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

Country/Region as subject
Publication year range
1.
Muscle Nerve ; 64(1): 95-99, 2021 07.
Article in English | MEDLINE | ID: mdl-33835497

ABSTRACT

INTRODUCTION/AIMS: Paramyotonia congenita (PMC) is a skeletal muscle sodium channelopathy characterized by paradoxical myotonia, cold sensitivity, and exercise/cold-induced paralysis. Treatment with sodium-channel-blocking antiarrhythmic agents may expose patients to a risk of arrhythmia or may be poorly tolerated or ineffective. In this study we explored the effectiveness of non-antiarrhythmic sodium-channel blockers in two patients with PMC. METHODS: Earlier treatment with mexiletine was discontinued for gastrointestinal side effects in one of the patients and lack of clinical benefit in the other. One patient received lacosamide, ranolazine, and buprenorphine, and the other was given buprenorphine only. Drug efficacy was assessed by clinical scores, timed tests, and by long and short exercise tests. RESULTS: In both patients, buprenorphine improved pain scores by at least 50%, stiffness and weakness levels, and handgrip/eyelid-opening times. The fall in compound muscle action potential (CMAP) during short exercise normalized in both patients at baseline, and improved after cooling. During long exercise, one patient showed an earlier recovery of CMAP, and the other patient had a less severe decrease (<60%). With buprenorphine, the fall in CMAP induced by cooling normalized in one patient (from -72% to -4%) and improved (from -49% to -37%) in the other patient. DISCUSSION: Buprenorphine showed promising results for the treatment of exercise-induced paralysis and cold intolerance in the two patients assessed. The exercise test may be useful for quantitative assessment of treatment response. Further studies on a larger number of patients, under carefully controlled conditions, should be considered to address the effectiveness and long-term tolerability of this therapeutic option.


Subject(s)
Analgesics, Opioid/therapeutic use , Buprenorphine/therapeutic use , Myotonic Disorders/diagnosis , Myotonic Disorders/drug therapy , Analgesics, Opioid/pharmacology , Buprenorphine/pharmacology , Exercise Test/drug effects , Exercise Test/methods , Humans , Male , Middle Aged , Myotonic Disorders/genetics , NAV1.4 Voltage-Gated Sodium Channel/genetics , Treatment Outcome
2.
Muscle Nerve ; 59(2): 240-243, 2019 02.
Article in English | MEDLINE | ID: mdl-30390395

ABSTRACT

INTRODUCTION: Paramyotonia congenita (PMC) is a nondystrophic myotonic disorder that is believed to be caused by a defect in Nav 1.4 sodium channel inactivation. Ranolazine, which acts by enhancing slow inactivation of sodium channels, has been proposed as a therapeutic option, but in vivo studies are lacking. METHODS: We conducted an open-label, single-center trial of ranolazine to evaluate efficacy and tolerability in patients with PMC. Subjective symptoms of stiffness, weakness, and pain as well as clinical and electrical myotonia were evaluated. Baseline measures were compared with those after 4 weeks of treatment with ranolazine. RESULTS: Ranolazine was tolerated well without any serious adverse events. Both subjective symptoms and clinical myotonia were significantly improved. Duration of myotonia was reduced according to electromyography, but this change was not statistically significant in all tested muscles. DISCUSSION: Our findings support the use of ranolazine as a treatment for myotonia in PMC and suggest that a randomized, placebo-controlled trial is warranted. Muscle Nerve 59:240-243, 2019.


Subject(s)
Myotonic Disorders/drug therapy , Ranolazine/therapeutic use , Sodium Channel Blockers/therapeutic use , Adult , Electromyography , Female , Hand Strength/physiology , Humans , Male , Middle Aged , Muscle Weakness/etiology , Myotonic Disorders/complications , Pain/etiology , Severity of Illness Index , Stiff-Person Syndrome/etiology
3.
Pharmacol Res ; 141: 224-235, 2019 03.
Article in English | MEDLINE | ID: mdl-30611854

ABSTRACT

Sodium channel myotonia and paramyotonia congenita are caused by gain-of-function mutations in the skeletal muscle voltage-gated sodium channel hNav1.4. The first-line drug is the sodium channel blocker mexiletine; however, some patients show side effects or limited responses. We previously showed that two hNav1.4 mutations, p.G1306E and p.P1158L, reduce mexiletine potency in vitro, whereas another sodium channel blocker, flecainide, is less sensitive to mutation-induced gating defects. This observation was successfully translated to p.G1306E and p.P1158L carriers. Thus, the aim of this study was to perform a pharmacological characterization of myotonic Nav1.4 mutations clustered near the fast inactivation gate of the channel. We chose seven mutations (p.V1293I, p.N1297S, p.N1297K, p.F1298C, p.G1306E, p.I1310N, and p.T1313M) from the database of Italian and French networks for muscle channelopathies. Recombinant hNav1.4 mutants were expressed in HEK293T cells for functional and pharmacological characterization using the patch-clamp technique. All the studied mutations impair the kinetics and/or voltage dependence of fast inactivation, which is likely the main mechanism responsible for myotonia. The severity of myotonia is well-correlated to the enhancement of window currents generated by the intersection of the activation and fast inactivation voltage dependence. Five of the six mutants displaying a significant positive shift of fast inactivation voltage dependence reduced mexiletine inhibition in an experimental condition mimicking myotonia. In contrast, none of the mutations impairs flecainide block nor does p.T1313M impair propafenone block, indicating that class Ic antiarrhythmics may constitute a valuable alternative. Our study suggests that mutation-driven therapy would be beneficial to myotonic patients, greatly improving their quality of life.


Subject(s)
Myotonic Disorders/genetics , NAV1.4 Voltage-Gated Sodium Channel/genetics , Adolescent , Adult , Child , Child, Preschool , Female , HEK293 Cells , Humans , Infant, Newborn , Ion Channel Gating , Male , Middle Aged , Mutation , Myotonic Disorders/drug therapy , Young Adult
4.
Brain ; 140(9): 2295-2305, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-29050397

ABSTRACT

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.


Subject(s)
Myotonia Congenita/drug therapy , Myotonic Disorders/drug therapy , Triazines/therapeutic use , Adult , Aged , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Humans , Lamotrigine , Male , Middle Aged , Treatment Outcome , Triazines/adverse effects , Voltage-Gated Sodium Channel Blockers/adverse effects , Voltage-Gated Sodium Channel Blockers/therapeutic use , Young Adult
5.
JAMA ; 320(22): 2344-2353, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30535218

ABSTRACT

Importance: In rare diseases it is difficult to achieve high-quality evidence of treatment efficacy because of small cohorts and clinical heterogeneity. With emerging treatments for rare diseases, innovative trial designs are needed. Objective: To investigate the effectiveness of mexiletine in nondystrophic myotonia using an aggregated N-of-1 trials design and compare results between this innovative design and a previously conducted RCT. Design, Setting, and Participants: A series of aggregated, double-blind, randomized, placebo-controlled N-of-1-trials, performed in a single academic referral center. Thirty Dutch adult patients with genetically confirmed nondystrophic myotonia (38 patients screened) were enrolled between February 2014 and June 2015. Follow-up was completed in September 2016. Interventions: Mexiletine (600 mg daily) vs placebo during multiple treatment periods of 4 weeks. Main Outcomes and Measures: Reduction in daily-reported muscle stiffness on a scale of 1 to 9, with higher scores indicating more impairment. A Bayesian hierarchical model aggregated individual N-of-1 trial data to determine the posterior probability of reaching a clinically meaningful effect of a greater than 0.75-point difference. Results: Among 30 enrolled patients (mean age, 43.4 [SD, 15.24] years; 22% men; 19 CLCN1 and 11 SCN4A genotype), 27 completed the study and 3 dropped out (1 because of a serious adverse event). In 24 of the 27 completers, a clinically meaningful treatment effect was found. In the Bayesian hierarchical model, mexiletine resulted in a 100% posterior probability of reaching a clinically meaningful reduction in self-reported muscle stiffness for the nondystrophic myotonia group overall and the CLCN1 genotype subgroup and 93% posterior probability for the SCN4A genotype subgroup. In the total nondystrophic myotonia group, the median muscle stiffness score was 6.08 (interquartile range, 4.71-6.80) at baseline and was 2.50 (95% credible interval [CrI], 1.77-3.24) during the mexiletine period and 5.56 (95% CrI, 4.73-6.39) during the placebo period; difference in symptom score reduction, 3.06 (95% CrI, 1.96-4.15; n = 27) favoring mexiletine. The most common adverse event was gastrointestinal discomfort (21 mexiletine [70%], 1 placebo [3%]). One serious adverse event occurred (1 mexiletine [3%]; allergic skin reaction). Using frequentist reanalysis, mexiletine compared with placebo resulted in a mean reduction in daily-reported muscle stiffness of 3.12 (95% CI, 2.46-3.78), consistent with the previous RCT treatment effect of 2.69 (95% CI, 2.12-3.26). Conclusions and Relevance: In a series of N-of-1 trials of mexiletine vs placebo in patients with nondystrophic myotonia, there was a reduction in mean daily-reported muscle stiffness that was consistent with the treatment effect in a previous randomized clinical trial. These findings support the efficacy of mexiletine for treatment of nondystrophic myotonia as well as the feasibility of N-of-1 trials for assessing interventions in some chronic rare diseases. Trial Registration: ClinicalTrials.gov Identifier: NCT02045667.


Subject(s)
Mexiletine/therapeutic use , Myotonia/drug therapy , Myotonic Disorders/drug therapy , Voltage-Gated Sodium Channel Blockers/therapeutic use , Adult , Bayes Theorem , Double-Blind Method , Female , Humans , Male , Mexiletine/adverse effects , Models, Statistical , Randomized Controlled Trials as Topic , Rare Diseases , Voltage-Gated Sodium Channel Blockers/adverse effects
7.
Nucleic Acids Res ; 39(20): 8881-90, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21768123

ABSTRACT

Myotonic dystrophy type 2 (DM2) is an incurable neuromuscular disease caused by expanded CCUG repeats that may exhibit toxicity by sequestering the splicing regulator MBNL1. A series of triaminotriazine- and triaminopyrimidine-based small molecules (ligands 1-3) were designed, synthesized and tested as inhibitors of the MBNL1-CCUG interaction. Despite the structural similarities of the triaminotriazine and triaminopyrimidine units, the triaminopyrimidine-based ligands bind with low micromolar affinity to CCUG repeats (K(d) ∼ 0.1-3.6 µM) whereas the triaminotriazine ligands do not bind CCUG repeats. Importantly, these simple and small triaminopyrimidine ligands exhibit both strong inhibition (K(i) ∼ 2 µM) of the MBNL1-CCUG interaction and high selectivity for CCUG repeats over other RNA targets. These experiments suggest these compounds are potential lead agents for the treatment of DM2.


Subject(s)
Acridines/pharmacology , Pyrimidines/pharmacology , RNA-Binding Proteins/metabolism , RNA/drug effects , Acridines/chemistry , Base Sequence , DNA/chemistry , Humans , Intercalating Agents/chemical synthesis , Intercalating Agents/chemistry , Intercalating Agents/pharmacology , Ligands , Myotonic Disorders/drug therapy , Myotonic Dystrophy , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , RNA/chemistry , RNA/metabolism , RNA-Binding Proteins/antagonists & inhibitors , Repetitive Sequences, Nucleic Acid
8.
Neuromuscul Disord ; 31(11): 1124-1135, 2021 11.
Article in English | MEDLINE | ID: mdl-34702654

ABSTRACT

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.


Subject(s)
Mexiletine/therapeutic use , Myotonia/drug therapy , Adult , Aged , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Myotonia Congenita/drug therapy , Myotonic Disorders/drug therapy , Quality of Life , Treatment Outcome
9.
J Neuromuscul Dis ; 8(3): 357-381, 2021.
Article in English | MEDLINE | ID: mdl-33325393

ABSTRACT

BACKGROUND: Skeletal muscle ion channelopathies include non-dystrophic myotonias (NDM), periodic paralyses (PP), congenital myasthenic syndrome, and recently identified congenital myopathies. The treatment of these diseases is mainly symptomatic, aimed at reducing muscle excitability in NDM or modifying triggers of attacks in PP. OBJECTIVE: This systematic review collected the evidences regarding effects of pharmacological treatment on muscle ion channelopathies, focusing on the possible link between treatments and genetic background. METHODS: We searched databases for randomized clinical trials (RCT) and other human studies reporting pharmacological treatments. Preclinical studies were considered to gain further information regarding mutation-dependent drug effects. All steps were performed by two independent investigators, while two others critically reviewed the entire process. RESULTS: For NMD, RCT showed therapeutic benefits of mexiletine and lamotrigine, while other human studies suggest some efficacy of various sodium channel blockers and of the carbonic anhydrase inhibitor (CAI) acetazolamide. Preclinical studies suggest that mutations may alter sensitivity of the channel to sodium channel blockers in vitro, which has been translated to humans in some cases. For hyperkalemic and hypokalemic PP, RCT showed efficacy of the CAI dichlorphenamide in preventing paralysis. However, hypokalemic PP patients carrying sodium channel mutations may have fewer benefits from CAI compared to those carrying calcium channel mutations. Few data are available for treatment of congenital myopathies. CONCLUSIONS: These studies provided limited information about the response to treatments of individual mutations or groups of mutations. A major effort is needed to perform human studies for designing a mutation-driven precision medicine in muscle ion channelopathies.


Subject(s)
Channelopathies/drug therapy , Muscle, Skeletal/drug effects , Precision Medicine/methods , Humans , Hypokalemic Periodic Paralysis/drug therapy , Lamotrigine/therapeutic use , Mexiletine/therapeutic use , Mutation , Myasthenic Syndromes, Congenital/drug therapy , Myotonic Disorders/drug therapy , Randomized Controlled Trials as Topic , Sodium Channel Blockers/therapeutic use
10.
Pflugers Arch ; 460(2): 239-48, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20237798

ABSTRACT

Five hereditary sodium channelopathies of skeletal muscle have been identified. Prominent symptoms are either myotonia or weakness caused by an increase or decrease of muscle fiber excitability. The voltage-gated sodium channel NaV1.4, initiator of the muscle action potential, is mutated in all five disorders. Pathogenetically, both loss and gain of function mutations have been described, the latter being the more frequent mechanism and involving not just the ion-conducting pore, but aberrant pores as well. The type of channel malfunction is decisive for therapy which consists either of exerting a direct effect on the sodium channel, i.e., by blocking the pore, or of restoring skeletal muscle membrane potential to reduce the fraction of inactivated channels.


Subject(s)
Channelopathies/genetics , Myotonic Disorders/genetics , Sodium Channels/genetics , Action Potentials/physiology , Humans , Hypokalemic Periodic Paralysis/physiopathology , Membrane Potentials/physiology , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Myotonic Disorders/drug therapy , Myotonic Disorders/physiopathology , NAV1.4 Voltage-Gated Sodium Channel , Paralysis, Hyperkalemic Periodic/physiopathology , Potassium/adverse effects , Sodium Channels/chemistry , Sodium Channels/physiology
11.
Muscle Nerve ; 41(1): 133-7, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19768756

ABSTRACT

Pyridostigmine relieved episodic weakness in a family with paramyotonia congenita resulting from the R1448C mutation in the sodium channel gene. The transmission was autosomal dominant and the patients had paradoxical myotonia and exercise-induced weakness. On electrophysiological studies there were myotonic potentials, and there was progressive reduction of compound muscle action potential (CMAP) amplitudes after short exercise associated with clinical weakness. Pyridostigmine in doses of 60 mg three times daily abolished the drop in the postexercise CMAP amplitude and reduced the amplitude decrement to slow rate repetitive stimulation, but there continued to be a drop in amplitude on exposure to cold. The decline of the CMAP amplitude on exposure to cold was controlled by treatment with phenytoin. The clinical and electrophysiological features are discussed in relation to therapy with pyridostigmine and phenytoin.


Subject(s)
Muscle Weakness/drug therapy , Myotonic Disorders/complications , Pyridostigmine Bromide/therapeutic use , Adult , Cholinesterase Inhibitors/therapeutic use , Electromyography , Female , Follow-Up Studies , Humans , Muscle Weakness/etiology , Muscle Weakness/physiopathology , Myotonic Disorders/drug therapy , Myotonic Disorders/physiopathology , Pedigree
13.
Neuromuscul Disord ; 14(7): 405-16, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15210163

ABSTRACT

The antimyotonic activity of chiral derivatives of mexiletine and tocainide, selected as potent use-dependent blockers of skeletal muscle sodium channels, was evaluated in vivo acutely in myotonic ADR mice. The compounds had either aromatic (Me4 and Me6) or branched isopropyl groups (Me5 and To1) on the asymmetric centre, or had this latter one methylene apart from the amino group (Me2). Therapeutic doses of mexiletine (5-10 mg/kg) and tocainide (7-20 mg/kg) significantly reduced the long time of righting reflex (TRR), typical of ADR mice. Me4, Me5 and Me6 were 2-fold more potent than mexiletine. To1 fully normalised the TRR at 7 mg/kg. The electromyographic analysis confirmed a muscle-based activity for drug effectiveness on TRR. All the compounds reduced the myotonic hyperexcitability of intercostal muscle fibres when tested in vitro by current-clamp recordings, with a potency correlated with their action on sodium channels. On stimulus-evoked firing, the isopropyl analogues were 2-4-fold more potent than parent compounds, while the aromatic analogues were about 10-fold more potent than mexiletine. Patch-clamp recordings confirmed a normal-like pharmacological sensitivity of sodium channels of native ADR muscle fibres. Finally, the in vivo antimyotonic activity is due to the block of sodium channels and divergences with in vitro potency can be related to structure-based changes in drug pharmacokinetics.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Mexiletine/therapeutic use , Myotonic Disorders/drug therapy , Tocainide/therapeutic use , Action Potentials/drug effects , Analysis of Variance , Animals , Anti-Arrhythmia Agents/blood , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Conductivity , Electromyography/methods , Female , In Vitro Techniques , Inhibitory Concentration 50 , Male , Mexiletine/blood , Mice , Mice, Mutant Strains , Muscle Contraction/drug effects , Myotonic Disorders/blood , Patch-Clamp Techniques/methods , Sodium Channels/drug effects , Sodium Channels/physiology , Structure-Activity Relationship , Time Factors , Tocainide/analogs & derivatives , Tocainide/blood
14.
J Child Neurol ; 25(2): 212-5, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19168830

ABSTRACT

Paramyotonia congenita in 22 members of Arab (Omani) family is reported. Four generations were affected. All had early onset around 1 year of age, with myotonia and cold intolerance. Age of onset in the index case was at 3 months of age. Six members with the disease were examined and investigated. To our knowledge, this is the first report of the condition from this region. Childhood presentation and differential diagnosis is discussed.


Subject(s)
Arabs , Myotonic Disorders , Adolescent , Adult , Age of Onset , Cold Temperature , Diagnosis, Differential , Electromyography , Family , Female , Humans , Infant , Male , Myotonia/diagnosis , Myotonia/drug therapy , Myotonia/physiopathology , Myotonic Disorders/diagnosis , Myotonic Disorders/drug therapy , Myotonic Disorders/physiopathology , Oman , Pedigree , Young Adult
16.
Pharmacogenet Genomics ; 15(4): 235-44, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15864116

ABSTRACT

The effects of extracellular pH (6.2, 7.4 and 8.2) and 0.1 mM mexiletine, a channel blocker of the lidocaine type, are studied on two mutations of the fourth voltage sensor of the Nav1.4 sodium channel, R1448H/C. The fast inactivated channel state to which mexiletine preferentially binds is destabilized by the mutations. By contrast to the expected low response of R1448H/C carriers, mexiletine is particularly effective in preventing exercise-induced stiffness and paralysis from which these patients suffer. Our measurements performed in the whole-cell mode on stably transfected HEK cells show for the first time that the mutations strikingly accelerate closed-state inactivation and, as steady-state fast inactivation is shifted to more negative potentials, stabilize the fast inactivated channel state in the potential range around the resting potential. At pH 7.4 and 8.2, the phasic mexiletine block is larger for R1448C (55%) and R1448H (47%) than for wild-type channels (31%) due to slowed recovery from block (tau is approximately 520 ms for R1448C versus 270 ms for wild-type at pH 7.4) although the recovery from inactivation is slightly faster for the mutants (tau is approximately 1.9 ms for R1448C versus 3.8 ms for wild-type at pH 7.4). At pH 6.2, recovery from block is relatively fast (tau is approximately 35 ms for R1448H/C and 14 ms for wild-type) and thus shows no use-dependence. We conclude that enhanced closed-state inactivation expands the concept of a mutation-induced uncoupling of channel inactivation from activation to a new potential range and that the higher mexiletine efficacy in R1448H/C carriers compared to other myotonic patients offers a pharmacogenetic strategy for mutation-specific treatment.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Mexiletine/pharmacology , Sodium Channels/drug effects , Sodium Channels/genetics , Cell Line , Cysteine/genetics , Exercise , Histidine/genetics , Humans , Hydrogen-Ion Concentration , Mutation , Myotonic Disorders/drug therapy , Myotonic Disorders/metabolism , Paralysis , Patch-Clamp Techniques , Pharmacogenetics/methods , Time Factors , Transfection
17.
Neurology ; 60(3): 500-2, 2003 Feb 11.
Article in English | MEDLINE | ID: mdl-12578937

ABSTRACT

The efficacy and safety of creatine monohydrate (Cr) in patients with myotonic dystrophy type 2/proximal myotonic myopathy were studied in a small placebo-controlled double-blind trial. Twenty patients received either Cr or placebo for 3 months. After 3 months, there were no significant differences of muscle strength as assessed by hand-held dynamometry, testing of maximum grip strength, Medical Research Council scoring, and the Neuromuscular Symptom Score between the two groups. Some measures indicated trends toward mild improvement with Cr. Myalgia improved in two patients.


Subject(s)
Creatine/analogs & derivatives , Creatine/therapeutic use , Myotonic Disorders/drug therapy , Adult , Aged , Creatine/adverse effects , Dietary Supplements , Double-Blind Method , Female , Hand Strength , Humans , Male , Middle Aged , Muscle Weakness/diagnosis , Muscle Weakness/drug therapy , Muscle Weakness/etiology , Myotonic Disorders/complications , Myotonic Disorders/genetics , Pilot Projects , Treatment Outcome
18.
Neurol Sci ; 21(5 Suppl): S953-61, 2000.
Article in English | MEDLINE | ID: mdl-11382195

ABSTRACT

Myotonia and muscle weakness are cardinal features of myotonic disorders including the myotonic dystrophies and the non-dystrophic myotonias. Despite the recent progress in molecular genetics of these myotonic disorders, the precise mechanisms responsible for myotonia and for permanent or episodic muscle weakness are still unclear. Treatment has been mostly symptomatic, independent of the disease process involved. Moreover, there have been few randomized controlled trials of treatment for myotonic disorders and consequently no standardized treatment regimens are available. We present a review of selected treatment trials in the myotonic disorders and in muscle channelopathies, and discuss, on the basis of our experience in the myotonic disorders, the limits and advantages of treatment trials in this field. Future genotype-phenotype correlations using the patch-clamp technique are also illustrated.


Subject(s)
Ion Channels/metabolism , Muscle, Skeletal/metabolism , Myotonic Disorders/drug therapy , Calcium Channels/drug effects , Calcium Channels/genetics , Calcium Channels/metabolism , Chloride Channels/drug effects , Chloride Channels/genetics , Chloride Channels/metabolism , Humans , Ion Channels/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , Myotonic Disorders/metabolism , Myotonic Disorders/physiopathology , Myotonic Dystrophy/therapy , Paralysis, Hyperkalemic Periodic/metabolism , Paralysis, Hyperkalemic Periodic/physiopathology , Paralysis, Hyperkalemic Periodic/therapy , Sodium Channels/drug effects , Sodium Channels/genetics , Sodium Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL