Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Exp Neurol ; 361: 114303, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36563835

RESUMEN

It has long been accepted that myotonia (muscle stiffness) in patients with muscle channelopathies is due to myotonic discharges (involuntary firing of action potentials). In a previous study, we identified a novel phenomenon in myotonic muscle: development of plateau potentials, transient depolarizations to near -35 mV lasting for seconds to minutes. In the current study we examined whether plateau potentials contribute to myotonia. A recessive genetic model (ClCadr mice) with complete loss of muscle chloride channel (ClC-1) function was used to model severe myotonia congenita with complete loss of ClC-1 function and a pharmacologic model using anthracene-9-carboxylic acid (9 AC) was used to model milder myotonia congenita with incomplete loss of ClC-1 function. Simultaneous measurements of action potentials and myoplasmic Ca2+ from individual muscle fibers were compared to recordings of whole muscle force generation. In ClCadr muscle both myotonia and plateau potentials lasted 10s of seconds to minutes. During plateau potentials lasting 1-2 min, there was a gradual transition from high to low intracellular Ca2+, suggesting a transition in individual fibers from myotonia to flaccid paralysis in severe myotonia congenita. In 9 AC-treated muscles, both myotonia and plateau potentials lasted only a few seconds and Ca2+ remained elevated during the plateau potentials, suggesting plateau potentials contribute to myotonia without causing weakness. We propose, that in myotonic muscle, there is a novel state in which there is contraction in the absence of action potentials. This discovery provides a mechanism to explain reports of patients with myotonia who suffer from electrically silent muscle contraction lasting minutes.


Asunto(s)
Miotonía Congénita , Miotonía , Ratones , Animales , Miotonía/genética , Miotonía Congénita/genética , Miotonía Congénita/tratamiento farmacológico , Contracción Muscular , Potenciales de Acción/fisiología , Fibras Musculares Esqueléticas , Canales de Cloruro/genética , Modelos Animales de Enfermedad
2.
J Vet Intern Med ; 36(4): 1454-1459, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35815860

RESUMEN

CASE DESCRIPTION: A 10-month-old castrated male domestic longhair cat was evaluated for increasing frequency of episodic limb rigidity. CLINICAL FINDINGS: The cat presented for falling over and lying recumbent with its limbs in extension for several seconds when startled or excited. Upon examination, the cat had hypertrophied musculature, episodes of facial spasm, and a short-strided, stiff gait. DIAGNOSTICS: Electromyography (EMG) identified spontaneous discharges that waxed and waned in amplitude and frequency, consistent with myotonic discharges. A high impact 8-base pair (bp) deletion across the end of exon 3 and intron 3 of the chloride voltage-gated channel 1 (CLCN1) gene was identified using whole genome sequencing. TREATMENT AND OUTCOME: Phenytoin treatment was initiated at 3 mg/kg po q24 h and resulted in long-term improvement. CLINICAL RELEVANCE: This novel mutation within the CLCN1 gene is a cause of myotonia congenita in cats and we report for the first time its successful treatment.


Asunto(s)
Enfermedades de los Gatos , Miotonía Congénita , Animales , Enfermedades de los Gatos/diagnóstico , Enfermedades de los Gatos/tratamiento farmacológico , Enfermedades de los Gatos/genética , Gatos , Canales de Cloruro/genética , Electromiografía/veterinaria , Exones , Masculino , Mutación , Miotonía Congénita/diagnóstico , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/genética , Miotonía Congénita/veterinaria
3.
Elife ; 112022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35238775

RESUMEN

To date there are no therapies for patients with congenital myopathies, muscle disorders causing poor quality of life of affected individuals. In approximately 30% of the cases, patients with congenital myopathies carry either dominant or recessive mutations in the ryanodine receptor 1 (RYR1) gene; recessive RYR1 mutations are accompanied by reduction of RyR1 expression and content in skeletal muscles and are associated with fiber hypotrophy and muscle weakness. Importantly, muscles of patients with recessive RYR1 mutations exhibit increased content of class II histone deacetylases and of DNA genomic methylation. We recently created a mouse model knocked-in for the p.Q1970fsX16+ p.A4329D RyR1 mutations, which are isogenic to those carried by a severely affected child suffering from a recessive form of RyR1-related multi-mini core disease. The phenotype of the RyR1 mutant mice recapitulates many aspects of the clinical picture of patients carrying recessive RYR1 mutations. We treated the compound heterozygous mice with a combination of two drugs targeting DNA methylases and class II histone deacetylases. Here, we show that treatment of the mutant mice with drugs targeting epigenetic enzymes improves muscle strength, RyR1 protein content, and muscle ultrastructure. This study provides proof of concept for the pharmacological treatment of patients with congenital myopathies linked to recessive RYR1 mutations.


