Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Front Physiol ; 11: 533946, 2020.
Article in English | MEDLINE | ID: mdl-33329012

ABSTRACT

Reduced Cl- conductance causes inhibited muscle relaxation after forceful voluntary contraction due to muscle membrane hyperexcitability. This represents the pathomechanism of myotonia congenita. Due to the prevailing data suggesting that an increased potassium level is a main contributor, we studied the effect of a modulator of a big conductance Ca2+- and voltage-activated K+ channels (BK) modulator on contraction and relaxation of slow- and high-twitch muscle specimen before and after the pharmacological induction of myotonia. Human and murine muscle specimens (wild-type and BK-/-) were exposed to anthracene-9-carboxylic acid (9-AC) to inhibit CLC-1 chloride channels and to induce myotonia in-vitro. Functional effects of BK-channel activation and blockade were investigated by exposing slow-twitch (soleus) and fast-twitch (extensor digitorum longus) murine muscle specimens or human musculus vastus lateralis to an activator (NS1608) and a blocker (Paxilline), respectively. Muscle-twitch force and relaxation times (T90/10) were monitored. Compared to wild type, fast-twitch muscle specimen of BK-/- mice resulted in a significantly decreased T90/10 in presence of 9-AC. Paxilline significantly shortened T90/10 of murine slow- and fast-twitch muscles as well as human vastus lateralis muscle. Moreover, twitch force was significantly reduced after application of Paxilline in myotonic muscle. NS1608 had opposite effects to Paxilline and aggravated the onset of myotonic activity by prolongation of T90/10. The currently used standard therapy for myotonia is, in some individuals, not very effective. This in vitro study demonstrated that a BK channel blocker lowers myotonic stiffness and thus highlights its potential therapeutic option in myotonia congenital (MC).

2.
Pflugers Arch ; 472(10): 1533, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32885305

ABSTRACT

The original article contains an error during online publication. Table 2 was included during production round and now deleted. The Original article has been corrected.

3.
Pflugers Arch ; 472(10): 1481-1494, 2020 10.
Article in English | MEDLINE | ID: mdl-32748018

ABSTRACT

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


Subject(s)
Chloride Channels/genetics , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Myotonia Congenita/metabolism , Potassium/metabolism , Animals , Chlorides/metabolism , KCNQ Potassium Channels/metabolism , Large-Conductance Calcium-Activated Potassium Channels/agonists , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Male , Membrane Potentials , Mice , Muscle Contraction , Mutation , Myotonia Congenita/genetics , Myotonia Congenita/physiopathology , Potassium Channel Blockers/pharmacology
4.
Neuropediatrics ; 50(5): 308-312, 2019 10.
Article in English | MEDLINE | ID: mdl-31226716

ABSTRACT

Mutations that disrupt the TBC1D24 presynaptic protein have been implicated in various neurological disorders including epilepsy, chronic encephalopathy, DOORS (deafness, onychodystrophy, osteodystrophy, mental retardation, and seizures) syndrome, nonsyndromic hearing loss, and myoclonus. We present the case of a 22-month-old male with infantile-onset paroxysmal episodes of facial and limb myoclonus. The episodes were linked to biallelic variants in exon 2 of the TBC1D24 gene that lead to amino acid changes (c.304C >T/p.Pro102Ser and c.410T > C/p.Val137Ala), each variant being inherited from a parent. Follow-up imaging in adolescence revealed widened right cerebellar sulci. We discuss the evolving landscape of TBC1D24 associated phenotypes; this case adds to a growing body of evidence linking this gene to movement disorders in children.


Subject(s)
Ataxia/diagnosis , Ataxia/genetics , GTPase-Activating Proteins/genetics , Movement Disorders/diagnosis , Movement Disorders/genetics , Age of Onset , Ataxia/complications , Brain/diagnostic imaging , Brain/pathology , Humans , Infant , Male , Movement Disorders/complications , Mutation
5.
J Neurol ; 266(7): 1623-1632, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30937521

ABSTRACT

We report a young wheelchair-dependent patient with an unclear proximal myopathy and a heterozygous, de-novo Cav1.1-R1239G mutation suggesting hypokalemic periodic paralysis (HypoPP). Sonography showed a loss of the pennate pattern indicative of an edema, whereas fatty degeneration was excluded. Within 7 days of therapy with spironolactone, potassium and physical therapy, muscle strength almost completely normalized, a normal pennate pattern appeared and the edema was markedly reduced. She learned to walk without aid and to do sports and has continued to do so for 11 years until now. Over the years, we tested serum potassium values, muscle strength, muscle edema and muscular sodium content by 1.5 T, 3 T and 7 T 1H and 23Na magnetic resonance imaging. No fatty muscle degeneration developed. Muscular edema-like changes only occurred when she was pregnant and was set to reduced therapy. Because of the ability to do sports again, her mobility was further increased. Our observational study on this single patient may suggest that: (1) muscle imaging and molecular genetics are important diagnostic tools, (2) weakness in periodic paralysis may be reversible, and (3) continued adequate therapy may preserve muscle structure and strength on a longterm, whereas weakness due to fatty degeneration could be considered progressive and irreversible. Although HypoPP is a rare disease, it should be included in differential diagnosis not only if there is paroxysmal weakness, but also in cases of myopathy of unknown origin.


Subject(s)
Calcium Channels, L-Type , Hypokalemic Periodic Paralysis/diagnostic imaging , Hypokalemic Periodic Paralysis/drug therapy , Muscle Strength/drug effects , Muscle, Skeletal/diagnostic imaging , Spironolactone/administration & dosage , Adult , Calcium Channels, L-Type/genetics , Female , Humans , Hypokalemic Periodic Paralysis/genetics , Mineralocorticoid Receptor Antagonists/administration & dosage , Muscle Strength/physiology , Potassium/administration & dosage , Time Factors
6.
J Physiol ; 597(1): 225-235, 2019 01.
Article in English | MEDLINE | ID: mdl-30284249

ABSTRACT

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


Subject(s)
Myotonia Congenita/physiopathology , Osmolar Concentration , Animals , Bumetanide/pharmacology , Disease Models, Animal , Female , Male , Membrane Potentials/drug effects , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiology , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/physiology
7.
Sci Rep ; 8(1): 10372, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29991727

ABSTRACT

Hypokalemic periodic paralysis is a skeletal muscle disease characterized by episodic weakness associated with low serum potassium. We compared clinical and biophysical effects of R222W, the first hNaV1.4 domain I mutation linked to this disease. R222W patients exhibited a higher density of fibers with depolarized resting membrane potentials and produced action potentials that were attenuated compared to controls. Functional characterization of the R222W mutation in heterologous expression included the inactivation deficient IFM/QQQ background to isolate activation. R222W decreased sodium current and slowed activation without affecting probability. Consistent with the phenotype of muscle weakness, R222W shifted fast inactivation to hyperpolarized potentials, promoted more rapid entry, and slowed recovery. R222W increased the extent of slow inactivation and slowed its recovery. A two-compartment skeletal muscle fiber model revealed that defects in fast inactivation sufficiently explain action potential attenuation in patients. Molecular dynamics simulations showed that R222W disrupted electrostatic interactions within the gating pore, supporting the observation that R222W promotes omega current at hyperpolarized potentials. Sodium channel inactivation defects produced by R222W are the primary driver of skeletal muscle fiber action potential attenuation, while hyperpolarization-induced omega current produced by that mutation promotes muscle fiber depolarization.


Subject(s)
Action Potentials/genetics , Hypokalemic Periodic Paralysis/genetics , Muscle Weakness/physiopathology , Mutation , NAV1.4 Voltage-Gated Sodium Channel/genetics , Humans , Membrane Potentials , Molecular Dynamics Simulation , Muscle Fibers, Skeletal , Muscle Weakness/etiology
8.
Acta Myol ; 37(3): 193-203, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30838349

ABSTRACT

INTRODUCTION: Periodic paralyses (PP) are recurrent episodes of flaccid limb muscle weakness. Next to autosomal dominant forms, sporadic PP (SPP) cases are known but their genetics are unclear. METHODS: In a patient with hypokalemic SPP, we performed exome sequencing to identify a candidate gene. We sequenced this gene in 263 unrelated PP patients without any known causative mutations. Then we performed functional analysis of all variants found and molecular modelling for interpretation. RESULTS: Exome sequencing in the proband yielded three heterozygous variants predicted to be linked to disease. These encoded p.Thr140Met in the Kir2.2 potassium channel, p.Asp229Asn in protein kinase C theta, and p.Thr15943Ile in titin. Since all hitherto known causative PP genes code for ion channels, we studied the Kir2.2-encoding gene, KCNJ12, for involvement in PP pathogenesis. KCNJ12 screening in 263 PP patients revealed three further variants, each in a single individual and coding for p.Gly419Ser, p.Cys75Tyr, and p.Ile283Val. All four Kir2.2 variants were functionally expressed. Only p.Thr140Met displayed relevant functional alterations, i.e. homo-tetrameric channels produced almost no current, and hetero-tetrameric channels suppressed co-expressed wildtype Kir2.1 in a dominant-negative manner. Molecular modelling showed Kir2.2 p.Thr140Met to reduce movement of potassium ions towards binding sites in the hetero-tetramer pore compatible with a reduced maximal current. MD simulations revealed loss of hydrogen bonding with the p.Thr140Met substitution. DISCUSSION: The electrophysiological findings of p.Thr140Met are similar to those found in thyrotoxic PP caused by Kir2.6 mutations. Also, the homologous Thr140 residue is mutated in Kir2.6. This supports the idea that Kir2.2 p.Thr140Met conveys susceptibility to SPP and should be included in genetic screening.


Subject(s)
Genetic Predisposition to Disease/genetics , Hypokalemic Periodic Paralysis/genetics , Paralyses, Familial Periodic/genetics , Potassium Channels, Inwardly Rectifying/genetics , Adolescent , Adult , Computer Simulation , Electrophysiology , Humans , Hypokalemic Periodic Paralysis/physiopathology , Male , Middle Aged , Models, Molecular , Exome Sequencing
9.
Acta Myol ; 36(1): 2-13, 2017 03.
Article in English | MEDLINE | ID: mdl-28690388

ABSTRACT

In this pilot study we tested whether a low dose application of a mild diuretic substance such as eplerenone is beneficial in early stages of Duchenne muscular dystrophy using 23Na und 1H imaging, myometry, and clinical testing versus the glucocorticoid gold standard. Two 7-years old patients with DMD were examined on a 3T MRI system. 1H MRI and 23Na density-adapted 3-dimensional radial MRI sequences were performed both before and 1, 3 and 6 months after therapy with eplerenone respectively cortisone. We quantified fatty infiltration on T1-weighted images using subcutaneous fat as reference and fat fraction with a two-point DIXON sequence. Muscle oedema was quantified on STIR images and DIXON water maps with background noise as reference. We quantified Na+ by a muscular tissue concentration sequence with a 51.3mM Na+ with 5% agarose reference tube. A Na+ IR-sequence was used for determination of mainly myoplasmic Na+. Correspondingly myometry of muscles and tendons were assessed. Clinical tests (i.e. 4-steps-test) and blood counts (i.e. K+) were done by a pediatrician. Under eplerenone therapy we detected a reduction of muscular oedema, intracellular-weighted sodium IR signal and muscular sodium concentration. The oedema reduction in the DMD patient receiving eplerenone was more pronounced to the patient with cortisone. Myometric-measured tissue parameters such as muscle stiffness had a more pronounced effect in the child treated with eplerenone after a first increase in muscle stiffness both after eplerenone and cortisone treatment. Clinical abilities during both therapies were mostly constant. Eplerenone might be a possible new therapy option in DMD patients.


Subject(s)
Diuretics/therapeutic use , Glucocorticoids/therapeutic use , Magnetic Resonance Imaging , Muscular Dystrophy, Duchenne/diagnostic imaging , Muscular Dystrophy, Duchenne/drug therapy , Pregnenediones/therapeutic use , Spironolactone/analogs & derivatives , Adipose Tissue/diagnostic imaging , Child , Edema/diagnostic imaging , Elasticity , Eplerenone , Humans , Hydrogen , Imaging, Three-Dimensional , Male , Muscle, Skeletal/physiopathology , Pilot Projects , Sodium Radioisotopes , Spironolactone/therapeutic use
10.
Acta Myol ; 36(3): 125-134, 2017 Sep.
Article in English | MEDLINE | ID: mdl-29774303

ABSTRACT

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.


Subject(s)
Myotonia Congenita/complications , Myotonia Congenita/genetics , NAV1.4 Voltage-Gated Sodium Channel/genetics , Action Potentials , Adolescent , Adult , Age Factors , Child , Child, Preschool , Dyspnea/etiology , Exercise/physiology , Female , Flecainide/therapeutic use , Heterozygote , Humans , Hypertrophy , Infant , Infant, Newborn , Laryngismus/etiology , Male , Middle Aged , Muscle Weakness/etiology , Mutation , Myotonia Congenita/drug therapy , Myotonia Congenita/physiopathology , NAV1.4 Voltage-Gated Sodium Channel/physiology , Phenotype , Respiratory Sounds/etiology , Respiratory Tract Diseases/etiology , Voltage-Gated Sodium Channel Blockers/therapeutic use , Young Adult
11.
Sci Rep ; 6: 33372, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27646467

ABSTRACT

Malignant hyperthermia (MH) is a pharmacogenetic disorder of skeletal muscle metabolism which is characterized by generalized muscle rigidity, increased body temperature, rhabdomyolysis, and severe metabolic acidosis. The underlying mechanism of MH involves excessive Ca(2+) release in myotubes via the ryanodine receptor type 1 (RyR1). As RyR1 is also expressed in B-lymphocytes, this study investigated whether cellular metabolism of native B-lymphocytes was also altered in MH susceptible (MHS) individuals. A potent activator of RyR1, 4-chloro-m-cresol (4-CmC) was used to challenge native B-lymphocytes in a real-time, metabolic assay based on a pH-sensitive silicon biosensor chip. At the cellular level, a dose-dependent, phasic acidification occurred with 4-CmC. The acidification rate, an indicator of metabolic activation, was significantly higher in B-lymphocytes from MHS patients and required 3 to 5 fold lower concentrations of 4-CmC to evoke similar acidification rates to MHN. Native B-lymphocytes from MHS individuals are more sensitive to 4-CmC than those from MHN, reflecting a greater Ca(2+) turnover. The acidification response, however, was less pronounced than in muscle cells, presumably reflecting the lower expression of RyR1 in B-lymphocytes.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Disease Susceptibility , Energy Metabolism , Malignant Hyperthermia/immunology , Malignant Hyperthermia/metabolism , Adult , Biomarkers , Caffeine/metabolism , Caffeine/pharmacology , Calcium/metabolism , Cell Line, Transformed , Cresols/metabolism , Cresols/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Hydrogen-Ion Concentration , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Malignant Hyperthermia/genetics , Middle Aged , Mutation , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Young Adult
13.
Radiology ; 280(3): 848-59, 2016 09.
Article in English | MEDLINE | ID: mdl-27082780

ABSTRACT

Purpose To determine whether altered sodium (Na(+)) and chloride (Cl(-)) homeostasis can be visualized in periodic paralyses by using 7-T sodium 23 ((23)Na) and chlorine 35 ((35)Cl) magnetic resonance (MR) imaging. Materials and Methods Institutional review board approval and informed consent of all participants were obtained. (23)Na (repetition time msec/echo time msec, 160/0.35) and (35)Cl (40/0.6) MR imaging of both lower legs was performed with a 7-T whole-body system in patients with genetically confirmed hypokalemic periodic paralysis (Cav1.1-R1239H mutation, n = 5; Cav1.1-R528H mutation, n = 8) and Andersen-Tawil syndrome (n = 3) and in 16 healthy volunteers. Additionally, each participant underwent 3-T proton MR imaging on the same day by using T1-weighted, short-tau inversion-recovery, and Dixon-type sequences. Muscle edema was assessed on short-tau inversion-recovery images, fatty degeneration was assessed on T1-weighted images, and muscular fat fraction was quantified with Dixon-type imaging. Na(+) and Cl(-) were quantified in the soleus muscle by using three phantoms that contained 10-, 20-, and 30-mmol/L NaCl solution and 5% agarose gel as a reference. Parametric data for all subpopulations were tested by using one-way analysis of variance with the Dunnett test, and correlations were assessed with the Spearman rank correlation coefficient. Results Median muscular (23)Na concentration was higher in patients with Cav1.1-R1239H (34.7 mmol/L, P < .001), Cav1.1-R528H (32.0 mmol/L, P < .001), and Kir2.1 (24.3 mmol/L, P = .035) mutations than in healthy volunteers (19.9 mmol/L). Median muscular normalized (35)Cl signal intensity was higher in patients with Cav1.1-R1239H (27.6, P < .001) and Cav1.1-R528H (23.6, P < .001) than in healthy volunteers (12.6), but not in patients with the Kir2.1 mutation (14.3, P = .517). When compared with volunteers, patients with Cav1.1-R1239H and Cav1.1-R528H showed increased muscular edema (P < .001 and P = .003, respectively) and muscle fat fraction (P < .001 and P = .017, respectively). Conclusion With 7-T MR imaging, changes of Na(+) and Cl(-) homeostasis can be visualized in periodic paralyses and are most pronounced in the severe phenotype Cav1.1-R1239H, with up to daily paralytic episodes. (©) RSNA, 2016 An earlier incorrect version of this article appeared online. This article was corrected on April 18, 2016.


Subject(s)
Edema/diagnostic imaging , Hypokalemic Periodic Paralysis/diagnostic imaging , Leg/diagnostic imaging , Magnetic Resonance Imaging/methods , Adipose Tissue/diagnostic imaging , Adult , Aged , Case-Control Studies , Chlorine , Female , Humans , Hypokalemic Periodic Paralysis/genetics , Male , Middle Aged , Muscle, Skeletal/diagnostic imaging , Mutation , Phantoms, Imaging , Sodium Isotopes
14.
Cephalalgia ; 36(13): 1238-1247, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26763045

ABSTRACT

Introduction Familial hemiplegic migraine (FHM) is a rare autosomal dominant subtype of migraine with aura. The FHM3 subtype is caused by mutations in SCN1A, which is also the most frequent epilepsy gene encoding the voltage-gated Na+ channel NaV1.1. The aim of this study was to explore the clinical, genetic and pathogenetic features of a pure FHM3 family. Methods A three-generation family was enrolled in this study for genetic testing and assessment of clinical features. Whole cell patch-clamp was performed to determine the functions of identified mutant NaV1.1 channels, which were transiently expressed in human tsA201 cells together with ß1 and ß2 subunits. Results and conclusions We identified a novel SCN1A (p.Leu1624Pro) mutation in a pure FHM family with notably early-onset attacks at mean age of 7. L1624P locates in S3 of domain IV, the same domain as two of four known pure FHM3 mutations. Compared to WT channels, L1624P displayed an increased threshold-near persistent current in addition to other gain-of-function features such as: a slowing of fast inactivation, a positive shift in steady-state inactivation, a faster recovery and higher channel availability during repetitive stimulation. Similar to the known FHM3 mutations, this novel mutation predicts hyperexcitability of GABAergic inhibitory neurons.


Subject(s)
Genetic Predisposition to Disease/genetics , Genetic Testing/methods , Migraine with Aura/diagnosis , Migraine with Aura/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Polymorphism, Single Nucleotide/genetics , Adult , Child , Female , Genetic Markers/genetics , Humans , Male , Middle Aged , Mutation/genetics , Pedigree
15.
J Neurol Neurosurg Psychiatry ; 87(1): 49-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25882930

ABSTRACT

OBJECTIVE: To examine rare KCNJ18 variations recently reported to cause sporadic and thyrotoxic hypokalaemic periodic paralysis (TPP). METHODS: We sequenced KCNJ18 in 474 controls (400 Caucasians, 74 male Asians) and 263 unrelated patients with periodic paralysis (PP), including 30 patients with TPP without mutations in established PP genes. RESULTS: In 10 patients without TPP, we identified 9 heterozygous, novel variations (c.-3G>A, L15S, R81C, E273X, T309I, I340T, N365S, G394R, R401W) and a questionable heterozygous causative R399X stop variant. Studies on 40 relatives of these 10 patients showed that none of the variants were de novo in the patients and that R399X occurred in 3 non-affected relatives. Most affected amino acids lacked conservation and several clinically affected relatives did not carry the patient's variant. T309I, however, could be pathogenic under the pre-requisite of strongly reduced penetrance in females. Of the controls, 17 revealed 12 novel rare variants including the heterozygous E273X stop variant in three individuals. CONCLUSIONS: Our study shows many different, rare KCNJ18 alterations in patients as well as controls. Only perhaps one meets the requirements of a disease-causing mutation. Therefore, KCNJ18 alterations are seldom pathogenic. Additional studies are required before patients with PP can be genetically diagnosed on the basis of a KCNJ18 variant alone.


Subject(s)
Hypokalemic Periodic Paralysis/genetics , Potassium Channels, Inwardly Rectifying/genetics , Adolescent , Adult , Amino Acid Substitution/genetics , Asian People , Child , Conserved Sequence , DNA/genetics , Female , Genetic Variation , Heterozygote , Humans , Male , Middle Aged , Mutation/genetics , Penetrance , Young Adult
16.
Acta Myol ; 35(2): 86-89, 2016 Oct.
Article in English | MEDLINE | ID: mdl-28344437

ABSTRACT

Eplerenone, an aldosterone antagonist, repolarizes muscle membrane in-vitro and increases strength in-vivo in channelopathies. In Duchenne dystrophy, it is administered for cardiomyopathy. We studied its mechanism of action on skeletal muscle to test its suitability for increasing strength in Duchenne dystrophy. Using membrane potential measurements, quantitative PCR, ELISA, and Western blots, we examined the effects of eplerenone on skeletal muscle Na,K-ATPase. The repolarizing effect of eplerenone in muscle fibres was counteracted by oubain, an ATPase blocker. In our experiment, ATPA1A mRNA and total ATPase protein were not elevated. Instead, Tyr10 of the α1 subunit was dephosphorylated which would agree with ATPase activation. Dephosporylation of the coupled Akt kinase corroborated our findings. We conclude that eplerenone repolarizes muscle membrane by Na,K-ATPase activation by dephosphorylation at Tyr10. Since ATPase protein is known to be compensatorily increased in Duchenne patients without activity change, eplerenone treatment may be beneficial.


Subject(s)
Diaphragm/drug effects , Membrane Potentials/drug effects , Mineralocorticoid Receptor Antagonists/pharmacology , Muscular Dystrophy, Duchenne/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Spironolactone/analogs & derivatives , Animals , Cell Line , Diaphragm/metabolism , Enzyme Inhibitors/pharmacology , Eplerenone , Mice , Ouabain/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rats , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics , Spironolactone/pharmacology , Transcription, Genetic/drug effects
17.
Acta Myol ; 35(2): 107-108, 2016 Oct.
Article in English | MEDLINE | ID: mdl-28344441

ABSTRACT

Primary periodic paralyses (PPs) are autosomal dominant ion channel disorders characterized by episodic flaccid weakness associated with variations in serum potassium level. The main prophylactic therapy of choice for PPsis carbonic anhydrase inhibitors that are not always effective. In this report, we described two PP patients who were successfully treated with coenzyme Q10. They remained asymptomatic since initiation of treatment, which may be associated with promotion of energy synthesis, anti-oxidant activity, influence of the fiber type composition and regulation of the expression of gene. To our knowledge, this is the first report of primary periodic paralyses which have been successfully treated with CoQ10. More observations need to substantiate this clinical finding in PPs.


Subject(s)
Paralyses, Familial Periodic/drug therapy , Ubiquinone/analogs & derivatives , Vitamins/therapeutic use , Adult , Humans , Male , Middle Aged , NAV1.4 Voltage-Gated Sodium Channel/genetics , Paralyses, Familial Periodic/genetics , Ubiquinone/therapeutic use
18.
Muscle Nerve ; 52(4): 680-3, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26080010

ABSTRACT

INTRODUCTION: Two previously reported Norwegian patients with painful muscle cramps and giant myotonic discharges were genotyped and compared with those of members of 21 families harboring the same mutation. METHODS: Using primers specific for SCN4A and CLCN1, the DNA of the Norwegian family members was amplified and bidirectionally sequenced. Clinical and neurophysiological features of other families harboring the same mutation were studied. RESULTS: A G1306A mutation in the Nav1.4 voltage-gated sodium channel of skeletal muscle was identified. This mutation is known to cause myotonia fluctuans. No giant myotonic discharges or painful muscle cramps were found in the other G1306A families. CONCLUSIONS: Ephaptic transmission between neighboring muscle fibers may not only cause the unusual size of the myotonic discharges in this family, but also a more severe type of potassium-aggravated myotonia than myotonia fluctuans.


Subject(s)
Evoked Potentials, Motor/genetics , Family Health , Muscle Cramp/complications , Muscle Cramp/genetics , Mutation/genetics , NAV1.4 Voltage-Gated Sodium Channel/genetics , Adult , Electromyography , Female , Humans , Middle Aged
19.
Channels (Austin) ; 8(5): 467-71, 2014.
Article in English | MEDLINE | ID: mdl-25483590

ABSTRACT

Heterologous expression of sodium channel mutations in hypokalemic periodic paralysis reveals 2 variants on channel dysfunction. Charge-reducing mutations of voltage sensing S4 arginine residues alter channel gating as typically studied with expression in mammalian cells. These mutations also produce leak currents through the voltage sensor module, as typically studied with expression in Xenopus oocytes. DIIIS4 mutations at R3 in the skeletal muscle sodium channel produce gating defects and omega current consistent with the phenotype of reduced excitability. Here, we confirm DIIIS4 R3C gating defects in the oocyte expression system for fast inactivation and its recovery. We provide novel data for the effects of the cysteine mutation on voltage sensor movement, to further our understanding of sodium channel defects in hypokalemic periodic paralysis. Gating charge movement and its remobilization are selectively altered by the mutation at hyperpolarized membrane potential, as expected with reduced serum potassium.


Subject(s)
Hypokalemic Periodic Paralysis/genetics , Mutation/genetics , NAV1.4 Voltage-Gated Sodium Channel/chemistry , NAV1.4 Voltage-Gated Sodium Channel/metabolism , Animals , NAV1.4 Voltage-Gated Sodium Channel/genetics , Oocytes/metabolism , Protein Structure, Tertiary/genetics , Xenopus laevis
20.
Stud Health Technol Inform ; 198: 47-54, 2014.
Article in English | MEDLINE | ID: mdl-24825684

ABSTRACT

For patients suffering from rare diseases it is often hard to find an expert clinician. Existing registries rely on manual registration procedures and cannot easily be kept up to date. A prototype data collection system for discovering experts on rare diseases using MEDLINE has been successfully deployed. Initial manual analyses demonstrate proof of concept and deliver promising results. Examining the associations between authors, diseases and MeSH-Terms is expected to open up a variety of possibilities beyond expert discovery.


Subject(s)
Authorship , Bibliometrics , Clinical Competence/statistics & numerical data , Data Mining/methods , MEDLINE/statistics & numerical data , Rare Diseases/classification , Referral and Consultation/classification , Expert Testimony , Humans , Medical Subject Headings , Natural Language Processing , Vocabulary, Controlled
SELECTION OF CITATIONS
SEARCH DETAIL
...