Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 147(1): 224-239, 2024 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-37647766

RESUMEN

Genetic variants associated with developmental and epileptic encephalopathies have been identified in the GABRB3 gene that encodes the ß3 subunit of GABAA receptors. Typically, variants alter receptor sensitivity to GABA resulting in either gain- or loss-of-function, which correlates with patient phenotypes. However, it is unclear how another important receptor property, desensitization, contributes to the greater clinical severity of gain-of-function variants. Desensitization properties of 20 gain-of-function GABRB3 variant receptors were evaluated using two-electrode voltage-clamp electrophysiology. The parameters measured included current decay rates and steady-state currents. Selected variants with increased or reduced desensitization were also evaluated using whole-cell electrophysiology in transfected mammalian cell lines. Of the 20 gain-of-function variants assessed, 13 were found to alter receptor desensitization properties. Seven variants reduced desensitization at equilibrium, which acts to worsen gain-of-function traits. Six variants accelerated current decay kinetics, which limits gain-of-function traits. All affected patients displayed severe clinical phenotypes with intellectual disability and difficult-to-treat epilepsy. Nevertheless, variants that reduced desensitization at equilibrium were associated with more severe clinical outcomes. This included younger age of first seizure onset (median 0.5 months), movement disorders (dystonia and dyskinesia), epilepsy of infancy with migrating focal seizures (EIMFS) and risk of early mortality. Variants that accelerated current decay kinetics were associated with slightly milder phenotypes with later seizure onset (median 4 months), unclassifiable developmental and epileptic encephalopathies or Lennox-Gastaut syndrome and no movement disorders. Our study reveals that gain-of-function GABRB3 variants can increase or decrease receptor desensitization properties and that there is a correlation with the degree of disease severity. Variants that reduced the desensitization at equilibrium were clustered in the transmembrane regions that constitute the channel pore and correlated with greater disease severity, while variants that accelerated current decay were clustered in the coupling loops responsible for receptor activation and correlated with lesser severity.


Asunto(s)
Epilepsia Generalizada , Epilepsia , Trastornos del Movimiento , Animales , Humanos , Recién Nacido , Mutación con Ganancia de Función , Mutación/genética , Epilepsia/genética , Convulsiones , Mamíferos/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo
2.
Molecules ; 29(11)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38893312

RESUMEN

Gain-of-function mutations in the KCNT1 gene, which encodes the sodium-activated potassium channel known as SLACK, are associated with the rare but devastating developmental and epileptic encephalopathy known as epilepsy of infancy with migrating focal seizures (EIMFS). The design of small molecule inhibitors of SLACK channels represents a potential therapeutic approach to the treatment of EIMFS, other childhood epilepsies, and developmental disorders. Herein, we describe a hit optimization effort centered on a xanthine SLACK inhibitor (8) discovered via a high-throughput screen. Across three distinct regions of the chemotype, we synthesized 58 new analogs and tested each one in a whole-cell automated patch-clamp assay to develop structure-activity relationships for inhibition of SLACK channels. We further evaluated selected analogs for their selectivity versus a variety of other ion channels and for their activity versus clinically relevant SLACK mutants. Selectivity within the series was quite good, including versus hERG. Analog 80 (VU0948578) was a potent inhibitor of WT, A934T, and G288S SLACK, with IC50 values between 0.59 and 0.71 µM across these variants. VU0948578 represents a useful in vitro tool compound from a chemotype that is distinct from previously reported small molecule inhibitors of SLACK channels.


Asunto(s)
Bloqueadores de los Canales de Potasio , Relación Estructura-Actividad , Humanos , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/farmacología , Canales de potasio activados por Sodio , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Xantina/química , Xantina/farmacología , Técnicas de Placa-Clamp , Células HEK293 , Estructura Molecular , Xantinas/química , Xantinas/farmacología
3.
Bioorg Med Chem ; 95: 117487, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37812884

RESUMEN

Malignant migrating partial seizure of infancy (MMPSI) is a devastating and pharmacoresistant form of infantile epilepsy. MMPSI has been linked to multiple gain-of-function (GOF) mutations in the KCNT1 gene, which encodes for a potassium channel often referred to as SLACK. SLACK channels are sodium-activated potassium channels distributed throughout the central nervous system (CNS) and the periphery. The investigation described here aims to discover SLACK channel inhibitor tool compounds and profile their pharmacokinetic and pharmacodynamic properties. A SLACK channel inhibitor VU0531245 (VU245) was identified via a high-throughput screen (HTS) campaign. Structure-activity relationship (SAR) studies were conducted in five distinct regions of the hit VU245. VU245 analogs were evaluated for their ability to affect SLACK channel activity using a thallium flux assay in HEK-293 cells stably expressing wild-type (WT) human SLACK. Selected analogs were tested for metabolic stability in mouse liver microsomes and plasma-protein binding in mouse plasma. The same set of analogs was tested via thallium flux for activity versus human A934T SLACK and other structurally related potassium channels, including SLICK and Maxi-K. In addition, potencies for selected VU245 analogs were obtained using whole-cell electrophysiology (EP) assays in CHO cells stably expressing WT human SLACK through an automated patch clamp system. Results revealed that this scaffold tolerates structural changes in some regions, with some analogs demonstrating improved SLACK inhibitory activity, good selectivity against the other channels tested, and modest improvements in metabolic clearance. Analog VU0935685 represents a new, structurally distinct small-molecule inhibitor of SLACK channels that can serve as an in vitro tool for studying this target.


Asunto(s)
Canales de Potasio , Talio , Animales , Cricetinae , Humanos , Ratones , Cricetulus , Células HEK293 , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio/genética , Canales de Potasio/metabolismo , Canales de potasio activados por Sodio/genética , Canales de potasio activados por Sodio/metabolismo , Convulsiones , Talio/metabolismo , Oxadiazoles/química , Oxadiazoles/metabolismo
4.
Bioorg Med Chem Lett ; 76: 129013, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36184030

RESUMEN

In this Letter we describe structure-activity relationship (SAR) studies conducted in five distinct regions of a new 2-amino-N-phenylacetamides series of Slack potassium channel inhibitors exemplified by recently disclosed high-throughput screening (HTS) hit VU0606170 (4). New analogs were screened in a thallium (Tl+) flux assay in HEK-293 cells stably expressing wild-type human (WT) Slack. Selected analogs were screened in Tl+ flux versus A934T Slack and other Slo family members Slick and Maxi-K and evaluated in whole-cell electrophysiology (EP) assays using an automated patch clamp system. Results revealed the series to have flat SAR with significant structural modifications resulting in a loss of Slack activity. More minor changes led to compounds with Slack activity and Slo family selectivity similar to the HTS hit.


Asunto(s)
Canales de Potasio , Talio , Humanos , Células HEK293 , Proteínas del Tejido Nervioso/metabolismo , Canales de potasio activados por Sodio , Relación Estructura-Actividad
5.
J Neurosci ; 39(37): 7438-7449, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31350261

RESUMEN

Mutations in the KCNT1 (Slack, KNa1.1) sodium-activated potassium channel produce severe epileptic encephalopathies. Expression in heterologous systems has shown that the disease-causing mutations give rise to channels that have increased current amplitude. It is not known, however, whether such gain of function occurs in human neurons, nor whether such increased KNa current is expected to suppress or increase the excitability of cortical neurons. Using genetically engineered human induced pluripotent stem cell (iPSC)-derived neurons, we have now found that sodium-dependent potassium currents are increased several-fold in neurons bearing a homozygous P924L mutation. In current-clamp recordings, the increased KNa current in neurons with the P924L mutation acts to shorten the duration of action potentials and to increase the amplitude of the afterhyperpolarization that follows each action potential. Strikingly, the number of action potentials that were evoked by depolarizing currents as well as maximal firing rates were increased in neurons expressing the mutant channel. In networks of spontaneously active neurons, the mean firing rate, the occurrence of rapid bursts of action potentials, and the intensity of firing during the burst were all increased in neurons with the P924L Slack mutation. The feasibility of an increased KNa current to increase firing rates independent of any compensatory changes was validated by numerical simulations. Our findings indicate that gain-of-function in Slack KNa channels causes hyperexcitability in both isolated neurons and in neural networks and occurs by a cell-autonomous mechanism that does not require network interactions.SIGNIFICANCE STATEMENTKCNT1 mutations lead to severe epileptic encephalopathies for which there are no effective treatments. This study is the first demonstration that a KCNT1 mutation increases the Slack current in neurons. It also provides the first explanation for how this increased potassium current induces hyperexcitability, which could be the underlining factor causing seizures.


Asunto(s)
Epilepsia/genética , Células Madre Pluripotentes Inducidas/fisiología , Mutación/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas/fisiología , Canales de potasio activados por Sodio/genética , Potenciales de Acción/fisiología , Diferenciación Celular/fisiología , Epilepsia/fisiopatología , Células HEK293 , Humanos
6.
Cell Rep ; 43(3): 113904, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38457342

RESUMEN

The KCNT1 gene encodes the sodium-activated potassium channel Slack (KCNT1, KNa1.1), a regulator of neuronal excitability. Gain-of-function mutations in humans cause cortical network hyperexcitability, seizures, and severe intellectual disability. Using a mouse model expressing the Slack-R455H mutation, we find that Na+-dependent K+ (KNa) and voltage-dependent sodium (NaV) currents are increased in both excitatory and inhibitory cortical neurons. These increased currents, however, enhance the firing of excitability neurons but suppress that of inhibitory neurons. We further show that the expression of NaV channel subunits, particularly that of NaV1.6, is upregulated and that the length of the axon initial segment and of axonal NaV immunostaining is increased in both neuron types. Our study on the coordinate regulation of KNa currents and the expression of NaV channels may provide an avenue for understanding and treating epilepsies and other neurological disorders.


Asunto(s)
Epilepsia , Canales de Potasio , Humanos , Axones/metabolismo , Epilepsia/genética , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Canales de Potasio/metabolismo , Canales de potasio activados por Sodio , Animales , Ratones
7.
Pharm Pat Anal ; 11(2): 45-56, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35369761

RESUMEN

Slack channels are sodium-activated potassium channels that are encoded by the KCNT1 gene. Several KCNT1 gain of function mutations have been linked to malignant migrating partial seizures of infancy. Quinidine is an anti-arrhythmic drug that functions as a moderately potent inhibitor of Slack channels; however, quinidine use is limited by its poor selectivity, safety and pharmacokinetic profile. Slack channels represent an interesting target for developing novel therapeutics for the treatment of malignant migrating partial seizures of infancy and other childhood epilepsies; thus, ongoing efforts are directed toward the discovery of small-molecules that inhibit Slack currents. This review summarizes patent applications published in 2020-2021 that describe the discovery of novel small-molecule Slack inhibitors.


Asunto(s)
Epilepsia , Proteínas del Tejido Nervioso , Bloqueadores de los Canales de Potasio , Canales de potasio activados por Sodio , Niño , Epilepsia/tratamiento farmacológico , Humanos , Mutación , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de potasio activados por Sodio/antagonistas & inhibidores , Quinidina/uso terapéutico , Convulsiones/tratamiento farmacológico
8.
Trends Pharmacol Sci ; 42(8): 700-713, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34074526

RESUMEN

Gain-of-function (GOF) pathogenic variants of KCNT1, the gene encoding the largest known potassium channel subunit, KNa1.1, are associated with developmental and epileptic encephalopathies accompanied by severe psychomotor and intellectual disabilities. Blocking hyperexcitable KNa1.1 channels with quinidine, a class I antiarrhythmic drug, has shown variable success in patients in part because of dose-limiting off-target effects, poor blood-brain barrier (BBB) penetration, and low potency. In recent years, high-resolution cryogenic electron microscopy (cryo-EM) structures of the chicken KNa1.1 channel in different activation states have been determined, and animal models of the diseases have been generated. Alongside increasing information about the functional effects of GOF pathogenic variants on KNa1.1 channel behaviour and how they lead to hyperexcitability, these tools will facilitate the development of more effective treatment strategies. We review the range of KCNT1 variants and their functional effects, the challenges posed by current treatment strategies, and recent advances in finding more potent and selective therapeutic interventions for KCNT1-related epilepsies.


Asunto(s)
Epilepsia , Proteínas del Tejido Nervioso , Animales , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Humanos , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Canales de potasio activados por Sodio , Quinidina
9.
J Pediatr Neurosci ; 16(4): 323-326, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-36531778

RESUMEN

Epilepsy of infancy with migrating focal seizures (EIMFS) is a rare devastating infantile epileptic encephalopathy that is characterized by a unique electroencephalopgraphy (EEG) signature of multifocal simultaneous seizures. Although no definite etiology is understood for EIMFS, mutations in certain ion channels, are implicated. Similarly, phenylalanyl-tRNA synthetase 2 (FARS2) deficiency is a rare, autosomal recessive disorder of dysfunctional mitochondrial translation causing refractory seizures, lactic acidosis, and developmental regression with a variety of EEG findings. However, an EIMFS-like pattern on EEG in FARS2 deficiency has only recently been reported once. Herein, we describe a seven-week-old male with seizures where whole exome sequencing (WES) revealed pathogenic FARS2 variants and an EIMFS pattern on EEG. This case provides an insight on a novel genetic mechanism for EIMFS. We encourage early consideration of WES when EIMFS is detected to evaluate for FARS2 deficiency, especially in the setting of profound lactic acidosis.

10.
Epileptic Disord ; 22(3): 327-335, 2020 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-32597768

RESUMEN

Epilepsy of infancy with migrating focal seizures (EIMFS) is now a well-recognized early-onset syndrome included in the ILAE classification of the epilepsies. KCNT1 gain-of-function variants are identified in about half of patients. In the remaining cases, the underlying genetic component is far more heterogeneous with sporadic mutations occasionally reported in SCN1A, SCN2A, SLC12A5, TBC1D24, PLCB1, SLC25A22, and KCNQ2. Here, we report, for the first time, a homozygous deleterious variant in the FARS2 gene, identified using a 115-gene panel for monogenic epilepsies, in a patient with EIMFS. This boy was the second child born to healthy consanguineous parents. The first seizures occurred at six weeks of age. The patient rapidly developed severe epilepsy with focal discharges on EEG, migrating from one brain region to another, highly suggestive of EIMFS. At five months of age, he had daily multifocal clonic seizures and erratic myoclonic fits, which were not consistently related to spikes or spike-and-wave discharges. Neurological status was severely abnormal from onset and the patient died at 10 months of age from respiratory distress. Using the gene panel, a homozygous missense variant of FARS2 was identified, at Chr6 (GRCh37):g.5404829C>T, c.667C>T (NM_001318872.1), inherited from both parents, leading to an arginine-to-cysteine substitution, p.(Arg223Cys). FARS2 is a member of the mitochondrial aminoacyl tRNA transferase (ARS) enzymes. ARS variants are increasingly recognized causes of early-onset epileptic and neurodevelopmental encephalopathies, however, the associated epileptic phenotype is not completely described. This case shows that FARS2-related seizures can mimic EIMFS in the early stage of the disease. Furthermore, in the setting of migrating focal seizures of infancy, FARS2 should be considered as a further candidate gene, and increased lactate level and occurrence of refractory myoclonic seizures are possible key features to suspect FARS deficiency.


Asunto(s)
Proteínas Mitocondriales/genética , Fenilalanina-ARNt Ligasa/genética , Convulsiones/genética , Convulsiones/fisiopatología , Edad de Inicio , Codón sin Sentido , Consanguinidad , Resultado Fatal , Humanos , Lactante , Masculino
11.
Neurotherapeutics ; 16(3): 848-857, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31054119

RESUMEN

Pathogenic variants in KCNT1 represent an important cause of treatment-resistant epilepsy, for which an effective therapy has been elusive. Reports about the effectiveness of quinidine, a candidate precision therapy, have been mixed. We sought to evaluate the treatment responsiveness of patients with KCNT1-related epilepsy. We performed an observational study of 43 patients using a collaborative KCNT1 patient registry. We assessed treatment efficacy based upon clinical seizure reduction, side effects of quinidine therapy, and variant-specific responsiveness to treatment. Quinidine treatment resulted in a > 50% seizure reduction in 20% of patients, with rare patients achieving transient seizure freedom. Multiple other therapies demonstrated some success in reducing seizure frequency, including the ketogenic diet and vigabatrin, the latter particularly in patients with epileptic spasms. Patients with the best quinidine response had variants that clustered distal to the NADP domain within the RCK2 domain of the protein. Half of patients did not receive a quinidine trial. In those who did, nearly half did not achieve therapeutic blood levels. More favorable response to quinidine in patients with KCNT1 variants distal to the NADP domain within the RCK2 domain may suggest a variant-specific response.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia Refractaria/genética , Proteínas del Tejido Nervioso/genética , Canales de potasio activados por Sodio/genética , Adolescente , Niño , Preescolar , Epilepsia Refractaria/tratamiento farmacológico , Femenino , Humanos , Lactante , Masculino , Quinidina/uso terapéutico , Sistema de Registros , Resultado del Tratamiento
12.
Seizure ; 69: 99-104, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31004928

RESUMEN

PURPOSE: Next-generation sequencing (NGS) has made genetic testing of patients with epileptic encephalopathies easier - novel variants are discovered and new phenotypes described. Variants in the same gene - even the same variant - can cause different types of epilepsy and neurodevelopmental disorders. Our aim was to identify the genetic causes of epileptic encephalopathies in paediatric patients with complex phenotypes. METHODS: NGS was carried out for three patients with epileptic encephalopathies. Detailed clinical features, brain magnetic resonance imaging and electroencephalography were analysed. We searched the Human Gene Mutation Database for the published GABRG2 variants with clinical description of patients and composed a summary of the variants and their phenotypic features. RESULTS: We identified two novel de novo GABRG2 variants, p.P282T and p.S306F, with new phenotypes including neuroradiological evidence of neurodegeneration and epilepsy of infancy with migrating focal seizures (EIMFS). One patient carried previously reported p.P83S variant with autism spectrum disorder (ASD) phenotype that has not yet been described related to GABRG2 disorders and a more severe epilepsy phenotype than reported earlier. In all, the literature search yielded twenty-two articles describing 27 different variants that were divided into two categories: those with self-limiting epilepsies and febrile seizures and those with more severe drug-resistant epileptic encephalopathies. CONCLUSION: This study further expands the genotypic and phenotypic spectrum of epilepsies associated with GABRG2 variants. More knowledge is still needed about the influence of the environment, genetic background and other epilepsy susceptibility genes on the phenotype of the specific GABRG2 variants.


Asunto(s)
Trastorno del Espectro Autista/genética , Epilepsia/genética , Mutación/genética , Receptores de GABA-A/genética , Trastorno del Espectro Autista/diagnóstico , Niño , Preescolar , Epilepsias Mioclónicas/diagnóstico , Epilepsias Mioclónicas/genética , Epilepsia/diagnóstico , Femenino , Genotipo , Humanos , Lactante , Masculino , Fenotipo , Convulsiones Febriles/genética
13.
Epileptic Disord ; 21(1): 48-54, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30782581

RESUMEN

Several recent studies have reported potassium sodium-activated channel subfamily T member 1 (KCNT1) mutations in epilepsy patients on quinidine therapy. The efficacy and safety of quinidine for epilepsy treatment, however, remains controversial. We herein report the cases of four patients with KCNT1 mutations treated with quinidine. A reduction in seizures of more than 50% after quinidine treatment was observed in one patient with epilepsy of infancy with migrating focal seizures (EIMFS), whereas two patients with EIMFS and one with focal epilepsy did not achieve apparent seizure reduction. The relationship between quinidine dose and serum quinidine concentration was inconsistent, particularly at high quinidine doses. One patient with EIMFS developed ventricular tachycardia the day after an increase in quinidine dose from 114 to 126 mg/kg/day. The serum trough quinidine concentration and the corrected QT interval (QTc) before arrhythmia onset were 2.4 µg/ml and 420 ms, respectively, and peak serum quinidine concentration after arrhythmia onset was 9.4 µg/ml. Another patient with EIMFS showed aberrant intraventricular conduction with a quinidine dose of 74.5 mg/kg/day and a serum trough concentration of 3.2 µg/ml. Given that serum quinidine levels may elevate sharply after a dose increase, careful monitoring of electrocardiographs and serum concentrations is required. Based on a review of previous reports and our experience with this case, quinidine should be considered as a promising drug for patients with EIMFS harbouring KCNT1 mutations, however, its efficacy remains controversial due to the limited number of cases, and more information on optimal serum concentrations and appropriate titration methods is required.


Asunto(s)
Anticonvulsivantes/farmacología , Arritmias Cardíacas/inducido químicamente , Epilepsias Parciales/tratamiento farmacológico , Proteínas del Tejido Nervioso/genética , Canales de Potasio/genética , Quinidina/farmacología , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/efectos adversos , Anticonvulsivantes/sangre , Niño , Preescolar , Monitoreo de Drogas , Electrocardiografía , Femenino , Humanos , Lactante , Masculino , Canales de potasio activados por Sodio , Quinidina/administración & dosificación , Quinidina/efectos adversos , Quinidina/sangre
14.
Epileptic Disord ; 21(5): 443-448, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31617493

RESUMEN

Epilepsy of infancy with migrating focal seizures (EIMFS) is a rare epilepsy syndrome, characterized by an onset of multifocal seizures before the age of six months and a rather typical ictal EEG pattern. The ketogenic diet (KD) has been shown to be a treatment option in these patients with variable results. The KD is generally given by enteral formula or solid food, however, patients on the KD often have coexisting medical disorders that may impair the gastrointestinal tract and, in these cases, parenteral nutrition support may be needed. We present our experience with three patients who had been on the KD because of EIMFS, who were acutely unable to absorb nutrients through the intestinal tract. For these patients, we were unable to reach ketogenic ratios higher than 1.5:1 because of the limited fat intake via the parenteral route. This ratio, nevertheless, was adequate for maintenance of seizure control while allowing short-term bowel rest. Even though our report is limited as it provides no controlled evidence, ketogenic parenteral nutrition should be considered in children on the KD when enteral nutrition is not feasible. Special care should be taken to maintain ketosis and avoid undesired carbohydrates. Patients may respond well to ketogenic parenteral nutrition in spite of a lower ketogenic ratio.


Asunto(s)
Dieta Cetogénica , Epilepsias Parciales/dietoterapia , Epilepsia/dietoterapia , Convulsiones/dietoterapia , Adolescente , Dieta Cetogénica/métodos , Epilepsias Parciales/diagnóstico , Epilepsia/complicaciones , Epilepsia/diagnóstico , Síndromes Epilépticos/diagnóstico , Síndromes Epilépticos/dietoterapia , Femenino , Humanos , Lactante , Masculino , Nutrición Parenteral/métodos , Convulsiones/complicaciones , Convulsiones/diagnóstico , Resultado del Tratamiento
15.
Neurotherapeutics ; 15(4): 1112-1126, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30112700

RESUMEN

Epilepsy of infancy with migrating focal seizures (EIMFS) is a rare early-onset developmental epileptic encephalopathy resistant to anti-epileptic drugs. The most common cause for EIMFS is a gain-of-function mutation in the KCNT1 potassium channel gene, and treatment with the KCNT1 blocker quinidine has been suggested as a rational approach for seizure control in EIMFS patients. However, variable results on the clinical efficacy of quinidine have been reported. In the present study, we provide a detailed description of the clinical, genetic, in vitro, and in vivo electrophysiological profile and pharmacological responses to quinidine of 2 EIMFS unrelated patients with a heterozygous de novo KCNT1 mutation: c.2849G>A (p.R950Q) in patient 1 and c.2677G>A (p.E893K) in patient 2. When expressed heterologously in CHO cells, KCNT1 channels carrying each variant showed gain-of-function effects, and were more effectively blocked by quinidine when compared to wild-type KCNT1 channels. On the basis of these in vitro results, add-on quinidine treatment was started at 3 and 16 months of age in patients 1 and 2, respectively. The results obtained reveal that quinidine significantly reduced seizure burden (by about 90%) and improved quality of life in both patients, but failed to normalize developmental milestones, which persisted as severely delayed. Based on the present experience, early quinidine intervention associated with heart monitoring and control of blood levels is among the critical factors for therapy effectiveness in EIMFS patients with KCNT1 gain-of-function mutations. Multicenter studies are needed to establish a consensus protocol for patient recruitment, quinidine treatment modalities, and outcome evaluation, to optimize clinical efficacy and reduce risks as well as variability associated to quinidine use in such severe developmental encephalopathy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Canal de Potasio Kv.1.1/genética , Mutación/genética , Quinidina/uso terapéutico , Convulsiones/tratamiento farmacológico , Convulsiones/genética , Animales , Células CHO , Preescolar , Cricetulus , Relación Dosis-Respuesta a Droga , Electroencefalografía , Pruebas Genéticas , Humanos , Lactante , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Modelos Moleculares , Medicina de Precisión , Convulsiones/fisiopatología , Transfección
16.
Brain Dev ; 40(8): 724-727, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29625812

RESUMEN

SCN2A mutations have been identified in various encephalopathy phenotypes, ranging from benign familial neonatal-infantile seizure (BFNIS) to more severe forms of epileptic encephalopathy such as Ohtahara syndrome or epilepsy of infancy with migrating focal seizure (EIMFS). Thus far, no particularly effective treatment is available for severe epileptic encephalopathy caused by SCN2A mutations in children. We present the case of a boy who developed seizures on the third day of life and received a diagnosis of EIMFS based on his clinical presentations and electroencephalography reports. Antiepileptic drugs, namely oxcarbazepine, phenytoin, valproate, levetiracetam, and clonazepam, as well as adrenocorticotropic hormone therapy failed to reduce the severity of the seizures. Seizure pattern changed to infantile spasm with extensor thrust since 5 months of age. A ketogenic diet consisting of a medium-chain triglyceride recipe was introduced at 8 months of age and the seizures were resolved in the following 10 months. A de novo mutation in SCN2A (c.573G > T; p.W191C) was proven through next-generation sequencing.


Asunto(s)
Dieta Cetogénica , Epilepsia Refractaria/dietoterapia , Canal de Sodio Activado por Voltaje NAV1.2/genética , Convulsiones/dietoterapia , Espasmos Infantiles/dietoterapia , Encéfalo/fisiopatología , Epilepsia Refractaria/genética , Epilepsia Refractaria/fisiopatología , Humanos , Lactante , Masculino , Mutación , Convulsiones/genética , Convulsiones/fisiopatología , Espasmos Infantiles/genética , Espasmos Infantiles/fisiopatología
17.
Brain Dev ; 40(8): 728-732, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29699863

RESUMEN

A heterozygous mutation in the fibroblast growth factor 12 (FGF12) gene, which elevates the voltage dependence of neuronal sodium channel fast inactivation, was recently identified in some patients with epileptic encephalopathy. Here we report 1 Japanese patient diagnosed with early infantile epileptic encephalopathy (EIEE) and another diagnosed with epilepsy of infancy with migrating focal seizures (EIMFS). These 2 patients had an identical heterozygous missense mutation [c.341G>A:p.(Arg114His)] in FGF12 , which was identified with whole-exome sequencing. This mutation is identical to previously reported mutations in cases with early onset epileptic encephalopathy. One of our cases exhibited EIMFS, and this case responded to phenytoin and high-dose phenobarbital (PB). FGF12-related epileptic encephalopathy may exhibit diverse phenotypes and may respond to sodium channel blockers or high-dose PB.


Asunto(s)
Epilepsias Parciales/genética , Factores de Crecimiento de Fibroblastos/genética , Mutación Missense , Espasmos Infantiles/genética , Adulto , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Epilepsias Parciales/diagnóstico por imagen , Epilepsias Parciales/tratamiento farmacológico , Epilepsias Parciales/fisiopatología , Humanos , Lactante , Masculino , Fenotipo , Espasmos Infantiles/diagnóstico por imagen , Espasmos Infantiles/tratamiento farmacológico , Espasmos Infantiles/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA