Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Neurobiol Dis ; 196: 106513, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38663634

RESUMO

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.


Assuntos
Regulação para Baixo , Hipocampo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos Knockout , Animais , Hipocampo/metabolismo , Camundongos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.1/genética , Proteínas/metabolismo , Proteínas/genética , Camundongos Endogâmicos C57BL , Canal de Potássio Kv1.2/metabolismo , Canal de Potássio Kv1.2/genética , Neurônios/metabolismo
2.
Exp Brain Res ; 242(2): 477-490, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38184806

RESUMO

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.


Assuntos
Oxigênio , Traumatismo por Reperfusão , Camundongos , Animais , Glucose/metabolismo , Desflurano/farmacologia , Transdução de Sinais , Estresse Oxidativo , Neurônios/metabolismo , Apoptose , Canal de Potássio Kv1.1/metabolismo
3.
Cells ; 12(10)2023 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-37408217

RESUMO

Dominantly inherited missense mutations of the KCNA1 gene, which encodes the KV1.1 potassium channel subunit, cause Episodic Ataxia type 1 (EA1). Although the cerebellar incoordination is thought to arise from abnormal Purkinje cell output, the underlying functional deficit remains unclear. Here we examine synaptic and non-synaptic inhibition of Purkinje cells by cerebellar basket cells in an adult mouse model of EA1. The synaptic function of basket cell terminals was unaffected, despite their intense enrichment for KV1.1-containing channels. In turn, the phase response curve quantifying the influence of basket cell input on Purkine cell output was maintained. However, ultra-fast non-synaptic ephaptic coupling, which occurs in the cerebellar 'pinceau' formation surrounding the axon initial segment of Purkinje cells, was profoundly reduced in EA1 mice in comparison with their wild type littermates. The altered temporal profile of basket cell inhibition of Purkinje cells underlines the importance of Kv1.1 channels for this form of signalling, and may contribute to the clinical phenotype of EA1.


Assuntos
Ataxia , Canal de Potássio Kv1.1 , Mioquimia , Inibição Neural , Células de Purkinje , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Animais , Camundongos , Modelos Animais de Doenças , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Sinapses/fisiologia , Comunicação Celular , Transmissão Sináptica , Ataxia/genética , Ataxia/patologia , Ataxia/fisiopatologia , Mioquimia/genética , Mioquimia/patologia , Mioquimia/fisiopatologia , Potenciais Evocados , Camundongos Endogâmicos C57BL , Masculino , Feminino
4.
Nat Commun ; 14(1): 3281, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37280215

RESUMO

There are currently no drugs known to rescue the function of Kv1.1 voltage-gated potassium channels carrying loss-of-function sequence variants underlying the inherited movement disorder, Episodic Ataxia 1 (EA1). The Kwakwaka'wakw First Nations of the Pacific Northwest Coast used Fucus gardneri (bladderwrack kelp), Physocarpus capitatus (Pacific ninebark) and Urtica dioica (common nettle) to treat locomotor ataxia. Here, we show that extracts of these plants enhance wild-type Kv1.1 current, especially at subthreshold potentials. Screening of their constituents revealed that gallic acid and tannic acid similarly augment wild-type Kv1.1 current, with submicromolar potency. Crucially, the extracts and their constituents also enhance activity of Kv1.1 channels containing EA1-linked sequence variants. Molecular dynamics simulations reveal that gallic acid augments Kv1.1 activity via a small-molecule binding site in the extracellular S1-S2 linker. Thus, traditional Native American ataxia treatments utilize a molecular mechanistic foundation that can inform small-molecule approaches to therapeutically correcting EA1 and potentially other Kv1.1-linked channelopathies.


Assuntos
Ataxia , Canal de Potássio Kv1.1 , Humanos , Ataxia/tratamento farmacológico , Ataxia/genética , Ativação do Canal Iônico , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Mutação , Canadenses Indígenas , Medicina Tradicional
5.
ACS Chem Neurosci ; 13(23): 3342-3351, 2022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36417797

RESUMO

Demyelination, the loss of the insulating sheath of neurons, causes failed or slowed neuronal conduction and contributes to the neurological symptoms in multiple sclerosis, traumatic brain and spinal cord injuries, stroke, and dementia. In demyelinated neurons, the axonal potassium channels Kv1.1 and Kv1.2, generally under the myelin sheath, become exposed and upregulated. Therefore, imaging these channels using positron emission tomography can provide valuable information for disease diagnosis and monitoring. Here, we describe a novel tracer for Kv1 channels, [11C]3-methyl-4-aminopyridine ([11C]3Me4AP). [11C]3Me4AP was efficiently synthesized via Pd(0)-Cu(I) comediated Stille cross-coupling of a stannyl precursor containing a free amino group. Evaluation of its imaging properties in rats and nonhuman primates showed that [11C]3Me4AP has a moderate brain permeability and slow kinetics. Additional evaluation in monkeys showed that the tracer is metabolically stable and that a one-tissue compartment model can accurately model the regional brain time-activity curves. Compared to the related tracers [18F]3-fluoro-4-aminopyridine ([18F]3F4AP) and [11C]3-methoxy-4-aminopyridine ([11C]3MeO4AP), [11C]3Me4AP shows lower initial brain uptake, which indicates reduced permeability to the blood-brain barrier and slower kinetics, suggesting higher binding affinity consistent with in vitro studies. While the slow kinetics and strong binding affinity resulted in a tracer with less favorable properties for imaging the brain than its predecessors, these properties may make 3Me4AP useful as a therapeutic.


Assuntos
4-Aminopiridina , Encéfalo , Doenças Desmielinizantes , Canal de Potássio Kv1.1 , Canal de Potássio Kv1.2 , Imagem Molecular , Traçadores Radioativos , Animais , Ratos , 4-Aminopiridina/análogos & derivados , 4-Aminopiridina/síntese química , 4-Aminopiridina/farmacocinética , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Permeabilidade , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.2/metabolismo , Doenças Desmielinizantes/diagnóstico por imagem , Imagem Molecular/métodos , Primatas , Barreira Hematoencefálica/metabolismo
6.
J Biol Chem ; 298(11): 102467, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36087839

RESUMO

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.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Sequência de Aminoácidos , Bloqueadores dos Canais de Potássio/química , Peptídeos/química , Ligantes , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.2/metabolismo , Canal de Potássio Kv1.5/metabolismo
7.
Cells ; 11(2)2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35053355

RESUMO

The mathematical modeling of ion channel kinetics is an important tool for studying the electrophysiological mechanisms of the nerves, heart, or cancer, from a single cell to an organ. Common approaches use either a Hodgkin-Huxley (HH) or a hidden Markov model (HMM) description, depending on the level of detail of the functionality and structural changes of the underlying channel gating, and taking into account the computational effort for model simulations. Here, we introduce for the first time a novel system theory-based approach for ion channel modeling based on the concept of transfer function characterization, without a priori knowledge of the biological system, using patch clamp measurements. Using the shaker-related voltage-gated potassium channel Kv1.1 (KCNA1) as an example, we compare the established approaches, HH and HMM, with the system theory-based concept in terms of model accuracy, computational effort, the degree of electrophysiological interpretability, and methodological limitations. This highly data-driven modeling concept offers a new opportunity for the phenomenological kinetic modeling of ion channels, exhibiting exceptional accuracy and computational efficiency compared to the conventional methods. The method has a high potential to further improve the quality and computational performance of complex cell and organ model simulations, and could provide a valuable new tool in the field of next-generation in silico electrophysiology.


Assuntos
Canal de Potássio Kv1.1/metabolismo , Modelos Biológicos , Animais , Simulação por Computador , Ativação do Canal Iônico , Cadeias de Markov , Subunidades Proteicas/metabolismo , Ratos
8.
Sci Rep ; 11(1): 15180, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34312446

RESUMO

Kv1.1 containing potassium channels play crucial roles towards dampening neuronal excitability. Mice lacking Kv1.1 subunits (Kcna1-/-) display recurrent spontaneous seizures and often exhibit sudden unexpected death. Seizures in Kcna1-/- mice resemble those in well-characterized models of temporal lobe epilepsy known to involve limbic brain regions and spontaneous seizures result in enhanced cFos expression and neuronal death in the amygdala. Yet, the functional alterations leading to amygdala hyperexcitability have not been identified. In this study, we used Kcna1-/- mice to examine the contributions of Kv1.1 subunits to excitability in neuronal subtypes from basolateral (BLA) and central lateral (CeL) amygdala known to exhibit distinct firing patterns. We also analyzed synaptic transmission properties in an amygdala local circuit predicted to be involved in epilepsy-related comorbidities. Our data implicate Kv1.1 subunits in controlling spontaneous excitatory synaptic activity in BLA pyramidal neurons. In the CeL, Kv1.1 loss enhances intrinsic excitability and impairs inhibitory synaptic transmission, notably resulting in dysfunction of feed-forward inhibition, a critical mechanism for controlling spike timing. Overall, we find inhibitory control of CeL interneurons is reduced in Kcna1-/- mice suggesting that basal inhibitory network functioning is less able to prevent recurrent hyperexcitation related to seizures.


Assuntos
Tonsila do Cerebelo/metabolismo , Epilepsia do Lobo Temporal/metabolismo , Canal de Potássio Kv1.1/metabolismo , Animais , Complexo Nuclear Basolateral da Amígdala/metabolismo , Núcleo Central da Amígdala/metabolismo , Modelos Animais de Doenças , Retroalimentação Fisiológica , Feminino , Canal de Potássio Kv1.1/deficiência , Canal de Potássio Kv1.1/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Inibição Neural/fisiologia , Células Piramidais/metabolismo , Convulsões/metabolismo , Transmissão Sináptica/fisiologia
9.
Elife ; 102021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-34018923

RESUMO

In the postnatal brain, neurogenesis occurs only within a few regions, such as the hippocampal sub-granular zone (SGZ). Postnatal neurogenesis is tightly regulated by factors that balance stem cell renewal with differentiation, and it gives rise to neurons that participate in learning and memory formation. The Kv1.1 channel, a voltage-gated potassium channel, was previously shown to suppress postnatal neurogenesis in the SGZ in a cell-autonomous manner. In this study, we have clarified the physiological and molecular mechanisms underlying Kv1.1-dependent postnatal neurogenesis. First, we discovered that the membrane potential of neural progenitor cells is highly dynamic during development. We further established a multinomial logistic regression model for cell-type classification based on the biophysical characteristics and corresponding cell markers. We found that the loss of Kv1.1 channel activity causes significant depolarization of type 2b neural progenitor cells. This depolarization is associated with increased tropomyosin receptor kinase B (TrkB) signaling and proliferation of neural progenitor cells; suppressing TrkB signaling reduces the extent of postnatal neurogenesis. Thus, our study defines the role of the Kv1.1 potassium channel in regulating the proliferation of postnatal neural progenitor cells in mouse hippocampus.


Assuntos
Proliferação de Células , Hipocampo/metabolismo , Canal de Potássio Kv1.1/metabolismo , Glicoproteínas de Membrana/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Animais Recém-Nascidos , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/citologia , Técnicas In Vitro , Canal de Potássio Kv1.1/genética , Glicoproteínas de Membrana/genética , Potenciais da Membrana , Camundongos Endogâmicos ICR , Camundongos Knockout , Proteínas Tirosina Quinases/genética
10.
Mol Cell Neurosci ; 113: 103615, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33901631

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Gliose/genética , Canal de Potássio Kv1.1/genética , Respiração , Morte Súbita Inesperada na Epilepsia/etiologia , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Feminino , Gliose/patologia , Canal de Potássio Kv1.1/deficiência , Canal de Potássio Kv1.1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nervo Vago/metabolismo , Nervo Vago/fisiopatologia
11.
RNA Biol ; 18(7): 999-1013, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33393416

RESUMO

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.


Assuntos
Esclerose Lateral Amiotrófica/genética , Astrocitoma/genética , Transtorno Bipolar/genética , Neoplasias do Sistema Nervoso Central/genética , Epilepsia/genética , Edição de RNA , Esquizofrenia/genética , Espasmos Infantis/genética , Adenosina/genética , Adenosina/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Astrocitoma/metabolismo , Astrocitoma/patologia , Transtorno Bipolar/metabolismo , Transtorno Bipolar/patologia , Neoplasias do Sistema Nervoso Central/metabolismo , Neoplasias do Sistema Nervoso Central/patologia , Epilepsia/metabolismo , Epilepsia/patologia , Humanos , Inosina/genética , Inosina/metabolismo , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Receptor 5-HT2C de Serotonina/genética , Receptor 5-HT2C de Serotonina/metabolismo , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/patologia , Espasmos Infantis/metabolismo , Espasmos Infantis/patologia
12.
Mol Neurobiol ; 58(5): 2046-2060, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33411244

RESUMO

The Shaker-related potassium channel Kv1.1 subunit has important implications for controlling neuronal excitabilities. A particular recoding by A-to-I RNA editing at I400 of Kv1.1 mRNA is an underestimated mechanism for fine-tuning the properties of Kv1.1-containing channels. Knowledge about functional differences between edited (I400V) and non-edited Kv1.1 isoforms is insufficient, especially in neurons. To understand their different roles, the two Kv1.1 isoforms were overexpressed in the prefrontal cortex via local adeno-associated virus-mediated gene delivery. The I400V isoform showed a higher competitiveness in membrane translocalization, but failed to reduce current-evoked discharges and showed weaker impact on spiking-frequency adaptation in the transduced neurons. The non-edited Kv1.1 overexpression led to slight elevations in both fast- and non-inactivating current components of macroscopic potassium current. By contrast, the I400V overexpression did not impact the fast-inactivating current component. Further isolation of Kv1.1-specific current by its specific blocker dendrotoxin-κ showed that both isoforms did result in significant increases in current amplitude, whereas the I400V was less efficient in contributing the fast-inactivating current component. Voltage-dependent properties of the fast-inactivating current component did not alter for both isoforms. For recovery kinetics, the I400V showed a significant acceleration of recovery from fast inactivation. The gene delivery of the I400V rather than the wild type exhibited anxiolytic activities, which was assessed by an open field test. These results suggest that the Kv1.1 RNA editing isoforms have different properties and outcomes, reflecting the functional and phenotypic significance of the Kv1.1 RNA editing in neurons.


Assuntos
Canal de Potássio Kv1.1/metabolismo , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Animais , Canal de Potássio Kv1.1/genética , Memória de Curto Prazo/fisiologia , Camundongos , Atividade Motora/fisiologia , Edição de RNA , Isoformas de RNA
13.
Physiol Rep ; 9(1): e14702, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33427415

RESUMO

Epilepsy-associated Kv1.1 voltage-gated potassium channel subunits encoded by the Kcna1 gene have traditionally been considered absent in heart, but recent studies reveal they are expressed in cardiomyocytes where they could regulate intrinsic cardiac electrophysiology. Although Kv1.1 now has a demonstrated functional role in atria, its role in the ventricles has never been investigated. In this work, electrophysiological, histological, and gene expression approaches were used to explore the consequences of Kv1.1 deficiency in the ventricles of Kcna1 knockout (KO) mice at the organ, cellular, and molecular levels to determine whether the absence of Kv1.1 leads to ventricular dysfunction that increases the risk of premature or sudden death. When subjected to intracardiac pacing, KO mice showed normal baseline susceptibility to inducible ventricular arrhythmias (VA) but resistance to VA under conditions of sympathetic challenge with isoproterenol. Echocardiography revealed cardiac contractile dysfunction manifesting as decreased ejection fraction and fractional shortening. In whole-cell patch-clamp recordings, KO ventricular cardiomyocytes exhibited action potential prolongation indicative of impaired repolarization. Imaging, histological, and transcript analyses showed no evidence of structural or channel gene expression remodeling, suggesting that the observed deficits are likely electrogenic due to Kv1.1 deficiency. Immunoblots of patient heart samples detected the presence of Kv1.1 at relatively high levels, implying that Kv1.1 contributes to human cardiac electrophysiology. Taken together, this work describes an important functional role for Kv1.1 in ventricles where its absence causes repolarization and contractility deficits but reduced susceptibility to arrhythmia under conditions of sympathetic drive.


Assuntos
Arritmias Cardíacas/fisiopatologia , Ventrículos do Coração/fisiopatologia , Canal de Potássio Kv1.1/genética , Contração Miocárdica , Potenciais de Ação , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Modelos Animais de Doenças , Canal de Potássio Kv1.1/deficiência , Canal de Potássio Kv1.1/metabolismo , Camundongos , Camundongos Knockout
14.
Elife ; 92020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33164746

RESUMO

Many voltage-dependent ion channels are regulated by accessory proteins. We recently reported powerful regulation of Kv1.2 potassium channels by the amino acid transporter Slc7a5. In this study, we report that Kv1.1 channels are also regulated by Slc7a5, albeit with different functional outcomes. In heterologous expression systems, Kv1.1 exhibits prominent current enhancement ('disinhibition') with holding potentials more negative than -120 mV. Knockdown of endogenous Slc7a5 leads to larger Kv1.1 currents and strongly attenuates the disinhibition effect, suggesting that Slc7a5 regulation of Kv1.1 involves channel inhibition that can be reversed by supraphysiological hyperpolarizing voltages. We investigated chimeric combinations of Kv1.1 and Kv1.2, demonstrating that exchange of the voltage-sensing domain controls the sensitivity and response to Slc7a5, and localize a specific position in S1 with prominent effects on Slc7a5 sensitivity. Overall, our study highlights multiple Slc7a5-sensitive Kv1 subunits, and identifies the voltage-sensing domain as a determinant of Slc7a5 modulation of Kv1 channels.


Assuntos
Canal de Potássio Kv1.1/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Animais , Linhagem Celular , Eletrofisiologia , Fibroblastos , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Ativação do Canal Iônico , Canal de Potássio Kv1.1/genética , Transportador 1 de Aminoácidos Neutros Grandes/genética , Camundongos
15.
Epilepsia ; 61(12): 2836-2846, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33104247

RESUMO

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.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neurônios/fisiologia , Convulsões/etiologia , Animais , Regulação para Baixo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.1/fisiologia , Camundongos , Camundongos Knockout , Comunicação Parácrina , RNA Interferente Pequeno , Convulsões/fisiopatologia , Sinapses/metabolismo , Sinapses/fisiologia
16.
BMC Biol ; 18(1): 143, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33059680

RESUMO

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.


Assuntos
Canal de Potássio Kv1.1/genética , Poli(ADP-Ribose) Polimerases/genética , Proteína Quinase C-delta/genética , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Animais , Células HEK293 , Células HeLa , Humanos , Canal de Potássio Kv1.1/metabolismo , Camundongos , Poli(ADP-Ribose) Polimerases/metabolismo , Proteína Quinase C-delta/metabolismo , Proteínas Proto-Oncogênicas/metabolismo
17.
Int J Mol Sci ; 21(20)2020 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-33066705

RESUMO

(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.


Assuntos
Ataxia/genética , Ativação do Canal Iônico , Canal de Potássio Kv1.1/genética , Mutação , Mioquimia/genética , Animais , Humanos , Canal de Potássio Kv1.1/química , Canal de Potássio Kv1.1/metabolismo , Xenopus
18.
Metabolomics ; 16(10): 106, 2020 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-33021695

RESUMO

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.


Assuntos
Epilepsia/metabolismo , Metabolismo dos Lipídeos/fisiologia , Animais , Dieta Cetogênica/métodos , Modelos Animais de Doenças , Epilepsia/dietoterapia , Hipocampo/metabolismo , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout
19.
Int J Mol Sci ; 21(18)2020 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-32899446

RESUMO

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.


Assuntos
Ataxia/genética , Ataxia/metabolismo , Ataxia/fisiopatologia , Canais de Cálcio/genética , Ataxia Cerebelar/genética , Transportador 1 de Aminoácido Excitatório/genética , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.2/genética
20.
Bioorg Chem ; 100: 103918, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32428746

RESUMO

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.


Assuntos
Canal de Potássio Kv1.1/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Animais , Linhagem Celular , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/metabolismo , Desenho de Fármacos , Células HEK293 , Humanos , Canal de Potássio Kv1.1/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Neurônios/metabolismo , Bloqueadores dos Canais de Potássio/síntese química , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA