Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Physiol Rev ; 101(4): 1633-1689, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33769100

RESUMEN

Voltage-gated sodium channels initiate action potentials in nerve, skeletal muscle, and other electrically excitable cells. Mutations in them cause a wide range of diseases. These channelopathy mutations affect every aspect of sodium channel function, including voltage sensing, voltage-dependent activation, ion conductance, fast and slow inactivation, and both biosynthesis and assembly. Mutations that cause different forms of periodic paralysis in skeletal muscle were discovered first and have provided a template for understanding structure, function, and pathophysiology at the molecular level. More recent work has revealed multiple sodium channelopathies in the brain. Here we review the well-characterized genetics and pathophysiology of the periodic paralyses of skeletal muscle and then use this information as a foundation for advancing our understanding of mutations in the structurally homologous α-subunits of brain sodium channels that cause epilepsy, migraine, autism, and related comorbidities. We include studies based on molecular and structural biology, cell biology and physiology, pharmacology, and mouse genetics. Our review reveals unexpected connections among these different types of sodium channelopathies.


Asunto(s)
Encéfalo/fisiopatología , Canalopatías/fisiopatología , Músculo Esquelético/fisiopatología , Canales de Sodio , Animales , Canalopatías/genética , Humanos , Ratones , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/fisiopatología , Canales de Sodio/genética
2.
Proc Natl Acad Sci U S A ; 121(14): e2309000121, 2024 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-38547067

RESUMEN

Apneic events are frightening but largely benign events that often occur in infants. Here, we report apparent life-threatening apneic events in an infant with the homozygous SCN1AL263V missense mutation, which causes familial hemiplegic migraine type 3 in heterozygous family members, in the absence of epilepsy. Observations consistent with the events in the infant were made in an Scn1aL263V knock-in mouse model, in which apnea was preceded by a large brainstem DC-shift, indicative of profound brainstem depolarization. The L263V mutation caused gain of NaV1.1 function effects in transfected HEK293 cells. Sodium channel blockade mitigated the gain-of-function characteristics, rescued lethal apnea in Scn1aL263V mice, and decreased the frequency of severe apneic events in the patient. Hence, this study shows that SCN1AL263V can cause life-threatening apneic events, which in a mouse model were caused by profound brainstem depolarization. In addition to being potentially relevant to sudden infant death syndrome pathophysiology, these data indicate that sodium channel blockers may be considered therapeutic for apneic events in patients with these and other gain-of-function SCN1A mutations.


Asunto(s)
Apnea , Mutación con Ganancia de Función , Bloqueadores de los Canales de Sodio , Animales , Humanos , Ratones , Apnea/tratamiento farmacológico , Apnea/genética , Tronco Encefálico , Células HEK293 , Migraña con Aura/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Bloqueadores de los Canales de Sodio/uso terapéutico , Lactante , Femenino
3.
J Neurochem ; 2023 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-37654020

RESUMEN

The past two decades have witnessed a wide range of studies investigating genetic variants of voltage-gated sodium (NaV ) channels, which are involved in a broad spectrum of diseases, including several types of epilepsy. We have reviewed here phenotypes and pathological mechanisms of genetic epilepsies caused by variants in NaV α and ß subunits, as well as of some relevant interacting proteins (FGF12/FHF1, PRRT2, and Ankyrin-G). Notably, variants of all these genes can induce either gain- or loss-of-function of NaV leading to either neuronal hyperexcitability or hypoexcitability. We present the results of functional studies obtained with different experimental models, highlighting that they should be interpreted considering the features of the experimental system used. These systems are models, but they have allowed us to better understand pathophysiological issues, ameliorate diagnostics, orientate genetic counseling, and select/develop therapies within a precision medicine framework. These studies have also allowed us to gain insights into the physiological roles of different NaV channels and of the cells that express them. Overall, our review shows the progress that has been made, but also the need for further studies on aspects that have not yet been clarified. Finally, we conclude by highlighting some significant themes of general interest that can be gleaned from the results of the work of the last two decades.

4.
Epilepsia ; 64(5): 1331-1347, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36636894

RESUMEN

OBJECTIVE: This study was undertaken to refine the spectrum of SCN1A epileptic disorders other than Dravet syndrome (DS) and genetic epilepsy with febrile seizures plus (GEFS+) and optimize antiseizure management by correlating phenotype-genotype relationship and functional consequences of SCN1A variants in a cohort of patients. METHODS: Sixteen probands carrying SCN1A pathogenic variants were ascertained via a national collaborative network. We also performed a literature review including individuals with SCN1A variants causing non-DS and non-GEFS+ phenotypes and compared the features of the two cohorts. Whole cell patch clamp experiments were performed for three representative SCN1A pathogenic variants. RESULTS: Nine of the 16 probands (56%) had de novo pathogenic variants causing developmental and epileptic encephalopathy (DEE) with seizure onset at a median age of 2 months and severe intellectual disability. Seven of the 16 probands (54%), five with inherited and two with de novo variants, manifested focal epilepsies with mild or no intellectual disability. Sodium channel blockers never worsened seizures, and 50% of patients experienced long periods of seizure freedom. We found 13 SCN1A missense variants; eight of them were novel and never reported. Functional studies of three representative variants showed a gain of channel function. The literature review led to the identification of 44 individuals with SCN1A variants and non-DS, non-GEFS+ phenotypes. The comparison with our cohort highlighted that DEE phenotypes are a common feature. SIGNIFICANCE: The boundaries of SCN1A disorders are wide and still expanding. In our cohort, >50% of patients manifested focal epilepsies, which are thus a frequent feature of SCN1A pathogenic variants beyond DS and GEFS+. SCN1A testing should therefore be included in the diagnostic workup of pediatric, familial and nonfamilial, focal epilepsies. Alternatively, non-DS/non-GEFS+ phenotypes might be associated with gain of channel function, and sodium channel blockers could control seizures by counteracting excessive channel function. Functional analysis evaluating the consequences of pathogenic SCN1A variants is thus relevant to tailor the appropriate antiseizure medication.


Asunto(s)
Epilepsias Mioclónicas , Epilepsias Parciales , Canal de Sodio Activado por Voltaje NAV1.1 , Humanos , Causalidad , Epilepsias Mioclónicas/tratamiento farmacológico , Epilepsias Mioclónicas/genética , Mutación con Ganancia de Función , Discapacidad Intelectual/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Fenotipo , Bloqueadores de los Canales de Sodio/uso terapéutico
5.
Brain ; 145(11): 3816-3831, 2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-35696452

RESUMEN

Brain voltage-gated sodium channel NaV1.1 (SCN1A) loss-of-function variants cause the severe epilepsy Dravet syndrome, as well as milder phenotypes associated with genetic epilepsy with febrile seizures plus. Gain of function SCN1A variants are associated with familial hemiplegic migraine type 3. Novel SCN1A-related phenotypes have been described including early infantile developmental and epileptic encephalopathy with movement disorder, and more recently neonatal presentations with arthrogryposis. Here we describe the clinical, genetic and functional evaluation of affected individuals. Thirty-five patients were ascertained via an international collaborative network using a structured clinical questionnaire and from the literature. We performed whole-cell voltage-clamp electrophysiological recordings comparing sodium channels containing wild-type versus variant NaV1.1 subunits. Findings were related to Dravet syndrome and familial hemiplegic migraine type 3 variants. We identified three distinct clinical presentations differing by age at onset and presence of arthrogryposis and/or movement disorder. The most severely affected infants (n = 13) presented with congenital arthrogryposis, neonatal onset epilepsy in the first 3 days of life, tonic seizures and apnoeas, accompanied by a significant movement disorder and profound intellectual disability. Twenty-one patients presented later, between 2 weeks and 3 months of age, with a severe early infantile developmental and epileptic encephalopathy and a movement disorder. One patient presented after 3 months with developmental and epileptic encephalopathy only. Associated SCN1A variants cluster in regions of channel inactivation associated with gain of function, different to Dravet syndrome variants (odds ratio = 17.8; confidence interval = 5.4-69.3; P = 1.3 × 10-7). Functional studies of both epilepsy and familial hemiplegic migraine type 3 variants reveal alterations of gating properties in keeping with neuronal hyperexcitability. While epilepsy variants result in a moderate increase in action current amplitude consistent with mild gain of function, familial hemiplegic migraine type 3 variants induce a larger effect on gating properties, in particular the increase of persistent current, resulting in a large increase of action current amplitude, consistent with stronger gain of function. Clinically, 13 out of 16 (81%) gain of function variants were associated with a reduction in seizures in response to sodium channel blocker treatment (carbamazepine, oxcarbazepine, phenytoin, lamotrigine or lacosamide) without evidence of symptom exacerbation. Our study expands the spectrum of gain of function SCN1A-related epilepsy phenotypes, defines key clinical features, provides novel insights into the underlying disease mechanisms between SCN1A-related epilepsy and familial hemiplegic migraine type 3, and identifies sodium channel blockers as potentially efficacious therapies. Gain of function disease should be considered in early onset epilepsies with a pathogenic SCN1A variant and non-Dravet syndrome phenotype.


Asunto(s)
Artrogriposis , Epilepsias Mioclónicas , Epilepsia , Migraña con Aura , Trastornos del Movimiento , Espasmos Infantiles , Humanos , Epilepsias Mioclónicas/tratamiento farmacológico , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/diagnóstico , Epilepsia/genética , Epilepsia/diagnóstico , Mutación con Ganancia de Función , Canal de Sodio Activado por Voltaje NAV1.1/genética , Fenotipo , Recién Nacido , Lactante
6.
Stem Cells ; 35(2): 374-385, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27664080

RESUMEN

Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and a leading cause of autism. FXS is due to the silencing of the Fragile X Mental Retardation Protein (FMRP), an RNA binding protein mainly involved in translational control, dendritic spine morphology and synaptic plasticity. Despite extensive studies, there is currently no cure for FXS. With the purpose to decipher the initial molecular events leading to this pathology, we developed a stem-cell-based disease model by knocking-down the expression of Fmr1 in mouse embryonic stem cells (ESCs). Repressing FMRP in ESCs increased the expression of amyloid precursor protein (APP) and Ascl1. When inducing neuronal differentiation, ßIII-tubulin, p27kip1 , NeuN, and NeuroD1 were upregulated, leading to an accelerated neuronal differentiation that was partially compensated at later stages. Interestingly, we observed that neurogenesis is also accelerated in the embryonic brain of Fmr1-knockout mice, indicating that our cellular model recapitulates the molecular alterations present in vivo. Importantly, we rescued the main phenotype of the Fmr1 knockdown cell line, not only by reintroducing FMRP but also by pharmacologically targeting APP processing, showing the role of this protein in the pathophysiology of FXS during the earliest steps of neurogenesis. Our work allows to define an early therapeutic window but also to identify more effective molecules for treating this disorder. Stem Cells 2017;35:374-385.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Neurogénesis , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/genética , Forma de la Célula/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Cinética , Ratones , Ratones Noqueados , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal/genética
8.
Proc Natl Acad Sci U S A ; 110(43): 17546-51, 2013 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-24101488

RESUMEN

Familial hemiplegic migraine (FHM) is a rare subtype of migraine with aura. Mutations causing FHM type 3 have been identified in SCN1A, the gene encoding the Nav1.1 Na(+) channel, which is also a major target of epileptogenic mutations and is particularly important for the excitability of GABAergic neurons. However, functional studies of NaV1.1 FHM mutations have generated controversial results. In particular, it has been shown that the NaV1.1-L1649Q mutant is nonfunctional when expressed in a human cell line because of impaired plasma membrane expression, similarly to NaV1.1 mutants that cause severe epilepsy, but we have observed gain-of-function effects for other NaV1.1 FHM mutants. Here we show that NaV1.1-L1649Q is nonfunctional because of folding defects that are rescuable by incubation at lower temperatures or coexpression of interacting proteins, and that a partial rescue is sufficient for inducing an overall gain of function because of the modifications in gating properties. Strikingly, when expressed in neurons, the mutant was partially rescued and was a constitutive gain of function. A computational model showed that 35% rescue can be sufficient for inducing gain of function. Interestingly, previously described folding-defective epileptogenic NaV1.1 mutants show loss of function also when rescued. Our results are consistent with gain of function as the functional effect of NaV1.1 FHM mutations and hyperexcitability of GABAergic neurons as the pathomechanism of FHM type 3.


Asunto(s)
Activación del Canal Iónico/genética , Migraña con Aura/genética , Mutación , Canal de Sodio Activado por Voltaje NAV1.1/genética , Algoritmos , Sustitución de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Simulación por Computador , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Humanos , Activación del Canal Iónico/fisiología , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Migraña con Aura/patología , Migraña con Aura/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.1/química , Canal de Sodio Activado por Voltaje NAV1.1/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Técnicas de Placa-Clamp
9.
Neurobiol Dis ; 75: 100-14, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25576396

RESUMEN

Mutations of the voltage gated Na(+) channel Na(V)1.1 (SCN1A) are important causes of different genetic epilepsies and can also cause familial hemiplegic migraine (FHM-III). In previous studies, some rescuable epileptogenic folding defective mutants located in domain IV of Na(V)1.1 have been identified, showing partial loss of function also with maximal rescue. Variable rescue may be one of the causes of phenotypic variability, and rescue might be exploited for therapeutic approaches. Recently, we have identified a folding defective FHM-III Na(V)1.1 mutant that showed overall gain of function when rescued, consistent with a differential pathomechanism. Here, we have evaluated functional properties and cell surface expression of six Na(V)1.1 epileptogenic missense mutations in different rescuing conditions, including a novel one that we have developed expressing a selective sodium channel toxin (CsEI) targeted to the endoplasmic reticulum (ER). All the mutants showed loss of function and reduced cell surface expression, consistently with possibility of rescue. Four of them were rescuable by incubation at low temperature and interactions with different co-expressed proteins or a pharmacological chaperone (phenytoin). Notably, CsEI was able to rescue four mutants. Thus, Na(V)1.1 folding defective mutants can be relatively common and mutations inducing rescuable folding defects are spread in all Na(V)1.1 domains. Importantly, epileptogenic mutants showed overall loss of function even upon rescue, differently than FHM-III ones. The effectiveness of CsEI demonstrates that interactions in the ER are sufficient for inducing rescue, and provides a proof of concept for developing possible therapeutic approaches that may overcome some limitations of pharmacological chaperones.


Asunto(s)
Retículo Endoplásmico/metabolismo , Epilepsia/genética , Epilepsia/metabolismo , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Western Blotting , Línea Celular Transformada , Retículo Endoplásmico/efectos de los fármacos , Escherichia coli , Humanos , Inmunohistoquímica , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Moduladores del Transporte de Membrana/farmacología , Modelos Neurológicos , Canal de Sodio Activado por Voltaje NAV1.1/química , Técnicas de Placa-Clamp , Pliegue de Proteína , Venenos de Escorpión/farmacología , Transfección
10.
Epilepsia ; 54(5): 927-35, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23398611

RESUMEN

PURPOSE: To report the identification of the T1174S SCN1A (NaV 1.1) mutation in a three-generation family with both epileptic and familial hemiplegic migraine (FHM) phenotypes and clarify the pathomechanism. METHODS: The five affected individuals underwent detailed clinical analyses. Mutation analyses was performed by direct sequencing of SCN1A; functional studies by expression in tsA-201 cells. A computational model was used to compare the effects of T1174S with those of a typical FHM mutation (Q1489K). KEY FINDINGS: The proband had benign occipital epilepsy (BOE); two relatives had simple febrile seizures (FS) and later developed BOE. Two additional relatives had FHM without epilepsy or FS. All affected members and one obliged carrier carried the T1174S mutation. Functional effects were divergent: positive shift of the activation curve and deceleration of recovery from fast inactivation, consistent with loss of function, and increase of persistent current (I(NaP)), consistent with gain of function. The I(NaP) increase was inhibited by dialysis of the cytoplasm, consistent with a modulation. Therefore, as shown by the computational model, T1174S could in some conditions induce overall loss of function, and in others gain of function; Q1489K induced gain of function in all the conditions. SIGNIFICANCE: Modulation of the properties of T1174S can lead to a switch between overall gain and loss of function, consistent with a switch between promigraine end epileptogenic effect and, thus, with coexistence of epileptic and FHM phenotypes in the same family. These findings may help to shed light on the complex genotype-phenotype relationship of SCN1A mutations.


Asunto(s)
Migraña con Aura/complicaciones , Migraña con Aura/genética , Mutación/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Convulsiones/complicaciones , Convulsiones/genética , Adolescente , Adulto , Línea Celular Transformada , Simulación por Computador , Análisis Mutacional de ADN , Estimulación Eléctrica , Femenino , Humanos , Italia , Masculino , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Persona de Mediana Edad , Modelos Moleculares , Técnicas de Placa-Clamp , Fenotipo , Serina/genética , Treonina/genética , Adulto Joven
11.
Epilepsia ; 54(8): e112-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23899126

RESUMEN

There has been increased interest in a possible association between epilepsy channelopathies and cardiac arrhythmias, such as long QT syndrome (LQTS). We report a kindred that features LQTS, idiopathic epilepsy, and increased risk of sudden death. Genetic study showed a previously unreported heterozygous point mutation (c.246T>C) in the KCNH2 gene. Functional studies showed that the mutation induces severe loss of function. This observation provides further evidence for a possible link between idiopathic epilepsy and LQTS.


Asunto(s)
Muerte Súbita , Epilepsia/genética , Canales de Potasio Éter-A-Go-Go/genética , Salud de la Familia , Síndrome de QT Prolongado/genética , Adolescente , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Fenómenos Biofísicos/genética , Línea Celular Transformada , Análisis Mutacional de ADN , Canal de Potasio ERG1 , Estimulación Eléctrica , Electrocardiografía , Epilepsia/complicaciones , Femenino , Humanos , Síndrome de QT Prolongado/complicaciones , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Técnicas de Placa-Clamp , Mutación Puntual/genética , Transfección , Gemelos Dicigóticos/genética
12.
Biomed Pharmacother ; 165: 115173, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37453200

RESUMEN

Nav1.1 is an important pharmacological target as this voltage-gated sodium channel is involved in neurological and cardiac syndromes. Channel activators are actively sought to try to compensate for haploinsufficiency in several of these pathologies. Herein we used a natural source of new peptide compounds active on ion channels and screened for drugs capable to inhibit channel inactivation as a way to compensate for decreased channel function. We discovered that JzTx-34 is highly active on Nav1.1 and subsequently performed a full structure-activity relationship investigation to identify its pharmacophore. These experiments will help interpret the mechanism of action of this and formerly identified peptides as well as the future identification of new peptides. We also reveal structural determinants that make natural ICK peptides active against Nav1.1 challenging to synthesize. Altogether, the knowledge gained by this study will help facilitate the discovery and development of new compounds active on this critical ion channel target.


Asunto(s)
Péptidos , Canales de Sodio Activados por Voltaje , Humanos , Péptidos/farmacología , Péptidos/química , Relación Estructura-Actividad
13.
J Clin Invest ; 131(21)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34491914

RESUMEN

Spreading depolarizations (SDs) are involved in migraine, epilepsy, stroke, traumatic brain injury, and subarachnoid hemorrhage. However, the cellular origin and specific differential mechanisms are not clear. Increased glutamatergic activity is thought to be the key factor for generating cortical spreading depression (CSD), a pathological mechanism of migraine. Here, we show that acute pharmacological activation of NaV1.1 (the main Na+ channel of interneurons) or optogenetic-induced hyperactivity of GABAergic interneurons is sufficient to ignite CSD in the neocortex by spiking-generated extracellular K+ build-up. Neither GABAergic nor glutamatergic synaptic transmission were required for CSD initiation. CSD was not generated in other brain areas, suggesting that this is a neocortex-specific mechanism of CSD initiation. Gain-of-function mutations of NaV1.1 (SCN1A) cause familial hemiplegic migraine type-3 (FHM3), a subtype of migraine with aura, of which CSD is the neurophysiological correlate. Our results provide the mechanism linking NaV1.1 gain of function to CSD generation in FHM3. Thus, we reveal the key role of hyperactivity of GABAergic interneurons in a mechanism of CSD initiation, which is relevant as a pathological mechanism of Nav1.1 FHM3 mutations, and possibly also for other types of migraine and diseases in which SDs are involved.


Asunto(s)
Depresión de Propagación Cortical , Neuronas GABAérgicas/metabolismo , Interneuronas/metabolismo , Trastornos Migrañosos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Neocórtex/metabolismo , Animales , Neuronas GABAérgicas/patología , Interneuronas/patología , Ratones , Ratones Transgénicos , Trastornos Migrañosos/genética , Trastornos Migrañosos/patología , Canal de Sodio Activado por Voltaje NAV1.1/genética , Neocórtex/patología
14.
J Neurosci ; 28(29): 7273-83, 2008 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-18632931

RESUMEN

Familial hemiplegic migraine (FHM) is an autosomal dominant inherited subtype of severe migraine with aura. Mutations causing FHM (type 3) have been identified in SCN1A, the gene encoding neuronal voltage-gated Na(v)1.1 Na(+) channel alpha subunit, but functional studies have been done using the cardiac Na(v)1.5 isoform, and the observed effects were similar to those of some epileptogenic mutations. We studied the FHM mutation Q1489K by transfecting tsA-201 cells and cultured neurons with human Na(v)1.1. We show that the mutation has effects on the gating properties of the channel that can be consistent with both hyperexcitability and hypoexcitability. Simulation of neuronal firing and long depolarizing pulses mimicking promigraine conditions revealed that the effect of the mutation is a gain of function consistent with increased neuronal firing. However, during high-frequency discharges and long depolarizations, the effect became a loss of function. Recordings of firing of transfected neurons showed higher firing frequency at the beginning of long discharges. This self-limited capacity to induce neuronal hyperexcitability may be a specific characteristic of migraine mutations, able to both trigger the cascade of events that leads to migraine and counteract the development of extreme hyperexcitability typical of epileptic seizures. Thus, we found a possible difference in the functional effects of FHM and familial epilepsy mutations of Nav1.1.


Asunto(s)
Potenciales de Acción/fisiología , Activación del Canal Iónico/fisiología , Migraña con Aura/genética , Migraña con Aura/fisiopatología , Mutación , Proteínas del Tejido Nervioso/fisiología , Canales de Sodio/fisiología , Potenciales de Acción/genética , Animales , Línea Celular , Células Cultivadas , Glutamina/genética , Humanos , Activación del Canal Iónico/genética , Lisina/genética , Migraña con Aura/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1 , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Ratas , Canales de Sodio/genética
15.
Hum Mutat ; 30(7): E747-60, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19402159

RESUMEN

Mutations of voltage-gated Na(+) channels are the most common known cause of genetically determined epilepsy; Na(v)1.1 (SCN1A) is the most frequent target. They can cause both mild and severe forms, also in patients harboring the same mutation. We have recently characterized in a family with extreme phenotypes the first epileptogenic folding-defective Na(+) channel mutant (Na(v)1.1-M1841T), whose loss of function is attenuated by interactions with associated proteins and drugs. We hypothesized that in vivo variability of the interactions may modulate the functional effect and thus the phenotype (Rusconi et al., 2007). Here we characterize another Na(v)1.1 folding-defective mutant (Na(v)1.1-R1916G) that, however, has been identified in a GEFS+ family with relatively mild phenotypes. This novel mutant shows a number of specific characteristics, but, similarly to Na(v)1.1-M1841T, it can be rescued by interactions with associated proteins and drugs. Thus, loss of function caused by folding defects that can be attenuated by molecular interactions may be a common pathogenic mechanism for Na(v)1.1 epileptogenic mutants. Folding defects can be present also in families showing only mild phenotypes in which, however, severe phenotypes could emerge within a permissive genetic background.


Asunto(s)
Epilepsia/etiología , Proteínas Mutantes , Proteínas del Tejido Nervioso/genética , Canales de Sodio/genética , Línea Celular , ADN Complementario , Epilepsia/genética , Salud de la Familia , Humanos , Proteínas Mutantes/fisiología , Canal de Sodio Activado por Voltaje NAV1.1 , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/fisiología , Técnicas de Placa-Clamp , Fenotipo , Pliegue de Proteína , Canales de Sodio/química , Canales de Sodio/fisiología , Transfección
16.
Biochim Biophys Acta ; 1770(8): 1161-8, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17544584

RESUMEN

The gene of the four disulfide-bridged Centruroides suffusus suffusus toxin II was cloned into the expression vector pQE30 containing a 6His-tag and a FXa proteolytic cleavage region. This recombinant vector was transfected into Escherichia coli BL21 cells and expressed under induction with isopropyl thiogalactoside (IPTG). The level of expression was 24.6 mg/l of culture medium, and the His tagged recombinant toxin (HisrCssII) was found exclusively in inclusion bodies. After solubilization the HisrCssII peptide was purified by affinity and hydrophobic interaction chromatography. The reverse-phase HPLC profile of the HisrCssII product obtained from the affinity chromatography step showed several peptide fractions having the same molecular mass of 9392.6 Da, indicating that HisrCssII was oxidized forming several distinct disulfide bridge arrangements. The multiple forms of HisrCssII after reduction eluted from the column as a single protein component of 9400.6 Da. Similarly, an in vitro folding of the reduced HisrCssII generated a single oxidized component of HisrCssII, which was cleaved by the proteolytic enzyme FXa to the recombinant CssII (rCssII). The molecular mass of rCssII was 7538.6 Da as expected. Since native CssII (nCssII) is amidated at the C-terminal residue whereas the rCssII is heterologously expressed in the format of free carboxyl end, there is a difference of 1 Da, when comparing both peptides (native versus heterologously expressed). Nevertheless, they show similar toxicity when injected intracranially into mice, and both nCssII and rCssII show the typical electrophysiological properties of beta-toxins in Na(v)1.6 channels, which is for the first time demonstrated here. Binding and displacement experiments conducted with radiolabelled CssII confirms the electrophysiological results. Several problems associated with the heterologously expressed toxins containing four disulfide bridges are discussed.


Asunto(s)
Disulfuros/química , Pliegue de Proteína , Venenos de Escorpión/química , Venenos de Escorpión/metabolismo , Animales , Línea Celular , Dicroismo Circular , Clonación Molecular , Relación Dosis-Respuesta a Droga , Escherichia coli/genética , Genes Sintéticos , Histidina/química , Humanos , Técnicas In Vitro , Cuerpos de Inclusión/metabolismo , Inyecciones Intraperitoneales , Isopropil Tiogalactósido/farmacología , Dosificación Letal Mediana , Masculino , Ratones , Ratones Endogámicos , Peso Molecular , Neurotoxinas/química , Neurotoxinas/genética , Neurotoxinas/metabolismo , Neurotoxinas/farmacología , Oxidación-Reducción , Técnicas de Placa-Clamp , Plásmidos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Venenos de Escorpión/genética , Venenos de Escorpión/aislamiento & purificación , Venenos de Escorpión/farmacología , Canales de Sodio/metabolismo , Transfección
17.
Neurosci Lett ; 667: 92-102, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29129678

RESUMEN

Migraine and epilepsy are episodic disorders with distinct features, but they have some clinical and pathophysiological overlaps. We review here clinical overlaps between seizures and migraine attacks, activities of neuronal networks observed during seizures and migraine attacks, and molecular and cellular mechanisms of migraine identified in genetic forms, focusing on genetic variants identified in hemiplegic migraine and their functional effects. Epilepsy and migraine can be generated by dysfunctions of the same neuronal networks, but these dysfunctions can be disease-specific, even if pathogenic mutations target the same protein. Studies of rare monogenic forms have allowed the identification of some molecular/cellular dysfunctions that provide a window on pathological mechanisms: we have begun to disclose the tip of the iceberg.


Asunto(s)
Epilepsia/genética , Trastornos Migrañosos/genética , Migraña con Aura/genética , Mutación/genética , Animales , Canales de Calcio/genética , Epilepsia/fisiopatología , Humanos , Trastornos Migrañosos/fisiopatología , Convulsiones/genética , Convulsiones/fisiopatología
18.
Neuropharmacology ; 132: 31-42, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28571716

RESUMEN

Channelopathies comprise various diseases caused by defects of ion channels. Modifications of their biophysical properties are common and have been widely studied. However, ion channels are heterogeneous multi-molecular complexes that are extensively modulated and undergo a maturation process comprising numerous steps of structural modifications and intracellular trafficking. Perturbations of these processes can give rise to aberrant channels that cause pathologies. Here we review channelopathies of the nervous system associated with dysfunctions at the post-translational level (folding, trafficking, degradation, subcellular localization, interactions with associated proteins and structural post-translational modifications). We briefly outline the physiology of ion channels' maturation and discuss examples of defective mechanisms, focusing in particular on voltage-gated sodium channels, which are implicated in numerous neurological disorders. We also shortly introduce possible strategies to develop therapeutic approaches that target these processes. This article is part of the Special Issue entitled 'Channelopathies.'


Asunto(s)
Canalopatías/metabolismo , Sistema Nervioso/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Humanos
19.
Front Mol Neurosci ; 11: 232, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30038559

RESUMEN

The SCN1A gene encodes for the voltage-dependent Nav1.1 Na+ channel, an isoform mainly expressed in GABAergic neurons that is the target of hundreds of epileptogenic mutations. More recently, it has been shown that the SCN1A gene is also the target of mutations responsible for familial hemiplegic migraine (FHM-3), a rare autosomal dominant subtype of migraine with aura. Studies of these mutations indicate that they induce gain of function of the channel. Surprisingly, the mutation L1649Q responsible for pure FHM-3 showed a complete loss of function, but, when partially rescued it induced an overall gain of function because of modification of the gating properties of the mutant channel. Here, we report the characterization of the L1670W SCN1A mutation that has been previously identified in a Chinese family with pure FHM-3, and that we have identified also in a Caucasian American family with pure FHM-3. Notably, one patient in our family had severe neurological deterioration after brain radiation for cancer treatment. Functional analysis of L1670W reveals that the mutation is responsible for folding/trafficking defects and, when they are rescued by incubation at lower temperature or by expression in neurons, modifications of the gating properties lead to an overall gain of function. Therefore, L1670W is the second mutation responsible for FHM-3 with this pathophysiological mechanism, showing that it may be a recurrent mechanism for Nav1.1 hemiplegic migraine mutations.

20.
Lancet Neurol ; 17(8): 699-708, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30033060

RESUMEN

BACKGROUND: Genetic generalised epilepsy is the most common type of inherited epilepsy. Despite a high concordance rate of 80% in monozygotic twins, the genetic background is still poorly understood. We aimed to investigate the burden of rare genetic variants in genetic generalised epilepsy. METHODS: For this exome-based case-control study, we used three different genetic generalised epilepsy case cohorts and three independent control cohorts, all of European descent. Cases included in the study were clinically evaluated for genetic generalised epilepsy. Whole-exome sequencing was done for the discovery case cohort, a validation case cohort, and two independent control cohorts. The replication case cohort underwent targeted next-generation sequencing of the 19 known genes encoding subunits of GABAA receptors and was compared to the respective GABAA receptor variants of a third independent control cohort. Functional investigations were done with automated two-microelectrode voltage clamping in Xenopus laevis oocytes. FINDINGS: Statistical comparison of 152 familial index cases with genetic generalised epilepsy in the discovery cohort to 549 ethnically matched controls suggested an enrichment of rare missense (Nonsyn) variants in the ensemble of 19 genes encoding GABAA receptors in cases (odds ratio [OR] 2·40 [95% CI 1·41-4·10]; pNonsyn=0·0014, adjusted pNonsyn=0·019). Enrichment for these genes was validated in a whole-exome sequencing cohort of 357 sporadic and familial genetic generalised epilepsy cases and 1485 independent controls (OR 1·46 [95% CI 1·05-2·03]; pNonsyn=0·0081, adjusted pNonsyn=0·016). Comparison of genes encoding GABAA receptors in the independent replication cohort of 583 familial and sporadic genetic generalised epilepsy index cases, based on candidate-gene panel sequencing, with a third independent control cohort of 635 controls confirmed the overall enrichment of rare missense variants for 15 GABAA receptor genes in cases compared with controls (OR 1·46 [95% CI 1·02-2·08]; pNonsyn=0·013, adjusted pNonsyn=0·027). Functional studies for two selected genes (GABRB2 and GABRA5) showed significant loss-of-function effects with reduced current amplitudes in four of seven tested variants compared with wild-type receptors. INTERPRETATION: Functionally relevant variants in genes encoding GABAA receptor subunits constitute a significant risk factor for genetic generalised epilepsy. Examination of the role of specific gene groups and pathways can disentangle the complex genetic architecture of genetic generalised epilepsy. FUNDING: EuroEPINOMICS (European Science Foundation through national funding organisations), Epicure and EpiPGX (Sixth Framework Programme and Seventh Framework Programme of the European Commission), Research Unit FOR2715 (German Research Foundation and Luxembourg National Research Fund).


Asunto(s)
Epilepsia Generalizada/genética , Secuenciación del Exoma/métodos , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Receptores de GABA-A/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Niño , Preescolar , Estudios de Cohortes , Epilepsia Generalizada/etnología , Europa (Continente) , Salud de la Familia , Femenino , Humanos , Lactante , Recién Nacido , Cooperación Internacional , Masculino , Persona de Mediana Edad , Modelos Moleculares , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA