Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
J Clin Invest ; 134(1)2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38165038

RESUMO

Myotonic dystrophy type 1 (DM1) involves misregulated alternative splicing for specific genes. We used exon or nucleotide deletion to mimic altered splicing of genes central to muscle excitation-contraction coupling in mice. Mice with forced skipping of exon 29 in the CaV1.1 calcium channel combined with loss of ClC-1 chloride channel function displayed markedly reduced lifespan, whereas other combinations of splicing mimics did not affect survival. The Ca2+/Cl- bi-channelopathy mice exhibited myotonia, weakness, and impairment of mobility and respiration. Chronic administration of the calcium channel blocker verapamil rescued survival and improved force generation, myotonia, and respiratory function. These results suggest that Ca2+/Cl- bi-channelopathy contributes to muscle impairment in DM1 and is potentially mitigated by common clinically available calcium channel blockers.


Assuntos
Canalopatias , Miotonia , Distrofia Miotônica , Camundongos , Animais , Distrofia Miotônica/tratamento farmacológico , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Cálcio/metabolismo , Cloretos/metabolismo , Miotonia/metabolismo , Verapamil/farmacologia , Verapamil/metabolismo , Canalopatias/genética , Canalopatias/metabolismo , Processamento Alternativo , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Músculo Esquelético/metabolismo
2.
J Physiol ; 600(12): 2835-2851, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35436004

RESUMO

Acquired and inherited dysfunction in voltage-gated sodium channels underlies a wide range of diseases. In addition to defects in trafficking and expression, sodium channelopathies are caused by dysfunction in one or several gating properties, for instance activation or inactivation. Disruption of channel inactivation leads to increased late sodium current, which is a common defect in seizure disorders, cardiac arrhythmias skeletal muscle myotonia and pain. An increase in late sodium current leads to repetitive action potentials in neurons and skeletal muscles, and prolonged action potential duration in the heart. In this Topical Review, we compare the effects of late sodium current in brain, heart, skeletal muscle and peripheral nerves.


Assuntos
Miotonia , Arritmias Cardíacas , Humanos , Miotonia/metabolismo , Dor , Sódio/metabolismo , Síndrome
3.
Sci Rep ; 8(1): 9714, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29946067

RESUMO

Dominantly inherited channelopathies of the skeletal muscle voltage-gated sodium channel NaV1.4 include hypokalaemic and hyperkalaemic periodic paralysis (hypoPP and hyperPP) and myotonia. HyperPP and myotonia are caused by NaV1.4 channel overactivity and overlap clinically. Instead, hypoPP is caused by gating pore currents through the voltage sensing domains (VSDs) of NaV1.4 and seldom co-exists clinically with myotonia. Recessive loss-of-function NaV1.4 mutations have been described in congenital myopathy and myasthenic syndromes. We report two families with the NaV1.4 mutation p.R1451L, located in VSD-IV. Heterozygous carriers in both families manifest with myotonia and/or hyperPP. In contrast, a homozygous case presents with both hypoPP and myotonia, but unlike carriers of recessive NaV1.4 mutations does not manifest symptoms of myopathy or myasthenia. Functional analysis revealed reduced current density and enhanced closed state inactivation of the mutant channel, but no evidence for gating pore currents. The rate of recovery from inactivation was hastened, explaining the myotonia in p.R1451L carriers and the absence of myasthenic presentations in the homozygous proband. Our data suggest that recessive loss-of-function NaV1.4 variants can present with hypoPP without congenital myopathy or myasthenia and that myotonia can present even in carriers of homozygous NaV1.4 loss-of-function mutations.


Assuntos
Paralisia Periódica Hipopotassêmica/genética , Paralisia Periódica Hipopotassêmica/metabolismo , Miotonia/genética , Miotonia/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.4/metabolismo , Adulto , Animais , Eletrofisiologia , Células HEK293 , Heterozigoto , Sequenciamento de Nucleotídeos em Larga Escala , Homozigoto , Humanos , Masculino , Mutação/genética , Canal de Sódio Disparado por Voltagem NAV1.4/genética , Linhagem , Estrutura Secundária de Proteína , Adulto Jovem
4.
Hum Mutat ; 37(1): 74-83, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26510092

RESUMO

Mutations in the gene coding for the skeletal muscle Cl(-) channel (CLCN1) lead to dominant or recessive myotonia. Here, we identified and characterized CLCN1 mutations in Costa Rican patients, who had been clinically diagnosed with myotonic dystrophy type 1 but who were negative for DM1 mutations. CLCN1 mutations c.501C>G, p.F167L and c.1235A>C, p.Q412P appeared to have recessive inheritance but patients had atypical clinical phenotypes; c.313C>T, p.R105C was found in combination with c.501C>G, p.F167L in an apparently recessive family and the c.461A>G, p.Q154R variant was associated with a less clear clinical picture. In Xenopus oocytes, none of the mutations exhibited alterations of fast or slow gating parameters or single channel conductance, and mutations p.R105C, p.Q154R, and p.F167L were indistinguishable from wild-type (WT). p.Q412P displayed a dramatically reduced current density, surface expression and exerted no dominant negative effect in the context of the homodimeric channel. Fluorescently tagged constructs revealed that p.Q412P is expressed inefficiently. Our study confirms p.F167L and p.R105C as myotonia mutations in the Costa Rican population, whereas p.Q154R may be a benign variant. p.Q412P most likely induces a severe folding defect, explaining the lack of dominance in patients and expression systems, but has WT properties once expressed in the plasma membrane.


Assuntos
Canais de Cloreto/genética , Estudos de Associação Genética , Mutação , Miotonia/diagnóstico , Miotonia/genética , Potenciais de Ação , Alelos , Animais , Canais de Cloreto/metabolismo , Feminino , Expressão Gênica , Humanos , Masculino , Miotonia/metabolismo , Oócitos/metabolismo , Linhagem , Fenótipo , Análise de Sequência de DNA
5.
EBioMedicine ; 2(9): 1034-47, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26501102

RESUMO

Loss of Muscleblind-like 1 (Mbnl1) is known to alter Clc-1 splicing to result in myotonia. Mbnl1(ΔE3/ΔE3)/Mbnl3(ΔE2) mice, depleted of Mbnl1 and Mbnl3, demonstrate a profound enhancement of myotonia and an increase in the number of muscle fibers with very low Clc-1 currents, where gClmax values approach ~ 1 mS/cm(2), with the absence of a further enhancement in Clc-1 splice errors, alterations in polyA site selection or Clc-1 localization. Significantly, Mbnl1(ΔE3/ΔE3)/Mbnl3(ΔE2) muscles demonstrate an aberrant accumulation of Clc-1 RNA on monosomes and on the first polysomes. Mbnl1 and Mbnl3 bind Clc-1 RNA and both proteins bind Hsp70 and eEF1A, with these associations being reduced in the presence of RNA. Thus binding of Mbnl1 and Mbnl3 to Clc-1 mRNA engaged with ribosomes can facilitate an increase in the local concentration of Hsp70 and eEF1A to assist Clc-1 translation. Dual depletion of Mbnl1 and Mbnl3 therefore initiates both Clc-1 splice errors and translation defects to synergistically enhance myotonia. As the HSA(LR) model for myotonic dystrophy (DM1) shows similar Clc-1 defects, this study demonstrates that both splice errors and translation defects are required for DM1 pathology to manifest. RESEARCH IN CONTEXT: Research in context: Myotonic Dystrophy type 1 (DM1) is a dominant disorder resulting from the expression of expanded CUG repeat RNA, which aberrantly sequesters and inactivates the muscleblind-like (MBNL) family of proteins. In mice, inactivation of Mbnl1 is known to alter Clc-1 splicing to result in myotonia. We demonstrate that concurrent depletion of Mbnl1 and Mbnl3 results in a synergistic enhancement of myotonia, with an increase in muscle fibers showing low chloride currents. The observed synergism results from the aberrant accumulation of Clc-1 mRNA on monosomes and the first polysomes. This translation error reflects the ability of Mbnl1 and Mbnl3 to act as adaptors that recruit Hsp70 and eEF1A to the Clc-1 mRNA engaged with ribosomes, to facilitate translation. Thus our study demonstrates that Clc-1 RNA translation defects work coordinately with Clc-1 splice errors to synergistically enhance myotonia in mice lacking Mbnl1 and Mbnl3.


Assuntos
Proteínas de Transporte/genética , Canais de Cloreto/genética , Proteínas de Ligação a DNA/genética , Miotonia/genética , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Processamento Alternativo , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Canais de Cloreto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , Immunoblotting , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Miotonia/metabolismo , Miotonia/fisiopatologia , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Distrofia Miotônica/fisiopatologia , Fator 1 de Elongação de Peptídeos/metabolismo , Ligação Proteica , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribossomos/genética , Ribossomos/metabolismo
6.
Sci Rep ; 5: 15382, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26502825

RESUMO

Mutations in the muscle chloride channel gene (CLCN1) cause myotonia congenita, an inherited condition characterized by muscle stiffness upon sudden forceful movement. We here studied the functional consequences of four disease-causing mutations that predict amino acid substitutions Q43R, S70L, Y137D and Q160H. Wild-type (WT) and mutant hClC-1 channels were heterologously expressed as YFP or CFP fusion protein in HEK293T cells and analyzed by whole-cell patch clamp and fluorescence recordings on individual cells. Q43R, Y137D and Q160H, but not S70L reduced macroscopic current amplitudes, but left channel gating and unitary current amplitudes unaffected. We developed a novel assay combining electrophysiological and fluorescence measurements at the single-cell level in order to measure the probability of ion channel surface membrane insertion. With the exception of S70L, all tested mutations significantly reduced the relative number of homodimeric hClC-1 channels in the surface membrane. The strongest effect was seen for Q43R that reduced the surface insertion probability by more than 99% in Q43R homodimeric channels and by 92 ± 3% in heterodimeric WT/Q43R channels compared to homodimeric WT channels. The new method offers a sensitive approach to investigate mutations that were reported to cause channelopathies, but display only minor changes in ion channel function.


Assuntos
Canais de Cloreto/metabolismo , Músculos/metabolismo , Mutação , Miotonia/metabolismo , Canais de Cloro CLC-2 , Canais de Cloreto/química , Dimerização , Células HEK293 , Humanos , Miotonia/genética , Propriedades de Superfície
7.
Rinsho Byori ; 62(3): 246-54, 2014 Mar.
Artigo em Japonês | MEDLINE | ID: mdl-24800500

RESUMO

Myotonic dystrophy (DM), the most common hereditary muscle disease in adults, is caused by the unstable genomic expansion of simple sequence repeats. This disease is characterized by myotonia and various multisystemic complications, most commonly those of the cardiac, endocrine, and central nervous systems. The cardiac abnormalities, especially cardiac conduction defects, significantly contribute to morbidity and mortality in DM patients. Therefore, understanding the pathophysiology of cardiac conduction defects in DM is important. The pathomechanism of DM has been thoroughly investigated. The mutant RNA transcripts containing the expanded repeat give rise to a toxic gain-of-function by perturbing splicing factors in the nucleus, leading to the misregulation of alternative pre-mRNA splicing. In particular, several studies, including ours, have shown that myotonia is caused by alternative splicing of the CLCN1 gene coding the voltage-gated chloride channel in skeletal muscle through an "RNA-dominant mechanism". Since the aberrantly spliced isoform does not seem to form a functional channel, the feature of skeletal muscle in DM can be interpreted as a "channelopathy" caused by reduced chloride channel protein. Similarly, we recently identified a misregulation of alternative splicing in an ion channel gene which is known to be responsible for arrhythmic disease showing Mendelian inheritance. Here, we review the cardiac manifestation and RNA-dominant mechanism of DM, and discuss the possible pathophysiology of cardiac conduction defects by referring to hereditary arrhythmic diseases, such as long QT syndrome and Brugada syndrome.


Assuntos
Arritmias Cardíacas/metabolismo , Sistema de Condução Cardíaco/anormalidades , Canais Iônicos/metabolismo , Miotonia/genética , Distrofia Miotônica/genética , Animais , Arritmias Cardíacas/genética , Síndrome de Brugada , Doença do Sistema de Condução Cardíaco , Canais de Cloreto/genética , Sistema de Condução Cardíaco/metabolismo , Humanos , Canais Iônicos/genética , Miotonia/metabolismo , Distrofia Miotônica/metabolismo , Splicing de RNA/fisiologia
8.
PLoS One ; 8(12): e81063, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324661

RESUMO

Mutations in the cytoplasmic tail (CT) of voltage gated sodium channels cause a spectrum of inherited diseases of cellular excitability, yet to date only one mutation in the CT of the human skeletal muscle voltage gated sodium channel (hNaV1.4F1705I) has been linked to cold aggravated myotonia. The functional effects of altered regulation of hNaV1.4F1705I are incompletely understood. The location of the hNaV1.4F1705I in the CT prompted us to examine the role of Ca(2+) and calmodulin (CaM) regulation in the manifestations of myotonia. To study Na channel related mechanisms of myotonia we exploited the differences in rat and human NaV1.4 channel regulation by Ca(2+) and CaM. hNaV1.4F1705I inactivation gating is Ca(2+)-sensitive compared to wild type hNaV1.4 which is Ca(2+) insensitive and the mutant channel exhibits a depolarizing shift of the V1/2 of inactivation with CaM over expression. In contrast the same mutation in the rNaV1.4 channel background (rNaV1.4F1698I) eliminates Ca(2+) sensitivity of gating without affecting the CaM over expression induced hyperpolarizing shift in steady-state inactivation. The differences in the Ca(2+) sensitivity of gating between wild type and mutant human and rat NaV1.4 channels are in part mediated by a divergence in the amino acid sequence in the EF hand like (EFL) region of the CT. Thus the composition of the EFL region contributes to the species differences in Ca(2+)/CaM regulation of the mutant channels that produce myotonia. The myotonia mutation F1705I slows INa decay in a Ca(2+)-sensitive fashion. The combination of the altered voltage dependence and kinetics of INa decay contribute to the myotonic phenotype and may involve the Ca(2+)-sensing apparatus in the CT of NaV1.4.


Assuntos
Cálcio/metabolismo , Calmodulina/metabolismo , Potenciais da Membrana/genética , Mutação , Miotonia/genética , Canal de Sódio Disparado por Voltagem NAV1.4/metabolismo , Sequência de Aminoácidos , Animais , Expressão Gênica , Células HEK293 , Humanos , Ativação do Canal Iônico , Cinética , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mutagênese Sítio-Dirigida , Miotonia/metabolismo , Miotonia/patologia , Canal de Sódio Disparado por Voltagem NAV1.4/genética , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Ratos
9.
Muscle Nerve ; 47(4): 483-7, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23381896

RESUMO

Myotonia is a heritable disorder in which patients are unable to willfully relax their muscles. The physiological basis for myotonia lies in well-established deficiencies of skeletal muscle chloride and sodium conductances. What is unclear is how normal muscle function can temporarily return with repeated movement, the so-called "warm-up" phenomenon. Electrophysiological analyses of the skeletal muscle voltage-gated sodium channel Nav 1.4 (gene name SCN4A), a key player in myotonia, have revealed several parallels between the Nav 1.4 biophysical signature, specifically slow-inactivation, and myotonic warm-up, which suggest that Nav 1.4 is critical not only in producing the myotonic reaction, but also in mediating the warm-up.


Assuntos
Fibras Musculares Esqueléticas/metabolismo , Miotonia/metabolismo , Transtornos Miotônicos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.4/metabolismo , Canais de Cloreto/metabolismo , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Miotonia/fisiopatologia , Transtornos Miotônicos/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.4/genética
10.
Biomark Med ; 7(1): 169-86, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23387498

RESUMO

Disease-specific biomarkers play a central diagnostic and therapeutic role in muscle pathology. Serum levels of a variety of muscle-derived enzymes are routinely used for the detection of muscle damage in diagnostic procedures, as well as for the monitoring of physical training status in sports medicine. Over the last few years, the systematic application of mass spectrometry-based proteomics for studying skeletal muscle degeneration has greatly expanded the range of muscle biomarkers, including new fiber-associated proteins involved in muscle transformation, muscular atrophy, muscular dystrophy, motor neuron disease, inclusion body myositis, myotonia, hypoxia, diabetes, obesity and sarcopenia of old age. These mass spectrometric studies have clearly established skeletal muscle proteomics as a reliable method for the identification of novel indicators of neuromuscular diseases.


Assuntos
Biomarcadores/sangue , Músculo Esquelético/metabolismo , Proteômica , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Humanos , Doença dos Neurônios Motores/metabolismo , Doença dos Neurônios Motores/patologia , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Miosite de Corpos de Inclusão/metabolismo , Miosite de Corpos de Inclusão/patologia , Miotonia/metabolismo , Miotonia/patologia , Obesidade/metabolismo , Obesidade/patologia , Proteoma/metabolismo
11.
Muscle Nerve ; 45(6): 897-900, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22581547

RESUMO

INTRODUCTION: Caveolin-3 is a major component of the caveolae in skeletal and cardiac muscle. Mutations in the caveolin-3 gene (CAV3) lead to a spectrum of clinical phenotypes including limb-girdle muscular dystrophy 1C, distal myopathy, rippling muscle disease, isolated hyperCKemia, and cardiomyopathy. CASE REPORT: A 24-year-old man with myalgia, muscle stiffness, and fatigue has normal strength and prominent myotonic discharges in the gastrocnemius. He also has epilepsy. He harbors a heterozygous CAV3 mutation, p.V57M. He has no mutations in CLCN1 and SCN4A, and he had normal genetic testing for myotonic dystrophy type 1 and type 2. CONCLUSIONS: Mutations in CAV3, and in particular p.V57M in CAV3, previously reported in isolated familial hyperCKemia, can be associated with electrical myotonia.


Assuntos
Caveolina 3/genética , Predisposição Genética para Doença/genética , Miotonia/genética , Adulto , Caveolina 3/metabolismo , Eletromiografia , Humanos , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Miotonia/metabolismo , Miotonia/fisiopatologia
12.
Am J Physiol Cell Physiol ; 302(7): C1019-25, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22237405

RESUMO

High interstitial K(+) concentration ([K(+)]) has been reported to impede normal propagation of electrical impulses along the muscle cell membrane (sarcolemma) and then also into the transverse tubule system; this is one considered underlying mechanism associated with the development of muscle fatigue. Interestingly, the extracellular buildup of lactic acid, once considered an additional cause for muscle fatigue, was recently shown to have force-restoring effects in such conditions. Specifically, it was proposed that elevated lactic acid (and intracellular acidosis) may lead to inhibition of voltage-gated chloride channels, thereby reestablishing better excitability of the muscle cell sarcolemma. In the present study, using an in vitro muscle contractile experimental setup to study functionally viable rectus abdominis muscle preparations obtained from normal swine, we examined the effects of 20 mM lactic acid and 512 µM 9-anthracenecarboxylic acid (9-AC; a voltage-gated chloride channel blocker) on the force recovery of K(+)-depressed (10 mM K(+)) twitch forces. We observed a similar muscle contractile restoration after both treatments. Interestingly, at elevated [K(+)], myotonia (i.e., hyperexcitability or afterdepolarizations), usually present in skeletal muscle with inherent or induced chloride channel dysfunctions, was not observed in the presence of either lactic acid or 9-AC. In part, these data confirm previous studies showing a force-restoring effect of lactic acid in high-[K(+)] conditions. In addition, we observed similar restorative effects of lactic acid and 9-AC, implicating a beneficial mechanism via voltage-gated chloride channel modulation.


Assuntos
Canais de Cloreto/metabolismo , Ácido Láctico/farmacologia , Fadiga Muscular/efeitos dos fármacos , Fadiga Muscular/fisiologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Animais , Antracenos/farmacologia , Canais de Cloreto/antagonistas & inibidores , Epinefrina/farmacologia , Humanos , Ácido Láctico/metabolismo , Masculino , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Esquelético/metabolismo , Miotonia/metabolismo , Miotonia/fisiopatologia , Ouabaína/farmacologia , Potássio/farmacologia , Suínos
13.
Mol Biosyst ; 7(8): 2480-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21629954

RESUMO

Myotonia is a symptom of various genetic and acquired skeletal muscular disorders and is characterized by hyperexcitability of the sarcolemma. Here, we have performed a comparative proteomic study of the genetic mouse models ADR, MTO and MTO*5J of human congenital myotonia in order to determine myotonia-specific changes in the global protein complement of gastrocnemius muscle. Proteomic analyses of myotonia in the mouse, which is caused by mutations in the gene encoding the muscular chloride channel Clc1, revealed a generally perturbed protein expression pattern in severely affected ADR and MTO muscle, but less pronounced alterations in mildly diseased MTO*5J mice. Alterations were found in major metabolic pathways, the contractile machinery, ion handling elements, the cellular stress response and cell signaling mechanisms, clearly confirming a glycolytic-to-oxidative transformation process in myotonic fast muscle. In the long-term, a detailed biomarker signature of myotonia will improve our understanding of the pathobiochemical processes underlying this disorder and be helpful in determining how a single mutation in a tissue-specific gene can trigger severe downstream effects on the expression levels of a very large number of genes in contractile tissues.


Assuntos
Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miotonia/metabolismo , Proteoma/análise , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Canais de Cloreto/análise , Canais de Cloreto/metabolismo , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Camundongos , Camundongos Transgênicos , Proteínas Musculares/análise , Músculo Esquelético/química , Proteoma/metabolismo , Transdução de Sinais
14.
Channels (Austin) ; 5(2): 161-72, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21317558

RESUMO

The antianginal drug ranolazine exerts voltage- and use-dependent block (UDB) of several Na+ channel isoforms, including Na(v) 1.4. We hypothesized that ranolazine will similarly inhibit the paramyotonia congenita Na(v) 1.4 gain-of-function mutations, R1448C, R1448H, and R1448P that are associated with repetitive action potential firing. Whole-cell Na+ current (I(Na)) was recorded from HEK293 cells expressing the hNa(v) 1.4 WT or R1448 mutations. At a holding potential (HP) of -140 mV, ranolazine exerted UDB (10 Hz) of WT and R1448 mutations (IC 50 = 59 - 71 µM). The potency for ranolazine UDB increased when the frequency of stimulation was raised to 30 Hz (IC 50 = 20 - 27 uM). When the HP was changed to -70 mV to mimic the resting potential of an injured skeletal muscle fibre, the potency of ranolazine to block I(Na) further increased; values of ranolazine IC 50 for block of WT, R1448C, R1448H, and R1448P were 3.8, 0.9, 6.3, and 0.9 uM, respectively. Ranolazine (30 uM) also caused a hyperpolarizing shift in the voltage-dependence of inactivation of WT and R1448 mutations. The effects of ranolazine (30 uM) to reduce I(Na) were similar (~35% I(Na) inhibition) when different conditioning pulse durations (2-20 msec) were used. Ranolazine (10 µM) suppressed the abnormal I(Na) induced by slow voltage ramps for R1448C channels. In computer simulations, 3 µM ranolazine inhibited the sustained and excessive firing of skeletal muscle action potentials that are characteristic of myotonia. Taken together, the data indicate that ranolazine interacts with the open state and stabilizes the inactivated state(s) of Na(v)1.4 channels, causes voltage- and use-dependent block of I(Na) and suppresses persistent I(Na). These data further suggest that ranolazine might be useful to reduce the sustained action potential firing seen in paramyotonia congenita.


Assuntos
Acetanilidas/farmacologia , Proteínas Musculares/antagonistas & inibidores , Mutação , Transtornos Miotônicos/genética , Piperazinas/farmacologia , Biofísica/métodos , Relação Dose-Resposta a Droga , Eletrofisiologia/métodos , Inibidores Enzimáticos/farmacologia , Humanos , Concentração Inibidora 50 , Proteínas Musculares/química , Músculo Esquelético/metabolismo , Mutagênese , Miotonia/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.4 , Isoformas de Proteínas , Ranolazina , Canais de Sódio/química , Software
15.
J Clin Invest ; 118(4): 1437-49, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18317596

RESUMO

Hyperkalemic periodic paralysis (HyperKPP) produces myotonia and attacks of muscle weakness triggered by rest after exercise or by K+ ingestion. We introduced a missense substitution corresponding to a human familial HyperKPP mutation (Met1592Val) into the mouse gene encoding the skeletal muscle voltage-gated Na+ channel NaV1.4. Mice heterozygous for this mutation exhibited prominent myotonia at rest and muscle fiber-type switching to a more oxidative phenotype compared with controls. Isolated mutant extensor digitorum longus muscles were abnormally sensitive to the Na+/K+ pump inhibitor ouabain and exhibited age-dependent changes, including delayed relaxation and altered generation of tetanic force. Moreover, rapid and sustained weakness of isolated mutant muscles was induced when the extracellular K+ concentration was increased from 4 mM to 10 mM, a level observed in the muscle interstitium of humans during exercise. Mutant muscle recovered from stimulation-induced fatigue more slowly than did control muscle, and the extent of recovery was decreased in the presence of high extracellular K+ levels. These findings demonstrate that expression of the Met1592ValNa+ channel in mouse muscle is sufficient to produce important features of HyperKPP, including myotonia, K+-sensitive paralysis, and susceptibility to delayed weakness during recovery from fatigue.


Assuntos
Músculo Esquelético/metabolismo , Miotonia/metabolismo , Miotonia/patologia , Potássio/metabolismo , Canais de Sódio/metabolismo , Envelhecimento/fisiologia , Animais , Progressão da Doença , Eletrofisiologia , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Miotonia/genética , Oxirredução , Paralisia Periódica Hiperpotassêmica/genética , Paralisia Periódica Hiperpotassêmica/metabolismo , Paralisia Periódica Hiperpotassêmica/patologia , Fenótipo , RNA Mensageiro/genética , Sensibilidade e Especificidade , Canais de Sódio/genética
16.
Curr Opin Neurol ; 20(5): 558-63, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17885445

RESUMO

PURPOSE OF REVIEW: To provide a current review of clinical phenotypes, genetics, molecular pathophysiology, and electro-diagnostic testing strategies of periodic paralysis and nondystrophic myotonias. RECENT FINDINGS: The number of pathogenic mutations causing periodic paralysis and nondystrophic myotonias continues to increase. Important insight into the molecular pathogenesis of muscle sodium channelopathies has been revealed by the finding of 'leaky' closed sodium channels. Previously, alterations in sodium-channel activation or inactivation have been identified as important disease mechanisms. The recent discovery that substitutions of key arginine residues in the voltage-sensing segment of the channel may lead to a 'pore leak' when the channel is closed suggests a new mechanism. Since similar mutations exist in corresponding positions of other channels, this mechanism may apply to other channel diseases. The recognition of different electrophysiological patterns that are specific to muscle ion-channel genotypes will be useful in diagnosis and in guiding genetic testing. Recent studies demonstrate that magnetic resonance imaging may be used to detect intramuscular accumulation of sodium during episodes of weakness. SUMMARY: Recent advances have refined our ability to make a precise molecular diagnosis in muscle channelopathies. The description of a pore leak with voltage-sensor mutations may represent a new disease mechanism.


Assuntos
Canalopatias/fisiopatologia , Músculo Esquelético/fisiopatologia , Miotonia/fisiopatologia , Paralisia Periódica Hiperpotassêmica/fisiopatologia , Canalopatias/genética , Canalopatias/metabolismo , Diagnóstico Diferencial , Humanos , Canais Iônicos/química , Canais Iônicos/genética , Canais Iônicos/metabolismo , Biologia Molecular/métodos , Músculo Esquelético/metabolismo , Miotonia/genética , Miotonia/metabolismo , Miotonia Congênita/genética , Miotonia Congênita/metabolismo , Miotonia Congênita/fisiopatologia , Paralisia Periódica Hiperpotassêmica/genética , Paralisia Periódica Hiperpotassêmica/metabolismo
17.
Muscle Nerve ; 31(4): 503-5, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15536612

RESUMO

Myotonic dystrophy (DM1) is an autosomal-dominant multisystem disease characterized by progressive skeletal muscle weakness, myotonia, cataracts, cardiac arrhythmias, mild mental retardation, and endocrinopathies. Heterozygous loss of SIX5 in mice causes cataracts and cardiac conduction disease, and homozygous loss also leads to sterility and decreased testicular mass, reminiscent of DM1 in humans. The effect of SIX5 deficiency in muscle is unknown. In this study, we found that muscle contractile properties, electromyographic insertional activity, and muscle histology were normal in SIX5 deficient mice. The implications of these findings for the pathogenesis of DM1 are discussed.


Assuntos
Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Contração Muscular/fisiologia , Miotonia/genética , Miotonia/metabolismo , Animais , Camundongos , Camundongos Knockout
18.
Intern Med ; 44(12): 1247-51, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16415544

RESUMO

OBJECTIVE: In order to find some appropriate medicine to suppress myotonia without decreasing muscle strength experiments were performed on myotonic (mto) mice whose Cl channel does not develop due to stop codon and serves as an animal model of myotonia. In myotonic dystrophy dehydroepiandrosterone is low in the serum and it has been reported that intravenous injections of DHEAS to human cases improves myotonia and activities of daily living. MATERIALS AND METHODS: Three pairs of heterozygote mto mice, SWR/J-Clcn1(adr-mto/+) and ten Wistar rats were used. We performed intracellular recordings of myotonia from mto mice and the drug effects on insertion myotonia were recorded from the hemidiaphragm preparations of mto mice with different concentrations of DHEAS. Isometric twitch tension was recorded from rat hemidiaphragm preparations in Tyrode's solution and the effect of DHEAS on the muscle twitch tension was measured at different concentrations of DHEAS from 100 mg/l to 300 mg/l. The effect of mexiletine on ITT was also measured. RESULTS: In mto mice insertion myotonia was recorded as soon as the microelectrode was inserted in the muscle cells. When DHEAS was added to Tyrode's solution, insertion myotonia was suppressed. DHEAS decreased ITT up to 70% of the original value, though mexiletine decreased ITT to 30% of the original value. Therefore, the decrement of the muscle strength in DHEAS solution is much smaller than that of mexiletine. CONCLUSION: Since myotonic dystrophy shows progressive muscle weakness in addition to myotonia, medications like DHEAS are more favorable than the typical Na channel blocker.


Assuntos
Sulfato de Desidroepiandrosterona/uso terapêutico , Contração Isométrica/efeitos dos fármacos , Músculo Esquelético/fisiopatologia , Miotonia/tratamento farmacológico , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Modelos Animais de Doenças , Eletrofisiologia/métodos , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Miotonia/metabolismo , Miotonia/patologia , Ratos , Ratos Wistar
19.
J Physiol ; 554(Pt 2): 321-34, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14608015

RESUMO

Flecainide, a class IC antiarrhythmic, was shown to improve myotonia caused by sodium channel mutations in situations where the class IB antiarrhythmic drug mexiletine was less efficient. Yet little is known about molecular interactions between flecainide and human skeletal muscle sodium (hNa(v)1.4) channels. Whole-cell sodium currents (I(Na)) were recorded in tsA201 cells expressing wild-type (WT) and mutant hNa(v)1.4 channels (R1448C, paramyotonia congenita; G1306E, potassium-aggravated myotonia). At a holding potential (HP) of -120 mV, flecainide use-dependently blocked WT and G1306E I(Na) equally but was more potent on R1448C channels. For WT, the extent of block depended on a holding voltage more negative than the activation threshold, being greater at -90 mV as compared to -120 and -180 mV. This behaviour was exacerbated by the R1448C mutation since block at -120 mV was greater than that at -180 mV. Thus flecainide can bind to inactivated sodium channels in the absence of channel opening. Nevertheless, all the channels showed the same closed-state affinity constant (K(R) approximately 480 microM) and the same inactivated-state affinity constant (K(I) approximately 18 microM). Simulations according to the modulated receptor hypothesis mimic the voltage-dependent block of WT and mutant channels by flecainide and mexiletine. All the results suggest similar blocking mechanisms for the two drugs. Yet, since flecainide exerts use-dependent block at lower frequency than mexiletine, it may exhibit greater benefit in all myotonic syndromes. Moreover, flecainide blocks hNa(v)1.4 channel mutants with a rightward shift of availability voltage dependence more specifically than mexiletine, owing to a lower K(R)/K(I) ratio. This study offers a pharmacogenetic strategy to better address treatment in individual myotonic patients.


Assuntos
Flecainida/farmacologia , Proteínas Musculares/antagonistas & inibidores , Mutação , Miotonia/tratamento farmacológico , Bloqueadores dos Canais de Sódio/farmacologia , Relação Dose-Resposta a Droga , Flecainida/metabolismo , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Proteínas Musculares/metabolismo , Miotonia/genética , Miotonia/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.4 , Bloqueadores dos Canais de Sódio/metabolismo , Canais de Sódio/metabolismo
20.
J Neurol ; 249(11): 1493-502, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12420087

RESUMO

Ion channelopathies have common clinical features, recurrent patterns of mutations, and almost predictable mechanisms of pathogenesis. In skeletal muscle, disorders are associated with mutations in voltage-gated Na(+), K(+), Ca(2+), and Cl(-) channels leading to hypoexcitability, causing periodic paralysis and to hyperexcitabilty, resulting in myotonia or susceptibility to malignant hyperthermia.


Assuntos
Canais Iônicos/metabolismo , Músculo Esquelético/metabolismo , Doenças Musculares/metabolismo , Sarcolema/metabolismo , Humanos , Canais Iônicos/genética , Hipertermia Maligna/genética , Hipertermia Maligna/metabolismo , Hipertermia Maligna/fisiopatologia , Músculo Esquelético/inervação , Músculo Esquelético/fisiopatologia , Doenças Musculares/genética , Doenças Musculares/fisiopatologia , Mutação/genética , Miotonia/genética , Miotonia/metabolismo , Miotonia/fisiopatologia , Paralisia Periódica Hiperpotassêmica/genética , Paralisia Periódica Hiperpotassêmica/metabolismo , Paralisia Periódica Hiperpotassêmica/fisiopatologia , Sarcolema/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...