Asunto(s)
Enfermedades Musculares , Miotonía Congénita , Animales , ADN/metabolismo , Modelos Animales de Enfermedad , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Metiltransferasas/metabolismo , Ratones , Fuerza Muscular/genética , Músculo Esquelético/metabolismo , Mutación , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/genética , Calidad de Vida , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
4.
Neuromuscul Disord ; 31(11): 1124-1135, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34702654

RESUMEN

The MYOMEX study was a multicentre, randomised, double-blind, placebo-controlled, cross-over study aimed to compare the effects of mexiletine vs. placebo in patients with myotonia congenita (MC) and paramyotonia congenita (PC). The primary endpoint was the self-reported score of stiffness severity on a 100 mm visual analogic scale (VAS). Mexiletine treatment started at 200 mg/day and was up-titrated by 200 mg increment each three days to reach a maximum dose of 600 mg/day for total treatment duration of 18 days for each cross-over period. The modified intent-to-treat population included 25 patients (13 with MC and 12 with PC; mean age, 43.0 years; male, 68.0%). The median VAS score for mexiletine was 71.0 at baseline and decreased to 16.0 at the end of the treatment while the score did not change for placebo (81.0 at baseline vs. 78.0 at end of treatment). A mixed effects linear model analysis on ranked absolute changes showed a significant effect of treatment (p < 0.001). The overall score of the Individualized Neuromuscular Quality of Life questionnaire (INQoL) was significantly improved (p < 0.001). No clinically significant adverse events were reported. In conclusion, mexiletine improved stiffness and quality of life in patients with nondystrophic myotonia and was well tolerated.


Asunto(s)
Mexiletine/uso terapéutico , Miotonía/tratamiento farmacológico , Adulto , Anciano , Estudios Cruzados , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miotonía Congénita/tratamiento farmacológico , Trastornos Miotónicos/tratamiento farmacológico , Calidad de Vida , Resultado del Tratamiento
6.
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
7.
Exp Neurol ; 315: 52-59, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30738808

RESUMEN

Patients with myotonia congenita suffer from muscle stiffness caused by muscle hyperexcitability. Although loss-of-function mutations in the ClC-1 muscle chloride channel have been known for 25 years to cause myotonia congenita, this discovery has led to little progress on development of therapy. Currently, treatment is primarily focused on reducing hyperexcitability by blocking Na+ current. However, other approaches such as increasing K+ currents might also be effective. For example, the K+ channel activator retigabine, which opens KCNQ channels, is effective in treating epilepsy because it causes hyperpolarization of the resting membrane potential in neurons. In this study, we found that retigabine greatly reduced the duration of myotonia in vitro. Detailed study of its mechanism of action revealed that retigabine had no effect on any of the traditional measures of muscle excitability such as resting potential, input resistance or the properties of single action potentials. Instead it appears to shorten myotonia by activating K+ current during trains of action potentials. Retigabine also greatly reduced the severity of myotonia in vivo, which was measured using a muscle force transducer. Despite its efficacy in vivo, retigabine did not improve motor performance of mice with myotonia congenita. There are a number of potential explanations for the lack of motor improvement in vivo including central nervous system side effects. Nonetheless, the striking effectiveness of retigabine on muscle itself suggests that activating potassium currents is an effective method to treat disorders of muscle hyperexcitability.


Asunto(s)
Carbamatos/uso terapéutico , Moduladores del Transporte de Membrana/uso terapéutico , Miotonía Congénita/tratamiento farmacológico , Fenilendiaminas/uso terapéutico , Potenciales de Acción/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Técnicas In Vitro , Canales de Potasio KCNQ/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Ratones , Contracción Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Miotonía Congénita/psicología , Desempeño Psicomotor/efectos de los fármacos
8.
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
9.
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
10.
Brain ; 140(9): 2295-2305, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-29050397

RESUMEN

Mexiletine is the only drug with proven effect for treatment of non-dystrophic myotonia, but mexiletine is expensive, has limited availability and several side effects. There is therefore a need to identify other pharmacological compounds that can alleviate myotonia in non-dystrophic myotonias. Like mexiletine, lamotrigine is a sodium channel blocker, but unlike mexiletine, lamotrigine is available, inexpensive, and well tolerated. We investigated the potential of using lamotrigine for treatment of myotonia in patients with non-dystrophic myotonias. In this, randomized double-blind, placebo-controlled, two-period cross-over study, we included adult outpatients recruited from all of Denmark with clinical myotonia and genetically confirmed myotonia congenita and paramyotonia congenita for investigation at the Copenhagen Neuromuscular Center. A pharmacy produced the medication and placebo, and randomized patients in blocks of 10. Participants and investigators were all blinded to treatment until the end of the trial. In two 8-week periods, oral lamotrigine or placebo capsules were provided once daily, with increasing doses (from 25 mg, 50 mg, 150 mg to 300 mg) every second week. The primary outcome was a severity score of myotonia, the Myotonic Behaviour Scale ranging from asymptomatic (score 1) to invalidating myotonia (score 6), reported by the participants during Weeks 0 and 8 in each treatment period. Clinical myotonia was also measured and side effects were monitored. The study was registered at ClinicalTrials.gov (NCT02159963) and EudraCT (2013-003309-24). We included 26 patients (10 females, 16 males, age: 19-74 years) from 13 November 2013 to 6 July 2015. Twenty-two completed the entire study. One patient withdrew due to an allergic reaction to lamotrigine. Three patients withdrew for reasons not related to the trial intervention. The Myotonic Behaviour Scale at baseline was 3.2 ± 1.1, which changed after treatment with lamotrigine by 1.3 ± 0.2 scores (P < 0.001), but not with placebo (0.2 ± 0.1 scores, P = 0.4). The estimated effect size was 1.0 ± 0.2 (95% confidence interval = 0.5-1.5, P < 0.001, n = 22). The standardized effect size of lamotrigine was 1.5 (confidence interval: 1.2-1.8). Number needed to treat was 2.6 (P = 0.006, n = 26). No adverse or unsuspected event occurred. Common side effects occurred in both treatment groups; number needed to harm was 5.2 (P = 0.11, n = 26). Lamotrigine effectively reduced myotonia, emphasized by consistency between effects on patient-related outcomes and objective outcomes. The frequency of side effects was acceptable. Considering this and the high availability and low cost of the drug, we suggest that lamotrigine should be used as the first line of treatment for myotonia in treatment-naive patients with non-dystrophic myotonias.


Asunto(s)
Miotonía Congénita/tratamiento farmacológico , Trastornos Miotónicos/tratamiento farmacológico , Triazinas/uso terapéutico , Adulto , Anciano , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Lamotrigina , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Triazinas/efectos adversos , Bloqueadores del Canal de Sodio Activado por Voltaje/efectos adversos , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico , Adulto Joven
11.
Neurology ; 89(7): 710-713, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28710329

RESUMEN

OBJECTIVE: To determine open-label, pilot study whether ranolazine could improve signs and symptoms of myotonia and muscle stiffness in patients with myotonia congenita (MC). METHODS: Thirteen participants were assessed at baseline and 2, 4, and 5 weeks. Ranolazine was started after baseline assessment (500 mg twice daily), increased as tolerated after week 2 (1,000 mg twice daily), and maintained until week 4. Outcomes included change from baseline to week 4 in self-reported severity of symptoms (stiffness, weakness, and pain), Timed Up and Go (TUG), hand grip and eyelid myotonia, and myotonia on EMG. RESULTS: Self-reported severity of stiffness (p < 0.0001) and weakness (p < 0.01) was significantly improved compared with baseline. TUG and grip myotonia times were reduced (p = 0.03, p = 0.01). EMG of the abductor digiti minimi and tibialis anterior showed significantly reduced myotonia duration (p < 0.001, p < 0.01) at week 4. No participant discontinued ranolazine because of side effects. CONCLUSIONS: Ranolazine appeared to be well tolerated over a period of 4 weeks in individuals with MC, and ranolazine resulted in improvement of signs and symptoms of muscle stiffness. The findings of this study suggest that ranolazine should be investigated in a larger controlled study. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that ranolazine improves myotonia in myotonia congenita.


Asunto(s)
Fármacos Cardiovasculares/uso terapéutico , Miotonía Congénita/tratamiento farmacológico , Ranolazina/uso terapéutico , Adolescente , Adulto , Anciano , Electromiografía , Femenino , Estudios de Seguimiento , Fuerza de la Mano , Humanos , Masculino , Persona de Mediana Edad , Miotonía Congénita/fisiopatología , Proyectos Piloto , Autoinforme , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Adulto Joven
12.
Neurophysiol Clin ; 47(3): 247-252, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28153715

RESUMEN

OBJECTIVE: To investigate the cause of transient weakness in myotonia congenita (MC) and the mechanism of action of mexiletine in reducing weakness. METHODS: The changes in neuromuscular excitability produced by 1min of maximal voluntary contractions (MVC) were measured on the amplitude of compound muscle action potentials (CMAP) in two patients with either recessive or dominant MC, compared to control values obtained in 20 healthy subjects. Measurements were performed again in MC patients after mexiletine therapy. RESULTS: Transient reduction in maximal CMAP amplitude lasting several minutes after MVC was evident in MC patients, whereas no change was observed in controls. Mexiletine efficiently reduced this transient CMAP depression in both patients. DISCUSSION: Transient CMAP depression following sustained MVC may represent the electrophysiological correlate of the weakness clinically experienced by the patients. In MC, the low chloride conductance could induce self-sustaining action potentials after MVC, determining progressive membrane depolarization and a loss of excitability of muscle fibers, thus resulting in transient paresis. Mexiletine may prevent conduction block due to excessive membrane depolarization, thus reducing the transient CMAP depression following sustained MVC.


Asunto(s)
Mexiletine/uso terapéutico , Contracción Muscular , Debilidad Muscular/fisiopatología , Músculo Esquelético/fisiopatología , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/fisiopatología , Potenciales de Acción/efectos de los fármacos , Adulto , Electromiografía , Femenino , Humanos , Masculino , Debilidad Muscular/tratamiento farmacológico , Músculo Esquelético/efectos de los fármacos , Estimulación Eléctrica Transcutánea del Nervio , Resultado del Tratamiento
13.
Acta Myol ; 36(3): 125-134, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29774303

RESUMEN

INTRODUCTION: Myotonia permanens due to Nav1.4-G1306E is a rare sodium channelopathy with potentially life-threatening respiratory complications. Our goal was to study phenotypic variability throughout life. METHODS: Clinical neurophysiology and genetic analysis were performed. Using existing functional expression data we determined the sodium window by integration. RESULTS: In 10 unrelated patients who were believed to have epilepsy, respiratory disease or Schwartz-Jampel syndrome, we made the same prima facie diagnosis and detected the same heterologous Nav1.4-G1306E channel mutation as for our first myotonia permanens patient published in 1993. Eight mutations were de-novo, two were inherited from the affected parent each. Seven patients improved with age, one had a benign phenotype from birth, and two died of respiratory complications. The clinical features age-dependently varied with severe neonatal episodic laryngospasm in childhood and myotonia throughout life. Weakness of varying degrees was present. The responses to cold, exercise and warm-up were different for lower than for upper extremities. Spontaneous membrane depolarization increased frequency and decreased size of action potentials; self-generated repolarization did the opposite. The overlapping of steady-state activation and inactivation curves generated a 3.1-fold window area for G1306E vs. normal channels. DISCUSSION: Residue G1306 Neonatal laryngospasm and unusual distribution of myotonia, muscle hypertrophy, and weakness encourage direct search for the G1306E mutation, a hotspot for de-novo mutations. Successful therapy with the sodium channel blocker flecainide is due to stabilization of the inactivated state and special effectiveness for enlarged window currents. Our G1306E collection is the first genetically clarified case series from newborn period to adulthood and therefore helpful for counselling.


Asunto(s)
Miotonía Congénita/complicaciones , Miotonía Congénita/genética , Canal de Sodio Activado por Voltaje NAV1.4/genética , Potenciales de Acción , Adolescente , Adulto , Factores de Edad , Niño , Preescolar , Disnea/etiología , Ejercicio Físico/fisiología , Femenino , Flecainida/uso terapéutico , Heterocigoto , Humanos , Hipertrofia , Lactante , Recién Nacido , Laringismo/etiología , Masculino , Persona de Mediana Edad , Debilidad Muscular/etiología , Mutación , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.4/fisiología , Fenotipo , Ruidos Respiratorios/etiología , Enfermedades Respiratorias/etiología , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico , Adulto Joven
14.
Neurology ; 86(22): 2100-8, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27164696

RESUMEN

OBJECTIVE: We performed a clinical, functional, and pharmacologic characterization of the novel p.P1158L Nav1.4 mutation identified in a young girl presenting a severe myotonic phenotype. METHODS: Wild-type hNav1.4 channel and P1158L mutant were expressed in tsA201 cells for functional and pharmacologic studies using patch-clamp. RESULTS: The patient shows pronounced myotonia, slowness of movements, and generalized muscle hypertrophy. Because of general discomfort with mexiletine, she was given flecainide with satisfactory response. In vitro, mutant channels show a slower current decay and a rightward shift of the voltage dependence of fast inactivation. The voltage dependence of activation and slow inactivation were not altered. Mutant channels were less sensitive to mexiletine, whereas sensitivity to flecainide was not altered. The reduced inhibition of mutant channels by mexiletine was also observed using clinically relevant drug concentrations in a myotonic-like condition. CONCLUSIONS: Clinical phenotype and functional alterations of P1158L support the diagnosis of myotonia permanens. Impairment of fast inactivation is consistent with the possible role of the channel domain III S4-S5 loop in the inactivation gate docking site. The reduced sensitivity of P1158L to mexiletine may have contributed to the unsatisfactory response of the patient. The success of flecainide therapy underscores the usefulness of in vitro functional studies to help in the choice of the best drug for each individual.


Asunto(s)
Mutación , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/genética , Canal de Sodio Activado por Voltaje NAV1.4/genética , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico , Línea Celular , Niño , Diagnóstico Diferencial , Femenino , Flecainida/farmacología , Flecainida/uso terapéutico , Humanos , Mexiletine/efectos adversos , Mexiletine/farmacología , Mexiletine/uso terapéutico , Miotonía Congénita/diagnóstico , Miotonía Congénita/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.4/metabolismo , Pruebas de Farmacogenómica/métodos , Medicina de Precisión/métodos , Investigación Biomédica Traslacional , Bloqueadores del Canal de Sodio Activado por Voltaje/efectos adversos , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología
15.
Pediatrics ; 137(4)2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26944947

RESUMEN

Sodium channel myotonias are inherited muscle diseases linked to mutations in the voltage-gated sodium channel. These diseases may also affect newborns with variable symptoms. More recently, severe neonatal episodic laryngospasm (SNEL) has been described in a small number of patients. A timely diagnosis of SNEL is crucial because a specific treatment is now available that will likely reduced laryngospasm and improve vital and cerebral outcomes. We report here on an 8-year-old girl who had presented, at birth, with SNEL who subsequently developed myotonia permanens starting at age 3 years. Results of molecular analysis revealed a de novo SCN4A G1306E mutation. The girl was treated with carbamazepine, acetazolamide, and mexiletine, with little improvement; after switching her treatment to flecainide, she experienced a dramatic reduction in muscle stiffness and myotonic symptoms as well as an improvement in behavior.


Asunto(s)
Flecainida/uso terapéutico , Miotonía Congénita/tratamiento farmacológico , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico , Niño , Femenino , Humanos , Hipertrofia/etiología , Laringismo , Músculo Esquelético/patología , Mutación , Miotonía Congénita/complicaciones , Miotonía Congénita/genética , Canal de Sodio Activado por Voltaje NAV1.4/genética
16.
Clin Neurophysiol ; 126(2): 399-403, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25065301

RESUMEN

OBJECTIVE: We aim to demonstrate the effect of mexiletine on the compound muscle action potential (CMAP) amplitude transitory depression (TD) in a cohort of patients with recessive myotonia congenita. METHODS: We evaluated 21 patients with recessive myotonia congenita referred to our institute from 1990 to 2013 and treated with mexiletine chlorhydrate. All patients underwent prolonged 3 Hz repetitive nerve stimulation (3 Hz-PLRS) before and after the beginning of treatment. RESULTS: We observed in all subjects a reduction of CMAP amplitude TD after the beginning of treatment. The mean value of the TD nadir before starting mexiletine treatment was -62.0% and reduced to -28.8% after the therapy was started (51.6% reduction, p<0.001). CONCLUSIONS: The 3 Hz-PLRS is configured as a neurophysiological test able to indirectly detect and quantify, through the measurement of TD, the clinical phenomenon of the transitory weakness that occurs in myotonic syndromes due to CLCN1 mutations. SIGNIFICANCE: This neurophysiological test might be considered a helpful tool to assess the effect of anti-myotonic drugs, as mexiletine, in recessive myotonia congenita.


Asunto(s)
Potenciales Evocados Motores/efectos de los fármacos , Mexiletine/farmacología , Mexiletine/uso terapéutico , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/fisiopatología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Adolescente , Adulto , Anciano , Niño , Potenciales Evocados Motores/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miotonía Congénita/diagnóstico , Factores de Tiempo , Resultado del Tratamiento , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico , Adulto Joven
17.
Ann Neurol ; 77(2): 320-32, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25515836

RESUMEN

OBJECTIVE: Patients with myotonia congenita have muscle hyperexcitability due to loss-of-function mutations in the chloride channel in skeletal muscle, which causes spontaneous firing of muscle action potentials (myotonia), producing muscle stiffness. In patients, muscle stiffness lessens with exercise, a change known as the warmup phenomenon. Our goal was to identify the mechanism underlying warmup and to use this information to guide development of novel therapy. METHODS: To determine the mechanism underlying warmup, we used a recently discovered drug to eliminate muscle contraction, thus allowing prolonged intracellular recording from individual muscle fibers during induction of warmup in a mouse model of myotonia congenita. RESULTS: Changes in action potentials suggested slow inactivation of sodium channels as an important contributor to warmup. These data suggested that enhancing slow inactivation of sodium channels might offer effective therapy for myotonia. Lacosamide and ranolazine enhance slow inactivation of sodium channels and are approved by the US Food and Drug Administration for other uses in patients. We compared the efficacy of both drugs to mexiletine, a sodium channel blocker currently used to treat myotonia. In vitro studies suggested that both lacosamide and ranolazine were superior to mexiletine. However, in vivo studies in a mouse model of myotonia congenita suggested that side effects could limit the efficacy of lacosamide. Ranolazine produced fewer side effects and was as effective as mexiletine at a dose that produced none of mexiletine's hypoexcitability side effects. INTERPRETATION: We conclude that ranolazine has excellent therapeutic potential for treatment of patients with myotonia congenita.


Asunto(s)
Canales de Cloruro/antagonistas & inhibidores , Sistemas de Liberación de Medicamentos/métodos , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/fisiopatología , Bloqueadores de los Canales de Sodio/administración & dosificación , Acetanilidas/administración & dosificación , Animales , Canales de Cloruro/fisiología , Ratones , Ratones Transgénicos , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Miotonía Congénita/genética , Técnicas de Cultivo de Órganos , Piperazinas/administración & dosificación , Ranolazina
18.
BMJ Case Rep ; 20142014 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-24939454

RESUMEN

Mutations of the skeletal muscle voltage-gated sodium channel (NaV1.4) are an established cause of several clinically distinct forms of periodic paralysis and myotonia. Focal paresis has sometimes already been described. We report a case with atypical clinical manifestation comprising paramyotonia and cold-induced persistent and focal paralysis. A 27-year-old woman presented with paramyotonia congenita since her childhood. She experienced during her childhood one brief episode of generalised weakness. At the age of 27, she experienced a focal paresis lasting for several months. The known mutation p.Val1293Ile was found in the muscle sodium channel gene (SCN4A). Channel inactivation is involved in most Na(+) channelopathies. Fast inactivation is known to be responsible for the myotonia phenotype. We hypothesise that the V1293I mutation may also alter the slow inactivation in specific conditions, for example, prolonged cold exposure or prolonged and intensive exercise. This observation broadens the spectrum of clinical manifestations of this sodium channel mutation.


Asunto(s)
Miotonía Congénita/complicaciones , Parálisis/etiología , Femenino , Humanos , Mexiletine/uso terapéutico , Mutación/genética , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/genética , Canal de Sodio Activado por Voltaje NAV1.4/genética , Parálisis/tratamiento farmacológico , Resultado del Tratamiento , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico , Adulto Joven
19.
Neuromuscul Disord ; 24(8): 707-12, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24951453

RESUMEN

The spectrum of RYR1 mutation associated disease encompasses congenital myopathies, exercise induced rhabdomyolysis, malignant hyperthermia susceptibility and King-Denborough syndrome. We report the clinical phenotype of two siblings who presented in infancy with hypotonia and striking fatigable ptosis. Their response to pyridostigimine was striking, but genetic screening for congenital myasthenic syndromes was negative, prompting further evaluation. Muscle MRI was abnormal with a selective pattern of involvement evocative of RYR1-related myopathy. This directed sequencing of the RYR1 gene, which revealed two heterozygous c.6721C>T (p.Arg2241X) nonsense mutations and novel c.8888T>C (p.Leu2963Pro) mutations in both siblings. These cases broaden the RYR1-related disease spectrum to include a myasthenic-like phenotype, including partial response to pyridostigimine. RYR1-related myopathy should be considered in the presence of fatigable weakness especially if muscle imaging demonstrates structural abnormalities. Single fibre electromyography can also be helpful in cases like this.


Asunto(s)
Inhibidores de la Colinesterasa/uso terapéutico , Miotonía Congénita/tratamiento farmacológico , Miotonía Congénita/genética , Bromuro de Piridostigmina/uso terapéutico , Canal Liberador de Calcio Receptor de Rianodina/genética , Biopsia , Codón sin Sentido , Femenino , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Fatiga Muscular/efectos de los fármacos , Fatiga Muscular/fisiología , Músculos/efectos de los fármacos , Músculos/patología , Músculos/fisiopatología , Miotonía Congénita/patología , Miotonía Congénita/fisiopatología , Hermanos , Resultado del Tratamiento
20.
Exp Neurol ; 255: 96-102, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24613829

RESUMEN

Although the sodium channel blocker mexiletine is considered the first-line drug in myotonia, some patients experiment adverse effects, while others do not gain any benefit. Other antimyotonic drugs are thus needed to offer mexiletine alternatives. In the present study, we used a previously-validated rat model of myotonia congenita to compare six marketed sodium channel blockers to mexiletine. Myotonia was induced in the rat by injection of anthracen-9-carboxylic acid, a muscle chloride channel blocker. The drugs were given orally and myotonia was evaluated by measuring the time of righting reflex. The drugs were also tested on sodium currents recorded in a cell line transfected with the human skeletal muscle sodium channel hNav1.4 using patch-clamp technique. In vivo, carbamazepine and propafenone showed antimyotonic activity at doses similar to mexiletine (ED50 close to 5mg/kg); flecainide and orphenadrine showed greater potency (ED50 near 1mg/kg); lubeluzole and riluzole were the more potent (ED50 near 0.1mg/kg). The antimyotonic activity of drugs in vivo was linearly correlated with their potency in blocking hNav1.4 channels in vitro. Deviation was observed for propafenone and carbamazepine, likely due to pharmacokinetics and multiple targets. The comparison of the antimyotonic dose calculated in rats with the current clinical dose in humans strongly suggests that all the tested drugs may be used safely for the treatment of human myotonia. Considering the limits of mexiletine tolerability and the occurrence of non-responders, this study proposes an arsenal of alternative drugs, which may prove useful to increase the quality of life of individuals suffering from non-dystrophic myotonia. Further clinical trials are warranted to confirm these results.


Asunto(s)
Mexiletine/uso terapéutico , Músculo Esquelético/efectos de los fármacos , Miotonía Congénita/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/uso terapéutico , Animales , Carbamazepina/farmacología , Carbamazepina/uso terapéutico , Modelos Animales de Enfermedad , Flecainida/farmacología , Flecainida/uso terapéutico , Células HEK293 , Humanos , Mexiletine/farmacología , Orfenadrina/farmacología , Orfenadrina/uso terapéutico , Piperidinas/farmacología , Piperidinas/uso terapéutico , Propafenona/farmacología , Propafenona/uso terapéutico , Ratas , Ratas Wistar , Riluzol/farmacología , Riluzol/uso terapéutico , Bloqueadores de los Canales de Sodio/farmacología , Tiazoles/farmacología , Tiazoles/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA