotonía Congénita/fisiopatología , Linaje , Pez Cebra/genética
16.
Hum Mutat ; 39(9): 1273-1283, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29935101

RESUMEN

Myotonia congenita (MC) is a skeletal-muscle hyperexcitability disorder caused by loss-of-function mutations in the ClC-1 chloride channel. Mutations are scattered over the entire sequence of the channel protein, with more than 30 mutations located in the poorly characterized cytosolic C-terminal domain. In this study, we characterized, through patch clamp, seven ClC-1 mutations identified in patients affected by MC of various severities and located in the C-terminal region. The p.Val829Met, p.Thr832Ile, p.Val851Met, p.Gly859Val, and p.Leu861Pro mutations reside in the CBS2 domain, while p.Pro883Thr and p.Val947Glu are in the C-terminal peptide. We showed that the functional properties of mutant channels correlated with the clinical phenotypes of affected individuals. In addition, we defined clusters of ClC-1 mutations within CBS2 and C-terminal peptide subdomains that share the same functional defect: mutations between 829 and 835 residues and in residue 883 induced an alteration of voltage dependence, mutations between 851 and 859 residues, and in residue 947 induced a reduction of chloride currents, whereas mutations on 861 residue showed no obvious change in ClC-1 function. This study improves our understanding of the mechanisms underlying MC, sheds light on the role of the C-terminal region in ClC-1 function, and provides information to develop new antimyotonic drugs.


Asunto(s)
Canales de Cloruro/genética , Análisis Mutacional de ADN , Mutación/genética , Miotonía Congénita/genética , Adolescente , Adulto , Aminoácidos/genética , Femenino , Humanos , Activación del Canal Iónico/genética , Masculino , Persona de Mediana Edad , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/fisiopatología , Técnicas de Placa-Clamp , Péptidos/genética , Dominios Proteicos/genética
17.
Restor Neurol Neurosci ; 36(4): 459-467, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29889082

RESUMEN

BACKGROUND: The central nervous system involvement, in terms of a maladaptive sensory-motor plasticity, is well known in patients with dystrophic myotonias (DMs). To date, there are no data suggesting a central nervous system involvement in non-dystrophic myotonias (NDMs). OBJECTIVE: To investigate sensory-motor plasticity in patients with Myotonia Congenita (MC) and Paramyotonia Congenita (PMC) with or without mexiletine. METHODS: Twelve patients with a clinical, genetic, and electromyographic evidence of MC, fifteen with PMC, and 25 healthy controls (HC) were included in the study. TMS on both primary motor cortices (M1) and a rapid paired associative stimulation (rPAS) paradigm were carried out to assess M1 excitability and sensory-motor plasticity. RESULTS: patients showed a higher cortical excitability and a deterioration of the topographic specificity of rPAS aftereffects, as compared to HCs. There was no correlation among neurophysiological and clinical-demographic characteristics. Noteworthy, the patients who were under mexiletine showed a minor impairment of the topographic specificity of rPAS aftereffects as compared to those who did not take the drug. CONCLUSION: our findings could suggest the deterioration of cortical sensory-motor plasticity in patients with NDMs as a trait of the disease.


Asunto(s)
Antiarrítmicos/uso terapéutico , Mexiletine/uso terapéutico , Corteza Motora/fisiología , Miotonía Congénita/tratamiento farmacológico , Plasticidad Neuronal/fisiología , Adulto , Análisis de Varianza , Electromiografía , Potenciales Evocados Motores/efectos de los fármacos , Femenino , Estudios de Seguimiento , Humanos , Masculino , Miotonía Congénita/fisiopatología , Trastornos Miotónicos/fisiopatología , Trastornos Miotónicos/terapia , Estimulación Magnética Transcraneal , Adulto Joven
18.
Funct Neurol ; 33(4): 194-199, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30663965

RESUMEN

Some patients with multiple sclerosis (MS) complain of symptoms, such as myokymia, myotonia, spasms, and stiffness, which have been demonstrated to be due to a concurrent non-dystrophic myotonia, i.e. myotonia congenita or paramyotonia congenita. Beyond the known casual association between MS and non-dystrophic myotonia, a channelopathy representing a primary trait of MS rather than an epiphenomenon of demyelization (i.e., an acquired channelopathy) may exist. Indeed, the finding of MS patients with no genetic evidence of non-dystrophic myotonia but showing a clinical picture resembling this condition would support this hypothesis. Thirty patients with MS and no concurrent diagnosis of myotonia congenita or paramyotonia congenita were submitted to the Fournier protocol. Some of these MS patients presented abnormal muscle excitability with scarce myotonic discharges, but only a few of them had clinical features compatible with myotonia congenita or paramyotonia congenita syndromes. Even though the low number of recruited patients did not allow a robust statistical analysis, our data seemed to indicate the presence of an ion channel dysfunction that is independent of the acquired channelopathies and likely represents a common pathophysiological mechanism underlying a unique channelopathy simultaneously involving the peripheral and the central nervous system in individuals with MS. Confirming the presence of such a primary channelopathy in MS patients is of non-negligible importance, since dysfunction of ion channels may represent a suitable therapeutic target in MS.


Asunto(s)
Canalopatías/fisiopatología , Esclerosis Múltiple/fisiopatología , Trastornos Miotónicos/fisiopatología , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miotonía Congénita/fisiopatología
19.
Muscle Nerve ; 57(4): 586-594, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28877545

RESUMEN

INTRODUCTION: The gain-of-function mutations that underlie sodium channel myotonia (SCM) and paramyotonia congenital (PMC) produce differing clinical phenotypes. We used muscle velocity recovery cycles (MVRCs) to investigate membrane properties. METHODS: MVRCs and responses to trains of stimuli were compared in patients with SCM (n = 9), PMC (n = 8), and normal controls (n = 26). RESULTS: The muscle relative refractory period was reduced in SCM, consistent with faster recovery of the mutant sodium channels from inactivation. Both SCM and PMC showed an increased early supernormality and increased mean supernormality following multiple conditioning stimuli, consistent with slowed sodium channel inactivation. Trains of fast impulses caused a loss of amplitude in PMC, after which only half of the muscle fibers recovered, suggesting that the remainder stayed depolarized by persistent sodium currents. DISCUSSION: The differing effects of mutations on sodium channel function can be demonstrated in human subjects in vivo using this technique. Muscle Nerve 57: 586-594, 2018.


Asunto(s)
Potenciales de la Membrana , Fibras Musculares Esqueléticas/metabolismo , Miotonía Congénita/metabolismo , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miotonía Congénita/fisiopatología , Trastornos Miotónicos/metabolismo , Trastornos Miotónicos/fisiopatología , Periodo Refractario Electrofisiológico , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA