Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Cell Physiol ; 327(3): C790-C797, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39099423

RESUMEN

Within the tetramerization domain (T1) of most voltage-gated potassium channels (Kv) are highly conserved charged residues that line the T1-T1 interface. We investigated the Kv1.1 residue R86 located at the narrowest region of the T1 interface. A Kv1.1 R86Q mutation was reported in a child diagnosed with lower limb dyskinesia (Set KK, Ghosh D, Huq AHM, Luat AF. Mov Disord Clin Pract 4: 784-786, 2017). The child did not present with episodic ataxia 1 (EA1) symptoms typically associated with Kv1.1 loss-of-function mutations. We characterized the electrophysiological outcome of the R86Q substitution by expressing Kv1.1 in Xenopus laevis oocytes. Mutated α-subunits were able to form functional channels that pass delayed rectifier currents. Oocytes that expressed only mutated α-subunits produced a significant reduction in Kv1.1 current and showed a positive shift in voltage dependence of activation. In addition, there was substantially slower activation and faster deactivation implying a reduction in the time the channel is in its open state. Oocytes co-injected with both mutated and wild-type cRNA in equal amounts, to mimic the heterozygous condition of the disease, showed a decrease in current amplitude at -10 mV, a positive shift in activation voltage-dependence and faster deactivation kinetics when compared with the wild-type channel. These findings indicate that T1 plays a role in Kv1.1's voltage-dependent activation and in its kinetics of activation and deactivation.NEW & NOTEWORTHY This is the first Kv1.1 study to characterize the electrophysiological and structural phenotype of a tetramerization (T1) domain mutation. Surprisingly, the mutated α-subunits were able to tetramerize, albeit with different gating kinetics and voltage dependence. This novel finding points to a clear role of T1 in the channel's voltage dependence and gating. Mimicking the heterozygous condition resulted in milder alterations in channel function when compared with previously reported mutations. This is in agreement with the child's milder symptoms.


Asunto(s)
Activación del Canal Iónico , Canal de Potasio Kv.1.1 , Oocitos , Xenopus laevis , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv.1.1/metabolismo , Canal de Potasio Kv.1.1/química , Animales , Humanos , Oocitos/metabolismo , Cinética , Mutación , Potenciales de la Membrana , Multimerización de Proteína , Femenino
2.
Neurobiol Dis ; 199: 106592, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38971479

RESUMEN

Failure to recover from repeated hypercapnia and hypoxemia (HH) challenges caused by severe GCS and postictal apneas may contribute to sudden unexpected death in epilepsy (SUDEP). Our previous studies found orexinergic dysfunction contributes to respiratory abnormalities in a preclinical model of SUDEP, Kcna1-/- mice. Here, we developed two gas challenges consisting of repeated HH exposures and used whole body plethysmography to determine whether Kcna1-/- mice have detrimental ventilatory responses. Kcna1-/- mice exhibited an elevated ventilatory response to a mild repeated hypercapnia-hypoxia (HH) challenge compared to WT. Moreover, 71% of Kcna1-/- mice failed to survive a severe repeated HH challenge, whereas all WT mice recovered. We next determined whether orexin was involved in these differences. Pretreating Kcna1-/- mice with a dual orexin receptor antagonist rescued the ventilatory response during the mild challenge and all subjects survived the severe challenge. In ex vivo extracellular recordings in the lateral hypothalamus of coronal brain slices, we found reducing pH either inhibits or stimulates putative orexin neurons similar to other chemosensitive neurons; however, a significantly greater percentage of putative orexin neurons from Kcna1-/-mice were stimulated and the magnitude of stimulation was increased resulting in augmentation of the calculated chemosensitivity index relative to WT. Collectively, our data suggest that increased chemosensitive activity of orexin neurons may be pathologic in the Kcna1-/- mouse model of SUDEP, and contribute to elevated ventilatory responses. Our preclinical data suggest that those at high risk for SUDEP may be more sensitive to HH challenges, whether induced by seizures or other means; and the depth and length of the HH exposure could dictate the probability of survival.


Asunto(s)
Modelos Animales de Enfermedad , Hipercapnia , Hipoxia , Ratones Noqueados , Neuronas , Orexinas , Muerte Súbita e Inesperada en la Epilepsia , Animales , Hipercapnia/fisiopatología , Hipercapnia/metabolismo , Hipoxia/metabolismo , Hipoxia/fisiopatología , Orexinas/metabolismo , Ratones , Neuronas/metabolismo , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv.1.1/metabolismo , Masculino , Ratones Endogámicos C57BL
3.
Neurobiol Dis ; 196: 106513, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38663634

RESUMEN

In animal models of LGI1-dependent autosomal dominant lateral temporal lobe epilepsy, Kv1 channels are downregulated, suggesting their crucial involvement in epileptogenesis. The molecular basis of Kv1 channel-downregulation in LGI1 knock-out mice has not been elucidated and how the absence of this extracellular protein induces an important modification in the expression of Kv1 remains unknown. In this study we analyse by immunofluorescence the modifications in neuronal Kv1.1 and Kv1.2 distribution throughout the hippocampal formation of LGI1 knock-out mice. We show that Kv1 downregulation is not restricted to the axonal compartment, but also takes place in the somatodendritic region and is accompanied by a drastic decrease in Kv2 expression levels. Moreover, we find that the downregulation of these Kv channels is associated with a marked increase in bursting patterns. Finally, mass spectrometry uncovered key modifications in the Kv1 interactome that highlight the epileptogenic implication of Kv1 downregulation in LGI1 knock-out animals.


Asunto(s)
Regulación hacia Abajo , Hipocampo , Péptidos y Proteínas de Señalización Intracelular , Ratones Noqueados , Animales , Hipocampo/metabolismo , Ratones , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Canal de Potasio Kv.1.1/metabolismo , Canal de Potasio Kv.1.1/genética , Proteínas/metabolismo , Proteínas/genética , Ratones Endogámicos C57BL , Canal de Potasio Kv.1.2/metabolismo , Canal de Potasio Kv.1.2/genética , Neuronas/metabolismo
4.
Exp Brain Res ; 242(2): 477-490, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38184806

RESUMEN

Several volatile anesthetics have presented neuroprotective functions in ischemic injury. This study investigates the effect of desflurane (Des) on neurons following oxygen-glucose deprivation (OGD) challenge and explores the underpinning mechanism. Mouse neurons HT22 were subjected to OGD, which significantly reduced cell viability, increased lactate dehydrogenase release, and promoted cell apoptosis. In addition, the OGD condition increased oxidative stress in HT22 cells, as manifested by increased ROS and MDA contents, decreased SOD activity and GSH/GSSG ratio, and reduced nuclear protein level of Nrf2. Notably, the oxidative stress and neuronal apoptosis were substantially blocked by Des treatment. Bioinformatics suggested potassium voltage-gated channel subfamily A member 1 (Kcna1) as a target of Des. Indeed, the Kcna1 expression in HT22 cells was decreased by OGD but restored by Des treatment. Artificial knockdown of Kcna1 negated the neuroprotective effects of Des. By upregulating Kcna1, Des activated the Kv1.1 channel, therefore enhancing K+ currents and inducing neuronal repolarization. Pharmacological inhibition of the Kv1.1 channel reversed the protective effects of Des against OGD-induced injury. Collectively, this study demonstrates that Des improves electrical activity of neurons and alleviates OGD-induced neuronal injury by activating the Kcna1-dependent Kv1.1 channel.


Asunto(s)
Oxígeno , Daño por Reperfusión , Ratones , Animales , Glucosa/metabolismo , Desflurano/farmacología , Transducción de Señal , Estrés Oxidativo , Neuronas/metabolismo , Apoptosis , Canal de Potasio Kv.1.1/metabolismo
5.
J Biol Chem ; 298(11): 102467, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36087839

RESUMEN

Among voltage-gated potassium channel (KV) isoforms, KV1.6 is one of the most widespread in the nervous system. However, there are little data concerning its physiological significance, in part due to the scarcity of specific ligands. The known high-affinity ligands of KV1.6 lack selectivity, and conversely, its selective ligands show low affinity. Here, we present a designer peptide with both high affinity and selectivity to KV1.6. Previously, we have demonstrated that KV isoform-selective peptides can be constructed based on the simplistic α-hairpinin scaffold, and we obtained a number of artificial Tk-hefu peptides showing selective blockage of KV1.3 in the submicromolar range. We have now proposed amino acid substitutions to enhance their activity. As a result, we have been able to produce Tk-hefu-11 that shows an EC50 of ≈70 nM against KV1.3. Quite surprisingly, Tk-hefu-11 turns out to block KV1.6 with even higher potency, presenting an EC50 of ≈10 nM. Furthermore, we have solved the peptide structure and used molecular dynamics to investigate the determinants of selective interactions between artificial α-hairpinins and KV channels to explain the dramatic increase in KV1.6 affinity. Since KV1.3 is not highly expressed in the nervous system, we hope that Tk-hefu-11 will be useful in studies of KV1.6 and its functions.


Asunto(s)
Canales de Potasio con Entrada de Voltaje , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Secuencia de Aminoácidos , Bloqueadores de los Canales de Potasio/química , Péptidos/química , Ligandos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo , Canal de Potasio Kv.1.1/metabolismo , Canal de Potasio Kv.1.2/metabolismo , Canal de Potasio Kv1.5/metabolismo
6.
Mol Cell Neurosci ; 113: 103615, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33901631

RESUMEN

Cardiorespiratory collapse following a seizure is a suspected cause of sudden unexpected death in epilepsy (SUDEP), the leading cause of epilepsy-related mortality. In the commonly used Kcna1 gene knockout (Kcna1-/-) mouse model of SUDEP, cardiorespiratory profiling reveals an array of aberrant breathing patterns that could contribute to risk of seizure-related mortality. However, the brain structures mediating these respiratory abnormalities remain unknown. We hypothesize that Kv1.1 deficiency in respiratory control centers of the brain contribute to respiratory dysfunction in Kcna1-/- mice leading to increased SUDEP risk. Thus, in this study, we first used immunohistochemistry to map expression of Kv1.1 protein in cardiorespiratory brain regions of wild-type Kcna1+/+ (WT) mice. Next, GFAP and Iba1 immunostaining was used to test for the presence of astrogliosis and microgliosis, respectively, in the cardiorespiratory centers of Kcna1-/- mice, which could be indicative of seizure-related brain injury that could impair breathing. In WT mice, we detected Kv1.1 protein in all cardiorespiratory centers examined, including the basolateral amygdala, dorsal respiratory group, dorsal motor nucleus of vagus, nucleus ambiguus, ventral respiratory column, and pontine respiratory group, as well as chemosensory centers including the retrotrapezoid and median raphae nuclei. Extensive gliosis was observed in the same areas in Kcna1-/- mice suggesting that seizure-associated brain injury could contribute to respiratory abnormalities.


Asunto(s)
Encéfalo/metabolismo , Gliosis/genética , Canal de Potasio Kv.1.1/genética , Respiración , Muerte Súbita e Inesperada en la Epilepsia/etiología , Animales , Encéfalo/patología , Encéfalo/fisiopatología , Femenino , Gliosis/patología , Canal de Potasio Kv.1.1/deficiencia , Canal de Potasio Kv.1.1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Nervio Vago/metabolismo , Nervio Vago/fisiopatología
7.
RNA Biol ; 18(7): 999-1013, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33393416

RESUMEN

Adenosine-to-inosine (A-to-I) editing is one of the most prevalent post-transcriptional RNA modifications in metazoan. This reaction is catalysed by enzymes called adenosine deaminases acting on RNA (ADARs). RNA editing is involved in the regulation of protein function and gene expression. The numerous A-to-I editing sites have been identified in both coding and non-coding RNA transcripts. These editing sites are also found in various genes expressed in the central nervous system (CNS) and play an important role in neurological development and brain function. Aberrant regulation of RNA editing has been associated with the pathogenesis of neurological and psychiatric disorders, suggesting the physiological significance of RNA editing in the CNS. In this review, we discuss the current knowledge of editing on neurological disease and development.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Astrocitoma/genética , Trastorno Bipolar/genética , Neoplasias del Sistema Nervioso Central/genética , Epilepsia/genética , Edición de ARN , Esquizofrenia/genética , Espasmos Infantiles/genética , Adenosina/genética , Adenosina/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Astrocitoma/metabolismo , Astrocitoma/patología , Trastorno Bipolar/metabolismo , Trastorno Bipolar/patología , Neoplasias del Sistema Nervioso Central/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Epilepsia/metabolismo , Epilepsia/patología , Humanos , Inosina/genética , Inosina/metabolismo , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv.1.1/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Receptor de Serotonina 5-HT2C/genética , Receptor de Serotonina 5-HT2C/metabolismo , Receptores AMPA/genética , Receptores AMPA/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/patología , Espasmos Infantiles/metabolismo , Espasmos Infantiles/patología
8.
BMC Biol ; 18(1): 143, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33059680

RESUMEN

BACKGROUND: ADP-ribosylation is a ubiquitous post-translational modification that involves both mono- and poly-ADP-ribosylation. ARTD10, also known as PARP10, mediates mono-ADP-ribosylation (MARylation) of substrate proteins. A previous screen identified protein kinase C delta (PKCδ) as a potential ARTD10 substrate, among several other kinases. The voltage-gated K+ channel Kv1.1 constitutes one of the dominant Kv channels in neurons of the central nervous system and the inactivation properties of Kv1.1 are modulated by PKC. In this study, we addressed the role of ARTD10-PKCδ as a regulator of Kv1.1. RESULTS: We found that ARTD10 inhibited PKCδ, which increased Kv1.1 current amplitude and the proportion of the inactivating current component in HeLa cells, indicating that ARTD10 regulates Kv1.1 in living cells. An inhibitor of ARTD10, OUL35, significantly decreased peak amplitude together with the proportion of the inactivating current component of Kv1.1-containing channels in primary hippocampal neurons, demonstrating that the ARTD10-PKCδ signaling cascade regulates native Kv1.1. Moreover, we show that the pharmacological blockade of ARTD10 increases excitability of hippocampal neurons. CONCLUSIONS: Our results, for the first time, suggest that MARylation by ARTD10 controls neuronal excitability.


Asunto(s)
Canal de Potasio Kv.1.1/genética , Poli(ADP-Ribosa) Polimerasas/genética , Proteína Quinasa C-delta/genética , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/genética , Transducción de Señal , Animales , Células HEK293 , Células HeLa , Humanos , Canal de Potasio Kv.1.1/metabolismo , Ratones , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteína Quinasa C-delta/metabolismo , Proteínas Proto-Oncogénicas/metabolismo
9.
J Neurosci ; 39(16): 3159-3169, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30755487

RESUMEN

Refractory focal epilepsy is a devastating disease for which there is frequently no effective treatment. Gene therapy represents a promising alternative, but treating epilepsy in this way involves irreversible changes to brain tissue, so vector design must be carefully optimized to guarantee safety without compromising efficacy. We set out to develop an epilepsy gene therapy vector optimized for clinical translation. The gene encoding the voltage-gated potassium channel Kv1.1, KCNA1, was codon optimized for human expression and mutated to accelerate the recovery of the channels from inactivation. For improved safety, this engineered potassium channel (EKC) gene was packaged into a nonintegrating lentiviral vector under the control of a cell type-specific CAMK2A promoter. In a blinded, randomized, placebo-controlled preclinical trial, the EKC lentivector robustly reduced seizure frequency in a male rat model of focal neocortical epilepsy characterized by discrete spontaneous seizures. When packaged into an adeno-associated viral vector (AAV2/9), the EKC gene was also effective at suppressing seizures in a male rat model of temporal lobe epilepsy. This demonstration of efficacy in a clinically relevant setting, combined with the improved safety conferred by cell type-specific expression and integration-deficient delivery, identify EKC gene therapy as being ready for clinical translation in the treatment of refractory focal epilepsy.SIGNIFICANCE STATEMENT Pharmacoresistant epilepsy affects up to 0.3% of the population. Although epilepsy surgery can be effective, it is limited by risks to normal brain function. We have developed a gene therapy that builds on a mechanistic understanding of altered neuronal and circuit excitability in cortical epilepsy. The potassium channel gene KCNA1 was mutated to bypass post-transcriptional editing and was packaged in a nonintegrating lentivector to reduce the risk of insertional mutagenesis. A randomized, blinded preclinical study demonstrated therapeutic effectiveness in a rodent model of focal neocortical epilepsy. Adeno-associated viral delivery of the channel to both hippocampi was also effective in a model of temporal lobe epilepsy. These results support clinical translation to address a major unmet need.


Asunto(s)
Encéfalo/metabolismo , Epilepsia/terapia , Terapia Genética , Canal de Potasio Kv.1.1/genética , Convulsiones/terapia , Animales , Modelos Animales de Enfermedad , Epilepsia/genética , Vectores Genéticos , Canal de Potasio Kv.1.1/metabolismo , Masculino , Ratas , Convulsiones/genética
10.
Metabolomics ; 16(10): 106, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-33021695

RESUMEN

INTRODUCTION: Approximately 1% of the world's population is impacted by epilepsy, a chronic neurological disorder characterized by seizures. One-third of epileptic patients are resistant to AEDs, or have medically refractory epilepsy (MRE). One non-invasive treatment that exists for MRE includes the ketogenic diet, a high-fat, low-carbohydrate diet. Despite the KD's success in seizure attenuation, it has a few risks and its mechanisms remain poorly understood. The KD has been shown to improve metabolism and mitochondrial function in epileptic phenotypes. Potassium channels have implications in epileptic conditions as they have dual roles as metabolic sensors and control neuronal excitation. OBJECTIVES: The goal of this study was to explore changes in the lipidome in hippocampal and cortical tissue from Kv1.1-KO model of epilepsy. METHODS: FT-ICR/MS analysis was utilized to examine nonpolar metabolome of cortical and hippocampal tissue isolated from a Kv1.1 channel knockout mouse model of epilepsy (n = 5) and wild-type mice (n = 5). RESULTS: Distinct metabolic profiles were observed, significant (p < 0.05) features in hippocampus often being upregulated (FC ≥ 2) and the cortex being downregulated (FC ≤ 0.5). Pathway enrichment analysis shows lipid biosynthesis was affected. Partition ratio analysis revealed that the ratio of most metabolites tended to be increased in Kv1.1-/-. Metabolites in hippocampal tissue were commonly upregulated, suggesting seizure initiation in the hippocampus. Aberrant mitochondrial function is implicated by the upregulation of cardiolipin, a common component in the mitochondrial membrane. CONCLUSION: Generally, our study finds that the lipidome is changed in the hippocampus and cortex in response to Kv1.1-KO indicating changes in membrane structural integrity and synaptic transmission.


Asunto(s)
Epilepsia/metabolismo , Metabolismo de los Lípidos/fisiología , Animales , Dieta Cetogénica/métodos , Modelos Animales de Enfermedad , Epilepsia/dietoterapia , Hipocampo/metabolismo , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv.1.1/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados
11.
Epilepsia ; 61(12): 2836-2846, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33104247

RESUMEN

OBJECTIVE: Leucine-rich glioma-inactivated 1 (LGI1) is a secreted transsynaptic protein that interacts presynaptically with Kv1.1 potassium channels and a disintegrin and metalloprotease (ADAM) protein 23, and postsynaptically influences α-amino-3-hydroxy-5-methylisoxazole-4-propionate receptors through a direct link with the ADAM22 cell adhesion protein. Haploinsufficiency of LGI1 or autoantibodies directed against LGI1 are associated with human epilepsy, generating the hypothesis that a subacute reduction of LGI1 is sufficient to increase network excitability. METHODS: We tested this hypothesis in ex vivo hippocampal slices and in neuronal cultures, by subacutely reducing LGI1 expression with shRNA. RESULTS: Injection of shRNA-LGI1 in the hippocampus increased dentate granule cell excitability and low-frequency facilitation of mossy fibers to CA3 pyramidal cell neurotransmission. Application of the Kv1 family blocker, α-dendrotoxin, occluded this effect, implicating the involvement of Kv1.1. This subacute reduction of LGI1 was also sufficient to increase neuronal network activity in neuronal primary culture. SIGNIFICANCE: These results indicate that a subacute reduction in LGI1 potentiates neuronal excitability and short-term synaptic plasticity, and increases neuronal network excitability, opening new avenues for the treatment of limbic encephalitis and temporal lobe epilepsies.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/fisiología , Neuronas/fisiología , Convulsiones/etiología , Animales , Regulación hacia Abajo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Canal de Potasio Kv.1.1/metabolismo , Canal de Potasio Kv.1.1/fisiología , Ratones , Ratones Noqueados , Comunicación Paracrina , ARN Interferente Pequeño , Convulsiones/fisiopatología , Sinapsis/metabolismo , Sinapsis/fisiología
12.
Bioorg Chem ; 100: 103918, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32428746

RESUMEN

Members of the voltage-gated K+ channel subfamily (Kv1), involved in regulating transmission between neurons or to muscles, are associated with human diseases and, thus, putative targets for neurotherapeutics. This applies especially to those containing Kv1.1 α subunits which become prevalent in murine demyelinated axons and appear abnormally at inter-nodes, underlying the perturbed propagation of nerve signals. To overcome this dysfunction, akin to the consequential debilitation in multiple sclerosis (MS), small inhibitors were sought that are selective for the culpable hyper-polarising K+ currents. Herein, we report a new semi-podand - compound 3 - that was designed based on the modelling of its interactions with the extracellular pore region in a deduced Kv1.1 channel structure. After synthesis, purification, and structural characterisation, compound 3 was found to potently (IC50 = 8 µM) and selectively block Kv1.1 and 1.6 channels. The tested compound showed no apparent effect on native Nav and Cav channels expressed in F-11 cells. Compound 3 also extensively and selectively inhibited MS-related Kv1.1 homomer but not the brain native Kv1.1- or 1.6-containing channels. These collective findings highlight the therapeutic potential of compound 3 to block currents mediated by Kv1.1 channels enriched in demyelinated central neurons.


Asunto(s)
Canal de Potasio Kv.1.1/antagonistas & inhibidores , Neuronas/efectos de los fármacos , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/farmacología , Animales , Línea Celular , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Diseño de Fármacos , Células HEK293 , Humanos , Canal de Potasio Kv.1.1/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Neuronas/metabolismo , Bloqueadores de los Canales de Potasio/síntesis química , Ratas
13.
Int J Mol Sci ; 21(20)2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33066705

RESUMEN

(1) Background: Episodic ataxia type 1 is caused by mutations in the KCNA1 gene encoding for the voltage-gated potassium channel Kv1.1. There have been many mutations in Kv1.1 linked to episodic ataxia reported and typically investigated by themselves or in small groups. The aim of this article is to determine whether we can define a functional parameter common to all Kv1.1 mutants that have been linked to episodic ataxia. (2) Methods: We introduced the disease mutations linked to episodic ataxia in the drosophila analog of Kv1.1, the Shaker Kv channel, and expressed the channels in Xenopus oocytes. Using the cut-open oocyte technique, we characterized the gating and ionic currents. (3) Results: We found that the episodic ataxia mutations variably altered the different gating mechanisms described for Kv channels. The common characteristic was a conductance voltage relationship and inactivation shifted to less polarized potentials. (4) Conclusions: We suggest that a combination of a prolonged action potential and slowed and incomplete inactivation leads to development of ataxia when Kv channels cannot follow or adapt to high firing rates.


Asunto(s)
Ataxia/genética , Activación del Canal Iónico , Canal de Potasio Kv.1.1/genética , Mutación , Miocimia/genética , Animales , Humanos , Canal de Potasio Kv.1.1/química , Canal de Potasio Kv.1.1/metabolismo , Xenopus
14.
Int J Mol Sci ; 21(8)2020 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-32331416

RESUMEN

Kv1.1 belongs to the Shaker subfamily of voltage-gated potassium channels and acts as a critical regulator of neuronal excitability in the central and peripheral nervous systems. KCNA1 is the only gene that has been associated with episodic ataxia type 1 (EA1), an autosomal dominant disorder characterized by ataxia and myokymia and for which different and variable phenotypes have now been reported. The iterative characterization of channel defects at the molecular, network, and organismal levels contributed to elucidating the functional consequences of KCNA1 mutations and to demonstrate that ataxic attacks and neuromyotonia result from cerebellum and motor nerve alterations. Dysfunctions of the Kv1.1 channel have been also associated with epilepsy and kcna1 knock-out mouse is considered a model of sudden unexpected death in epilepsy. The tissue-specific association of Kv1.1 with other Kv1 members, auxiliary and interacting subunits amplifies Kv1.1 physiological roles and expands the pathogenesis of Kv1.1-associated diseases. In line with the current knowledge, Kv1.1 has been proposed as a novel and promising target for the treatment of brain disorders characterized by hyperexcitability, in the attempt to overcome limited response and side effects of available therapies. This review recounts past and current studies clarifying the roles of Kv1.1 in and beyond the nervous system and its contribution to EA1 and seizure susceptibility as well as its wide pharmacological potential.


Asunto(s)
Canalopatías/etiología , Canalopatías/terapia , Predisposición Genética a la Enfermedad , Canal de Potasio Kv.1.1/genética , Mutación , Alelos , Animales , Canalopatías/diagnóstico , Canalopatías/metabolismo , Manejo de la Enfermedad , Regulación de la Expresión Génica , Estudios de Asociación Genética , Genotipo , Humanos , Activación del Canal Iónico , Canal de Potasio Kv.1.1/química , Canal de Potasio Kv.1.1/metabolismo , Terapia Molecular Dirigida , Fenotipo , Relación Estructura-Actividad
15.
Int J Mol Sci ; 21(18)2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32899446

RESUMEN

The term Episodic Ataxias (EA) was originally used for a few autosomal dominant diseases, characterized by attacks of cerebellar dysfunction of variable duration and frequency, often accompanied by other ictal and interictal signs. The original group subsequently grew to include other very rare EAs, frequently reported in single families, for some of which no responsible gene was found. The clinical spectrum of these diseases has been enormously amplified over time. In addition, episodes of ataxia have been described as phenotypic variants in the context of several different disorders. The whole group is somewhat confused, since a strong evidence linking the mutation to a given phenotype has not always been established. In this review we will collect and examine all instances of ataxia episodes reported so far, emphasizing those for which the pathophysiology and the clinical spectrum is best defined.


Asunto(s)
Ataxia/genética , Ataxia/metabolismo , Ataxia/fisiopatología , Canales de Calcio/genética , Ataxia Cerebelosa/genética , Transportador 1 de Aminoácidos Excitadores/genética , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv.1.1/metabolismo , Canal de Sodio Activado por Voltaje NAV1.2/genética
16.
Neurobiol Dis ; 127: 502-511, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30974168

RESUMEN

Sudden unexpected death in epilepsy (SUDEP) is the leading cause of epilepsy-related mortality, but the relative importance of underlying cardiac and respiratory mechanisms remains unclear. To illuminate the interactions between seizures, respiration, cardiac function, and sleep that contribute to SUDEP risk, here we developed a mouse epilepsy monitoring unit (EMU) to simultaneously record video, electroencephalography (EEG), electromyography (EMG), plethysmography, and electrocardiography (ECG) in a commonly used genetic model of SUDEP, the Kcna1 knockout (Kcna1-/-) mouse. During interictal periods, Kcna1-/- mice exhibited an abnormal absence of post-sigh apneas and a 3-fold increase in respiratory variability. During spontaneous convulsive seizures, Kcna1-/- mice displayed an array of aberrant breathing patterns that always preceded cardiac abnormalities. These findings support respiratory dysfunction as a primary risk factor for susceptibility to deleterious cardiorespiratory sequelae in epilepsy and reveal a new role for Kcna1-encoded Kv1.1 channels in the regulation of basal respiratory physiology.


Asunto(s)
Epilepsia/fisiopatología , Canal de Potasio Kv.1.1/metabolismo , Sistema Respiratorio/fisiopatología , Convulsiones/complicaciones , Muerte Súbita e Inesperada en la Epilepsia/etiología , Animales , Modelos Animales de Enfermedad , Electrocardiografía , Electroencefalografía , Epilepsia/genética , Epilepsia/metabolismo , Canal de Potasio Kv.1.1/genética , Ratones , Ratones Noqueados , Respiración , Sistema Respiratorio/metabolismo , Factores de Riesgo , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/fisiopatología
17.
Hum Mol Genet ; 26(11): 2091-2103, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334922

RESUMEN

People with epilepsy have greatly increased probability of premature mortality due to sudden unexpected death in epilepsy (SUDEP). Identifying which patients are most at risk of SUDEP is hindered by a complex genetic etiology, incomplete understanding of the underlying pathophysiology and lack of prognostic biomarkers. Here we evaluated heterozygous Scn2a gene deletion (Scn2a+/-) as a protective genetic modifier in the Kcna1 knockout mouse (Kcna1-/-) model of SUDEP, while searching for biomarkers of SUDEP risk embedded in electroencephalography (EEG) and electrocardiography (ECG) recordings. The human epilepsy gene Kcna1 encodes voltage-gated Kv1.1 potassium channels that act to dampen neuronal excitability whereas Scn2a encodes voltage-gated Nav1.2 sodium channels important for action potential initiation and conduction. SUDEP-prone Kcna1-/- mice with partial genetic ablation of Nav1.2 channels (i.e. Scn2a+/-; Kcna1-/-) exhibited a two-fold increase in survival. Classical analysis of EEG and ECG recordings separately showed significantly decreased seizure durations in Scn2a+/-; Kcna1-/- mice compared with Kcna1-/- mice, without substantial modification of cardiac abnormalities. Novel analysis of the EEG and ECG together revealed a significant reduction in EEG-ECG association in Kcna1-/- mice compared with wild types, which was partially restored in Scn2a+/-; Kcna1-/- mice. The degree of EEG-ECG association was also proportional to the survival rate of mice across genotypes. These results show that Scn2a gene deletion acts as protective genetic modifier of SUDEP and suggest measures of brain-heart association as potential indices of SUDEP susceptibility.


Asunto(s)
Epilepsia/genética , Canal de Sodio Activado por Voltaje NAV1.2/genética , Canal de Sodio Activado por Voltaje NAV1.2/metabolismo , Animales , Biomarcadores , Encéfalo/fisiopatología , Muerte Súbita , Modelos Animales de Enfermedad , Electrocardiografía , Electroencefalografía , Epilepsia/complicaciones , Genotipo , Corazón/fisiopatología , Frecuencia Cardíaca , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv.1.1/metabolismo , Ratones , Ratones Noqueados , Convulsiones/genética
18.
Brain ; 141(3): 744-761, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29373639

RESUMEN

Despite the development of newer anti-seizure medications over the past 50 years, 30-40% of patients with epilepsy remain refractory to treatment. One explanation for this lack of progress is that the current screening process is largely biased towards transmembrane channels and receptors, and ignores intracellular proteins and enzymes that might serve as efficacious molecular targets. Here, we report the development of a novel drug screening platform that harnesses the power of zebrafish genetics and combines it with in vivo bioenergetics screening assays to uncover therapeutic agents that improve mitochondrial health in diseased animals. By screening commercially available chemical libraries of approved drugs, for which the molecular targets and pathways are well characterized, we were able to reverse-identify the proteins targeted by efficacious compounds and confirm the physiological roles that they play by utilizing other pharmacological ligands. Indeed, using an 870-compound screen in kcna1-morpholino epileptic zebrafish larvae, we uncovered vorinostat (Zolinza™; suberanilohydroxamic acid, SAHA) as a potent anti-seizure agent. We further demonstrated that vorinostat decreased average daily seizures by ∼60% in epileptic Kcna1-null mice using video-EEG recordings. Given that vorinostat is a broad histone deacetylase (HDAC) inhibitor, we then delineated a specific subset of HDACs, namely HDACs 1 and 3, as potential drug targets for future screening. In summary, we have developed a novel phenotypic, metabolism-based experimental therapeutics platform that can be used to identify new molecular targets for future drug discovery in epilepsy.


Asunto(s)
Histona Desacetilasas/metabolismo , Convulsiones/metabolismo , Convulsiones/terapia , Animales , Animales Modificados Genéticamente , Anticonvulsivantes/uso terapéutico , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Electrochoque/efectos adversos , Embrión no Mamífero , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Inhibidores de Histona Desacetilasas/uso terapéutico , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv.1.1/metabolismo , Ratones , Morfolinos , Pentilenotetrazol/toxicidad , Desempeño Psicomotor/fisiología , Convulsiones/etiología , Convulsiones/genética , Vorinostat/uso terapéutico , Pez Cebra
19.
Brain ; 141(11): 3144-3159, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30346486

RESUMEN

Leucine-rich glioma-inactivated 1 (LGI1) is a secreted neuronal protein that forms a trans-synaptic complex that includes the presynaptic disintegrin and metalloproteinase domain-containing protein 23 (ADAM23), which interacts with voltage-gated potassium channels Kv1.1, and the postsynaptic ADAM22, which interacts with AMPA receptors. Human autoantibodies against LGI1 associate with a form of autoimmune limbic encephalitis characterized by severe but treatable memory impairment and frequent faciobrachial dystonic seizures. Although there is evidence that this disease is immune-mediated, the underlying LGI1 antibody-mediated mechanisms are unknown. Here, we used patient-derived immunoglobulin G (IgG) antibodies to determine the main epitope regions of LGI1 and whether the antibodies disrupt the interaction of LGI1 with ADAM23 and ADAM22. In addition, we assessed the effects of patient-derived antibodies on Kv1.1, AMPA receptors, and memory in a mouse model based on cerebroventricular transfer of patient-derived IgG. We found that IgG from all patients (n = 25), but not from healthy participants (n = 20), prevented the binding of LGI1 to ADAM23 and ADAM22. Using full-length LGI1, LGI3, and LGI1 constructs containing the LRR1 domain (EPTP1-deleted) or EPTP1 domain (LRR3-EPTP1), IgG from all patients reacted with epitope regions contained in the LRR1 and EPTP1 domains. Confocal analysis of hippocampal slices of mice infused with pooled IgG from eight patients, but not pooled IgG from controls, showed a decrease of total and synaptic levels of Kv1.1 and AMPA receptors. The effects on Kv1.1 preceded those involving the AMPA receptors. In acute slice preparations of hippocampus, patch-clamp analysis from dentate gyrus granule cells and CA1 pyramidal neurons showed neuronal hyperexcitability with increased glutamatergic transmission, higher presynaptic release probability, and reduced synaptic failure rate upon minimal stimulation, all likely caused by the decreased expression of Kv1.1. Analysis of synaptic plasticity by recording field potentials in the CA1 region of the hippocampus showed a severe impairment of long-term potentiation. This defect in synaptic plasticity was independent from Kv1 blockade and was possibly mediated by ineffective recruitment of postsynaptic AMPA receptors. In parallel with these findings, mice infused with patient-derived IgG showed severe memory deficits in the novel object recognition test that progressively improved after stopping the infusion of patient-derived IgG. Different from genetic models of LGI1 deficiency, we did not observe aberrant dendritic sprouting or defective synaptic pruning as potential cause of the symptoms. Overall, these findings demonstrate that patient-derived IgG disrupt presynaptic and postsynaptic LGI1 signalling, causing neuronal hyperexcitability, decreased plasticity, and reversible memory deficits.


Asunto(s)
Inmunoglobulina G/farmacología , Canal de Potasio Kv.1.1/metabolismo , Memoria/fisiología , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Proteínas/inmunología , Receptores AMPA/metabolismo , Proteínas ADAM/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Encéfalo/citología , Encéfalo/metabolismo , Homólogo 4 de la Proteína Discs Large/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Canal de Potasio Kv.1.1/ultraestructura , Encefalitis Límbica/inmunología , Masculino , Memoria/efectos de los fármacos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Unión Proteica/efectos de los fármacos , Dominios Proteicos/efectos de los fármacos , Proteínas/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Sinapsis/ultraestructura
20.
Epilepsia ; 59(1): e1-e4, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29171006

RESUMEN

Metabolic alteration, either through the ketogenic diet (KD) or by genetic alteration of the BAD protein, can produce seizure protection in acute chemoconvulsant models of epilepsy. To assess the seizure-protective role of knocking out (KO) the Bad gene in a chronic epilepsy model, we used the Kcna1-/- model of epilepsy, which displays progressively increased seizure severity and recapitulates the early death seen in sudden unexplained death in epilepsy (SUDEP). Beginning on postnatal day 24 (P24), we continuously video monitored Kcna1-/- and Kcna1-/- Bad-/- double knockout mice to assess survival and seizure severity. We found that Kcna1-/- Bad-/- mice outlived Kcna1-/- mice by approximately 2 weeks. Kcna1-/- Bad-/- mice also spent significantly less time in seizure than Kcna1-/- mice on P24 and the day of death, showing that BadKO provides seizure resistance in a genetic model of chronic epilepsy.


Asunto(s)
Síndrome de Brugada/etiología , Síndrome de Brugada/genética , Epilepsia , Canal de Potasio Kv.1.1/genética , Proteína Letal Asociada a bcl/deficiencia , Factores de Edad , Animales , Síndrome de Brugada/metabolismo , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia/complicaciones , Epilepsia/genética , Epilepsia/prevención & control , Femenino , Canal de Potasio Kv.1.1/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Letal Asociada a bcl/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA