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










Base de dados
Intervalo de ano de publicação
1.
Neurobiol Dis ; 155: 105382, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33940180

RESUMO

The unique fast spiking (FS) phenotype of cortical parvalbumin-positive (PV) neurons depends on the expression of multiple subtypes of voltage-gated potassium channels (Kv). PV neurons selectively express Kcns3, the gene encoding Kv9.3 subunits, suggesting that Kcns3 expression is critical for the FS phenotype. KCNS3 expression is lower in PV neurons in the neocortex of subjects with schizophrenia, but the effects of this alteration are unclear, because Kv9.3 subunit function is poorly understood. Therefore, to assess the role of Kv9.3 subunits in PV neuron function, we combined gene expression analyses, computational modeling, and electrophysiology in acute slices from the cortex of Kcns3-deficient mice. Kcns3 mRNA levels were ~ 50% lower in cortical PV neurons from Kcns3-deficient relative to wildtype mice. While silent per se, Kv9.3 subunits are believed to amplify the Kv2.1 current in Kv2.1-Kv9.3 channel complexes. Hence, to assess the consequences of reducing Kv9.3 levels, we simulated the effects of decreasing the Kv2.1-mediated current in a computational model. The FS cell model with reduced Kv2.1 produced spike trains with irregular inter-spike intervals, or stuttering, and greater Na+ channel inactivation. As in the computational model, PV basket cells (PVBCs) from Kcns3-deficient mice displayed spike trains with strong stuttering, which depressed PVBC firing. Moreover, Kcns3 deficiency impaired the recruitment of PVBC firing at gamma frequency by stimuli mimicking synaptic input observed during cortical UP states. Our data indicate that Kv9.3 subunits are critical for PVBC physiology and suggest that KCNS3 deficiency in schizophrenia could impair PV neuron firing, possibly contributing to deficits in cortical gamma oscillations in the illness.


Assuntos
Potenciais de Ação/fisiologia , Neurônios/fisiologia , Parvalbuminas/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Córtex Pré-Frontal/fisiopatologia , Esquizofrenia/fisiopatologia , Animais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Esquizofrenia/genética
2.
Int J Mol Sci ; 22(4)2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-33562460

RESUMO

Post-translational regulations of Shaker-like voltage-gated K+ channels were reported to be essential for rapid responses to environmental stresses in plants. In particular, it has been shown that calcium-dependent protein kinases (CPKs) regulate Shaker channels in plants. Here, the focus was on KAT2, a Shaker channel cloned in the model plant Arabidopsis thaliana, where is it expressed namely in the vascular tissues of leaves. After co-expression of KAT2 with AtCPK6 in Xenopuslaevis oocytes, voltage-clamp recordings demonstrated that AtCPK6 stimulates the activity of KAT2 in a calcium-dependent manner. A physical interaction between these two proteins has also been shown by Förster resonance energy transfer by fluorescence lifetime imaging (FRET-FLIM). Peptide array assays support that AtCPK6 phosphorylates KAT2 at several positions, also in a calcium-dependent manner. Finally, K+ fluorescence imaging in planta suggests that K+ distribution is impaired in kat2 knock-out mutant leaves. We propose that the AtCPK6/KAT2 couple plays a role in the homeostasis of K+ distribution in leaves.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Sinalização do Cálcio , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Feminino , Transferência Ressonante de Energia de Fluorescência , Técnicas de Inativação de Genes , Técnicas In Vitro , Modelos Moleculares , Oócitos/metabolismo , Imagem Óptica , Técnicas de Patch-Clamp , Fosforilação , Folhas de Planta/metabolismo , Plantas Geneticamente Modificadas , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
3.
Inflammation ; 43(5): 1832-1845, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32519270

RESUMO

Potassium voltage-gated channel subfamily Q member 1 opposite strand 1 (KCNQ1OT1), a long non-coding RNA found in the KCNQ1 locus, has been evidenced to play important roles in the aggravation of inflammatory and oxidative stresses under hypoxia, but whether and how KCNQ1OT1 contributes to neuronal damages in the cerebral ischemic stroke remains unknown. In the present study, we found a dominant upregulation of KCNQ1OT1 both in the plasma of cerebral ischemia patients and in an oxygen-glucose deprivation and reperfusion (OGD/R) model in PC12 cells. KCNQ1OT1 knocking-down significantly ameliorated the inflammation, oxidative stress, and cell apoptosis induced by OGD/R. We further demonstrated that KCNQ1OT1 directly bound to and suppressed the expression of miR-140-3p. Overexpressing miR-140-3p significantly alleviated both the inflammation, oxidative stress, and cell apoptosis in OGD/R, while all those cytoprotective effects of miR-140-3p-overexpression were hindered by the co-overexpression of KCNQ1OT1. Furthermore, we found a direct interaction between miR-140-3p and the hypoxia-inducible factor-1α (HIF-1α), which was suppressed by the upregulation of KCNQ1OT1 in OGD/R. Our results indicate that KCNQ1OT1 exacerbates cerebral ischemia-reperfusion injury by targeted binding to miR-140-3p, thus interfering its direct interaction with HIF-1α. These data provide novel therapeutic targets in the cerebral ischemic stroke.


Assuntos
Isquemia Encefálica/metabolismo , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Isquemia Encefálica/patologia , Técnicas de Silenciamento de Genes/métodos , Humanos , Células PC12 , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Ligação Proteica , RNA Interferente Pequeno/farmacologia , Ratos , Traumatismo por Reperfusão/patologia
4.
Physiol Rep ; 7(3): e13957, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30737904

RESUMO

Sudden cardiac death (SCD) is the leading global cause of mortality. SCD often arises from cardiac ischemia reperfusion (IR) injury, pathologic sequence variants within ion channel genes, or a combination of the two. Alternative approaches are needed to prevent or ameliorate ventricular arrhythmias linked to SCD. Here, we investigated the efficacy of remote ischemic preconditioning (RIPC) of the limb versus the liver in reducing ventricular arrhythmias in a mouse model of SCD. Mice lacking the Kcne2 gene, which encodes a potassium channel ß subunit associated with acquired Long QT syndrome were exposed to IR injury via coronary ligation. This resulted in ventricular arrhythmias in all mice (15/15) and SCD in 5/15 mice during reperfusion. Strikingly, prior RIPC (limb or liver) greatly reduced the incidence and severity of all ventricular arrhythmias and completely prevented SCD. Biochemical and pharmacological analysis demonstrated that RIPC cardioprotection required ERK1/2 and/or AKT phosphorylation. A lack of alteration in GSK-3ß phosphorylation suggested against conventional reperfusion injury salvage kinase (RISK) signaling pathway protection. If replicated in human studies, limb RIPC could represent a noninvasive, nonpharmacological approach to limit dangerous ventricular arrhythmias associated with ischemia and/or channelopathy-linked SCD.


Assuntos
Arritmias Cardíacas/prevenção & controle , Morte Súbita Cardíaca/prevenção & controle , Extremidades/irrigação sanguínea , Precondicionamento Isquêmico/métodos , Fígado/irrigação sanguínea , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/enzimologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Arritmias Cardíacas/enzimologia , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Circulação Hepática , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Fosforilação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Fluxo Sanguíneo Regional , Transdução de Sinais
5.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R552-R562, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30758982

RESUMO

Activation of antiapoptotic signaling cascades, such as the reperfusion injury salvage kinase (RISK) and survivor activating factor enhancement (SAFE) pathways, is protective in a variety of tissues in the context of ischemia-reperfusion (IR) injury. Hepatic IR injury causes clinically significant hepatocellular damage in surgical procedures, including liver transplantation and hepatic resection, increasing associated morbidity and mortality. We previously found that the cardiovascular-expressed K+ voltage-gated channel ancillary subunit KCNE4 sex specifically influences the cardiac RISK/SAFE pathway response to IR and that Kcne4 deletion testosterone dependently exacerbates cardiac IR injury. Here, we discovered that germline Kcne4 deletion exacerbates hepatic IR injury damage in 13-mo-old male mice, despite a lack of Kcne4 expression in male mouse liver. Examining RISK/SAFE pathway induction, we found that Kcne4 deletion prevents the hepatic ERK1/2 phosphorylation response to IR injury. Conversely, in 13-mo-old female mice, Kcne4 deletion increased both baseline and post-IR GSK-3ß inhibitory phosphorylation, and pharmacological GSK-3ß inhibition was hepatoprotective. Finally, castration of male mice restored normal hepatic RISK/SAFE pathway responses in Kcne4-/- mice, eliminated Kcne4 deletion-dependent serum alanine aminotransferase elevation, and genotype independently augmented the hepatic post-IR GSK-3ß phosphorylation response. These findings support a role for KCNE4 as a systemic modulator of IR injury response and uncover hormonally influenced, sex-specific, KCNE4-dependent and -independent RISK/SAFE pathway induction.


Assuntos
Hepatopatias/prevenção & controle , Fígado/enzimologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Traumatismo por Reperfusão/prevenção & controle , Animais , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Deleção de Genes , Glicogênio Sintase Quinase 3 beta/metabolismo , Fígado/patologia , Hepatopatias/enzimologia , Hepatopatias/genética , Hepatopatias/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orquiectomia , Fosforilação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Fatores Sexuais , Transdução de Sinais , Testosterona/metabolismo
6.
J Vasc Res ; 53(3-4): 138-148, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27710966

RESUMO

Voltage-gated potassium (Kv) channels formed by Kv7 (KCNQ) α-subunits are recognized as crucial for vascular smooth muscle function, in addition to their established roles in the heart (Kv7.1) and the brain (Kv7.2-5). In vivo, Kv7 α-subunits are often regulated by KCNE subfamily ancillary (ß) subunits. We investigated the effects of targeted germline Kcne4 deletion on mesenteric artery reactivity in adult male and female mice. Kcne4 deletion increased mesenteric artery contractility in response to α-adrenoceptor agonist methoxamine, and decreased responses to Kv7.2-7.5 channel activator ML213, in male but not female mice. In contrast, Kcne4 deletion markedly decreased vasorelaxation in response to isoprenaline in both male and female mice. Kcne4 expression was 2-fold lower in the female versus the male mouse mesenteric artery, and Kcne4 deletion elicited only moderate changes of other Kcne transcripts, with no striking sex-specific differences. However, Kv7.4 protein expression in females was twice that in males, and was reduced in both sexes by Kcne4 deletion. Our findings confirm a crucial role for KCNE4 in regulation of Kv7 channel activity to modulate vascular tone, and provide the first known molecular mechanism for sex-specificity of this modulation that has important implications for vascular reactivity and may underlie sex-specific susceptibility to cardiovascular diseases.


Assuntos
Canais de Potássio KCNQ/metabolismo , Músculo Liso Vascular/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Vasoconstrição , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Anilidas/farmacologia , Animais , Compostos Bicíclicos com Pontes/farmacologia , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica , Genótipo , Canais de Potássio KCNQ/efeitos dos fármacos , Canais de Potássio KCNQ/genética , Masculino , Artérias Mesentéricas/metabolismo , Metoxamina/farmacologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Fatores Sexuais , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Vasodilatadores/farmacologia
7.
Sci Rep ; 6: 23118, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26984260

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is an increasing health problem worldwide, with genetic, epigenetic, and environmental components. Here, we describe the first example of NAFLD caused by genetic disruption of a mammalian potassium channel subunit. Mice with germline deletion of the KCNE2 potassium channel ß subunit exhibited NAFLD as early as postnatal day 7. Using mouse genetics, histology, liver damage assays and transcriptomics we discovered that iron deficiency arising from KCNE2-dependent achlorhydria is a major factor in early-onset NAFLD in Kcne2(─/─) mice, while two other KCNE2-dependent defects did not initiate NAFLD. The findings uncover a novel genetic basis for NAFLD and an unexpected potential factor in human KCNE2-associated cardiovascular pathologies, including atherosclerosis.


Assuntos
Anemia Ferropriva/complicações , Hepatopatia Gordurosa não Alcoólica/etiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Animais , Proteína C-Reativa/análise , Dieta Hiperlipídica , Feminino , Redes Reguladoras de Genes , Mutação em Linhagem Germinativa , Homocisteína/sangue , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Deleção de Sequência , Transcriptoma , Triglicerídeos/sangue
8.
Cardiovasc Res ; 110(2): 227-37, 2016 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-26952045

RESUMO

AIMS: Most cardiac arrhythmia-associated genes encode ion channel subunits and regulatory proteins that are also expressed outside the heart, suggesting that diseases linked to their disruption may be multifactorial. KCNE2 is a ubiquitously expressed potassium channel ß subunit associated with cardiac arrhythmia, atherosclerosis, and myocardial infarction (MI) in human populations. Here, we tested the hypothesis that Kcne2 disruption in mice would influence the acute outcome of experimentally induced MI. METHODS AND RESULTS: One-year-old male Kcne2⁺/⁺ and Kcne2⁻/⁻ mice were subjected to cardiac ischaemia/reperfusion injury (IRI) by left anterior descending coronary artery ligation. After reperfusion (3 h), infarct size and markers of tissue damage were quantified. Unexpectedly, post-reperfusion, Kcne2⁻/⁻ mice exhibited 40% lower infarct size, decreased myocardial apoptosis and damage, and more than two-fold lower serum levels of damage markers, lactate dehydrogenase and creatine kinase, than Kcne2⁺/⁺ mice. Kcne2 deletion, despite increasing normalized heart weight and prolonging baseline QTc by 70%, helped preserve post-infarct cardiac function (quantified by a Millar catheter), with parameters including left ventricular maximum pressure, max dP/dt (P < 0.01), contractility index, and pressure/time index (P < 0.05) all greater in Kcne2⁻/⁻ compared with Kcne2⁺/⁺ mice. Western blotting indicated two-fold-increased glycogen synthase kinase 3ß (GSK-3ß) phosphorylation (inactivation) before and after IRI (P < 0.05) in Kcne2⁻/⁻ mice compared with Kcne2⁺/⁺ mice. GSK-3ß inhibition by SB216763 mimicked in Kcne2⁺/⁺ mice the cardioprotective effects of Kcne2 deletion, but did not further enhance them in Kcne2⁻/⁻mice, suggesting that GSK-3ß inactivation was a primary cardioprotective mechanism arising from Kcne2 deletion. CONCLUSIONS: Kcne2 deletion preconditions the heart, attenuating the acute tissue damage caused by an imposed IRI. The findings contribute further evidence that genetic disruption of arrhythmia-associated ion channel genes has cardiac ramifications beyond abnormal electrical activity.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/genética , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Modelos Animais de Doenças , Indóis/administração & dosagem , Indóis/farmacologia , Isquemia/tratamento farmacológico , Isquemia/genética , Maleimidas/administração & dosagem , Maleimidas/farmacologia , Camundongos Knockout , Infarto do Miocárdio/metabolismo , Fosforilação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência
9.
Exp Hematol ; 42(12): 1053-8.e1, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25127743

RESUMO

Iron homeostasis is a dynamic process that is tightly controlled to balance iron uptake, storage, and export. Reduction of dietary iron from the ferric to the ferrous form is required for uptake by solute carrier family 11 (proton-coupled divalent metal ion transporters), member 2 (Slc11a2) into the enterocytes. Both processes are proton dependent and have led to the suggestion of the importance of acidic gastric pH for the absorption of dietary iron. Potassium voltage-gated channel subfamily E, member 2 (KCNE2), in combination with potassium voltage-gated channel, KQT-like subfamily, member 1 (KCNQ1), form a gastric potassium channel essential for gastric acidification. Deficiency of either Kcne2 or Kcnq1 results in achlorhydia, gastric hyperplasia, and neoplasia, but the impact on iron absorption has not, to our knowledge, been investigated. Here we report that Kcne2-deficient mice, in addition to the previously reported phenotypes, also present with iron-deficient anemia. Interestingly, impaired function of KCNQ1 results in iron-deficient anemia in Jervell and Lange-Nielsen syndrome patients. We speculate that impaired function of KCNE2 could result in the same clinical phenotype.


Assuntos
Anemia Ferropriva/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Anemia Ferropriva/sangue , Animais , Dieta Ocidental , Índices de Eritrócitos , Eritropoetina/sangue , Feminino , Ferritinas/sangue , Heterogeneidade Genética , Hematócrito , Humanos , Canal de Potássio KCNQ1/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Caracteres Sexuais , Especificidade da Espécie , Organismos Livres de Patógenos Específicos , Transferrina/análise
10.
Circ Cardiovasc Genet ; 7(1): 33-42, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24403551

RESUMO

BACKGROUND: Sudden cardiac death (SCD) is the leading global cause of mortality, exhibiting increased incidence in patients with diabetes mellitus. Ion channel gene perturbations provide a well-established ventricular arrhythmogenic substrate for SCD. However, most arrhythmia-susceptibility genes, including the KCNE2 K(+) channel ß subunit, are expressed in multiple tissues, suggesting potential multiplex SCD substrates. METHODS AND RESULTS: Using whole-transcript transcriptomics, we uncovered cardiac angiotensinogen upregulation and remodeling of cardiac angiotensinogen interaction networks in P21 Kcne2(-/-) mouse pups and adrenal remodeling consistent with metabolic syndrome in adult Kcne2(-/-) mice. This led to the discovery that Kcne2 disruption causes multiple acknowledged SCD substrates of extracardiac origin: diabetes mellitus, hypercholesterolemia, hyperkalemia, anemia, and elevated angiotensin II. Kcne2 deletion was also a prerequisite for aging-dependent QT prolongation, ventricular fibrillation and SCD immediately after transient ischemia, and fasting-dependent hypoglycemia, myocardial ischemia, and AV block. CONCLUSIONS: Disruption of a single, widely expressed arrhythmia-susceptibility gene can generate a multisystem syndrome comprising manifold electric and systemic substrates and triggers of SCD. This paradigm is expected to apply to other arrhythmia-susceptibility genes, the majority of which encode ubiquitously expressed ion channel subunits or regulatory proteins.


Assuntos
Arritmias Cardíacas/genética , Morte Súbita Cardíaca/etiologia , Síndrome do QT Longo/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Anemia/etiologia , Angiotensina II/metabolismo , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Animais , Arritmias Cardíacas/patologia , Diabetes Mellitus/etiologia , Dislipidemias/etiologia , Redes Reguladoras de Genes , Genótipo , Heterozigoto , Hiperpotassemia/etiologia , Isquemia/etiologia , Síndrome do QT Longo/patologia , Camundongos , Camundongos Knockout , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo
11.
FASEB J ; 28(2): 935-45, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24225147

RESUMO

Mutations in the human KCNE3 potassium channel ancillary subunit gene are associated with life-threatening ventricular arrhythmias. Most genes underlying inherited cardiac arrhythmias, including KCNE3, are not exclusively expressed in the heart, suggesting potentially complex disease etiologies. Here we investigated mechanisms of KCNE3-linked arrhythmogenesis in Kcne3(-/-) mice using real-time qPCR, echo- and electrocardiography, ventricular myocyte patch-clamp, coronary artery ligation/reperfusion, blood analysis, cardiac synaptosome exocytosis, microarray and pathway analysis, and multitissue histology. Kcne3 transcript was undetectable in adult mouse atria, ventricles, and adrenal glands, but Kcne3(-/-) mice exhibited 2.3-fold elevated serum aldosterone (P=0.003) and differentially expressed gene networks consistent with an adrenal-targeted autoimmune response. Furthermore, 8/8 Kcne3(-/-) mice vs. 0/8 Kcne3(+/+) mice exhibited an activated-lymphocyte adrenal infiltration (P=0.0002). Kcne3 deletion also caused aldosterone-dependent ventricular repolarization delay (19.6% mean QTc prolongation in females; P<0.05) and aldosterone-dependent predisposition to postischemia arrhythmogenesis. Thus, 5/11 Kcne3(-/-) mice vs. 0/10 Kcne3(+/+) mice exhibited sustained ventricular tachycardia during reperfusion (P<0.05). Kcne3 deletion is therefore arrhythmogenic by a novel mechanism in which secondary hyperaldosteronism, associated with an adrenal-specific lymphocyte infiltration, impairs ventricular repolarization. The findings highlight the importance of considering extracardiac pathogenesis when investigating arrhythmogenic mechanisms, even in inherited, monogenic channelopathies.


Assuntos
Arritmias Cardíacas/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Aldosterona/sangue , Animais , Arritmias Cardíacas/sangue , Arritmias Cardíacas/genética , Eletrocardiografia , Feminino , Masculino , Camundongos , Camundongos Mutantes , Canais de Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Reação em Cadeia da Polimerase em Tempo Real
12.
J Neurosci ; 31(46): 16541-9, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22090480

RESUMO

The mechanotransduction of vestibular sensory cells depends on the high endolymphatic potassium concentration ([K+]) maintained by a fine balance between K+ secretion and absorption by epithelial cells. Despite the crucial role of endolymph as an electrochemical motor for mechanotransduction, little is known about the processes that govern endolymph formation. To address these, we took advantage of an organotypic rodent model, which regenerates a genuine neonatal vestibular endolymphatic compartment, facilitating the determination of endolymphatic [K+] and transepithelial potential (Vt) during endolymph formation. While mature Vt levels are almost immediately achieved, K+ accumulates to reach a steady [K+] by day 5 in culture. Inhibition of sensory cell K+ efflux enhances [K+] regardless of the blocker used (FM1.43, amikacin, gentamicin, or gadolinium). Targeting K+ secretion with bumetanide partially and transiently reduces [K+], while ouabain application and Kcne1 deletion almost abolishes it. Immunofluorescence studies demonstrate that dark cells do not express Na-K-2Cl cotransporter 1 (the target of bumetanide) in cultured and young mouse utricles, while Na/K-ATPase (the target of ouabain) is found in dark cells and transitional cells. This global analysis of the involvement of endolymphatic homeostasis actors in the immature organ (1) confirms that KCNE1 channels are necessary for K+ secretion, (2) highlights Na/K-ATPase as the key endolymphatic K+ provider and shows that Na-K-2Cl cotransporter 1 has a limited impact on K+ influx, and (3) demonstrates that transitional cells are involved in K+ secretion in the early endolymphatic compartment.


Assuntos
Endolinfa/metabolismo , Células Epiteliais/fisiologia , Sáculo e Utrículo/crescimento & desenvolvimento , Sáculo e Utrículo/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Aminoglicosídeos/farmacologia , Animais , Animais Recém-Nascidos , Bumetanida/farmacologia , Endocitose/genética , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Feminino , Gadolínio/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Ouabaína/farmacologia , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Compostos de Piridínio/farmacocinética , Compostos de Amônio Quaternário/farmacocinética , Ratos , Ratos Wistar , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Fatores de Tempo
13.
FASEB J ; 25(12): 4264-73, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21859894

RESUMO

Cerebrospinal fluid (CSF) is crucial for normal function and mechanical protection of the CNS. The choroid plexus epithelium (CPe) is primarily responsible for secreting CSF and regulating its composition by mechanisms currently not fully understood. Previously, the heteromeric KCNQ1-KCNE2 K(+) channel was functionally linked to epithelial processes including gastric acid secretion and thyroid hormone biosynthesis. Here, using Kcne2(-/-) tissue as a negative control, we found cerebral expression of KCNE2 to be markedly enriched in the CPe apical membrane, where we also discovered expression of KCNQ1. Targeted Kcne2 gene deletion in C57B6 mice increased CPe outward K(+) current 2-fold. The Kcne2 deletion-enhanced portion of the current was inhibited by XE991 (10 µM) and margatoxin (10 µM) but not by dendrotoxin (100 nM), indicating that it arose from augmentation of KCNQ subfamily and KCNA3 but not KCNA1 K(+) channel activity. Kcne2 deletion in C57B6 mice also altered the polarity of CPe KCNQ1 and KCNA3 trafficking, hyperpolarized the CPe membrane by 9 ± 2 mV, and increased CSF [Cl(-)] by 14% compared with wild-type mice. These findings constitute the first report of CPe dysfunction caused by cation channel gene disruption and suggest that KCNE2 influences blood-CSF anion flux by regulating KCNQ1 and KCNA3 in the CPe.


Assuntos
Plexo Corióideo/metabolismo , Canal de Potássio KCNQ1/metabolismo , Canal de Potássio Kv1.3/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Cloretos/sangue , Cloretos/líquido cefalorraquidiano , Epitélio/metabolismo , Transporte de Íons , Canal de Potássio KCNQ1/química , Canal de Potássio Kv1.3/química , Potenciais da Membrana , Camundongos , Camundongos Knockout , Modelos Biológicos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Subunidades Proteicas
14.
J Physiol ; 589(Pt 14): 3595-609, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21576273

RESUMO

KCNE1 is a protein of low molecular mass that is known to regulate the chromanol 293B and clofilium-sensitive K+ channel, KCNQ1, in a number of tissues. Previous work on the kidney of KCNE1 and KCNQ1 knockout mice has revealed that these animals have different renal phenotypes, suggesting that KCNE1 may not regulate KCNQ1 in the renal system. In the current study, in vivo clearance approaches and whole cell voltage-clamp recordings from isolated renal proximal tubules were used to examine the physiological role of KCNE1. Data from wild-type mice were compared to those from KCNE1 knockout mice. In clearance studies the KCNE1 knockout mice had an increased fractional excretion of Na+, Cl−, HCO3(−) and water. This profile was mimicked in wild-type mice by infusion of chromanol 293B, while chromanol was without effect in KCNE1 knockout animals. Clofilium also increased the fractional excretion of Na+, Cl− and water, but this was observed in both wild-type and knockout mice, suggesting that KCNE1 was regulating a chromanol-sensitive but clofilium-insensitive pathway. In whole cell voltage clamp recordings from proximal tubules, a chromanol-sensitive, K+-selective conductance was identified that was absent in tubules from knockout animals. The properties of this conductance were not consistent with its being mediated by KCNQ1, suggesting that KCNE1 regulates another K+ channel in the renal proximal tubule. Taken together these data suggest that KCNE1 regulates a K+-selective conductance in the renal proximal tubule that plays a relatively minor role in driving the transport of Na+, Cl− and HCO3(−).


Assuntos
Cromanos/farmacologia , Canal de Potássio KCNQ1/metabolismo , Túbulos Renais Proximais/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Sulfonamidas/farmacologia , Animais , Bário/farmacologia , Bicarbonatos/metabolismo , Cloretos/metabolismo , Glucose/metabolismo , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Canal de Potássio KCNQ1/antagonistas & inibidores , Túbulos Renais Proximais/efeitos dos fármacos , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Compostos de Amônio Quaternário/farmacologia , Sódio/metabolismo
15.
Hear Res ; 276(1-2): 61-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21056098

RESUMO

Acoustic information is brought to the brain by auditory nerve fibers, all of which terminate in the cochlear nuclei, and is passed up the auditory pathway through the principal cells of the cochlear nuclei. A population of neurons variously known as T stellate, type I multipolar, planar multipolar, or chopper cells forms one of the major ascending auditory pathways through the brainstem. T Stellate cells are sharply tuned; as a population they encode the spectrum of sounds. In these neurons, phasic excitation from the auditory nerve is made more tonic by feedforward excitation, coactivation of inhibitory with excitatory inputs, relatively large excitatory currents through NMDA receptors, and relatively little synaptic depression. The mechanisms that make firing tonic also obscure the fine structure of sounds that is represented in the excitatory inputs from the auditory nerve and account for the characteristic chopping response patterns with which T stellate cells respond to tones. In contrast with other principal cells of the ventral cochlear nucleus (VCN), T stellate cells lack a low-voltage-activated potassium conductance and are therefore sensitive to small, steady, neuromodulating currents. The presence of cholinergic, serotonergic and noradrenergic receptors allows the excitability of these cells to be modulated by medial olivocochlear efferent neurons and by neuronal circuits associated with arousal. T Stellate cells deliver acoustic information to the ipsilateral dorsal cochlear nucleus (DCN), ventral nucleus of the trapezoid body (VNTB), periolivary regions around the lateral superior olivary nucleus (LSO), and to the contralateral ventral lemniscal nuclei (VNLL) and inferior colliculus (IC). It is likely that T stellate cells participate in feedback loops through both medial and lateral olivocochlear efferent neurons and they may be a source of ipsilateral excitation of the LSO.


Assuntos
Núcleo Coclear/citologia , Núcleo Coclear/fisiologia , Neurônios Aferentes/fisiologia , Animais , Vias Auditivas/fisiologia , Aves/fisiologia , Tronco Encefálico/fisiologia , Cóclea/fisiologia , Nervo Coclear/fisiologia , Dendritos/fisiologia , Fenômenos Eletrofisiológicos , Retroalimentação Fisiológica , Humanos , Inibição Neural , Neurotransmissores/metabolismo , Núcleo Olivar/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Receptores de N-Metil-D-Aspartato/metabolismo , Som , Sinapses/fisiologia , Fatores de Tempo
16.
Lancet Neurol ; 9(8): 776-85, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20580615

RESUMO

BACKGROUND: Voltage-gated potassium channels are thought to be the target of antibodies associated with limbic encephalitis. However, antibody testing using cells expressing voltage-gated potassium channels is negative; hence, we aimed to identify the real autoantigen associated with limbic encephalitis. METHODS: We analysed sera and CSF of 57 patients with limbic encephalitis and antibodies attributed to voltage-gated potassium channels and 148 control individuals who had other disorders with or without antibodies against voltage-gated potassium channels. Immunohistochemistry, immunoprecipitation, and mass spectrometry were used to characterise the antigen. An assay with HEK293 cells transfected with leucine-rich, glioma-inactivated 1 (LGI1) and disintegrin and metalloproteinase domain-containing protein 22 (ADAM22) or ADAM23 was used as a serological test. The identity of the autoantigen was confirmed by immunoabsorption studies and immunostaining of Lgi1-null mice. FINDINGS: Immunoprecipitation and mass spectrometry analyses showed that antibodies from patients with limbic encephalitis previously attributed to voltage-gated potassium channels recognise LGI1, a neuronal secreted protein that interacts with presynaptic ADAM23 and postsynaptic ADAM22. Immunostaining of HEK293 cells transfected with LGI1 showed that sera or CSF from patients, but not those from control individuals, recognised LGI1. Co-transfection of LGI1 with its receptors, ADAM22 or ADAM23, changed the pattern of reactivity and improved detection. LGI1 was confirmed as the autoantigen by specific abrogation of reactivity of sera and CSF from patients after immunoabsorption with LGI1-expressing cells and by comparative immunostaining of wild-type and Lgi1-null mice, which showed selective lack of reactivity in brains of Lgi1-null mice. One patient with limbic encephalitis and antibodies against LGI1 also had antibodies against CASPR2, an autoantigen we identified in some patients with encephalitis and seizures, Morvan's syndrome, and neuromyotonia. INTERPRETATION: LGI1 is the autoantigen associated with limbic encephalitis previously attributed to voltage-gated potassium channels. The term limbic encephalitis associated with antibodies against voltage-gated potassium channels should be changed to limbic encephalitis associated with LGI1 antibodies, and this disorder should be classed as an autoimmune synaptic encephalopathy. FUNDING: National Institutes of Health, National Cancer Institute, and Euroimmun.


Assuntos
Autoantígenos/imunologia , Encefalite Límbica/imunologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/imunologia , Proteínas/imunologia , Proteínas ADAM/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Autoanticorpos , Autoantígenos/genética , Linhagem Celular , Epilepsia/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Ligantes , Encefalite Límbica/genética , Encefalite Límbica/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Proteínas/genética , Proteínas/metabolismo , Ratos , Ratos Wistar
17.
J Biol Chem ; 281(33): 23740-7, 2006 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-16754665

RESUMO

Genes in the KCNE family encode single transmembrane domain ancillary subunits that co-assemble with voltage-gated potassium (Kv) channel alpha subunits to alter their function. KCNE2 (also known as MiRP1) is expressed in the heart, is associated with human cardiac arrhythmia, and modulates cardiac Kv alpha subunits hERG and KCNQ1 in vitro. KCNE2 and KCNQ1 are also expressed in parietal cells, leading to speculation they form a native channel complex there. Here, we disrupted the murine kcne2 gene and found that kcne2 (-/-) mice have a severe gastric phenotype with profoundly reduced parietal cell proton secretion, abnormal parietal cell morphology, achlorhydria, hypergastrinemia, and striking gastric glandular hyperplasia arising from an increase in the number of non-acid secretory cells. KCNQ1 exhibited abnormal distribution in gastric glands from kcne2 (-/-) mice, with increased expression in non-acid secretory cells. Parietal cells from kcne2 (+/-) mice exhibited normal architecture but reduced proton secretion, and kcne2 (+/-) mice were hypochlorhydric, indicating a gene-dose effect and a primary defect in gastric acid secretion. These data demonstrate that KCNE2 is essential for gastric acid secretion, the first genetic evidence that a member of the KCNE gene family is required for normal gastrointestinal function.


Assuntos
Ácido Gástrico/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Subunidades Proteicas/fisiologia , Acloridria/genética , Animais , Células Cultivadas , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Mucosa Gástrica/ultraestrutura , Gastrinas/antagonistas & inibidores , Gastrinas/biossíntese , Marcação de Genes , Hiperplasia/genética , Hipertrofia/genética , Canal de Potássio KCNQ1/biossíntese , Canal de Potássio KCNQ1/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Parietais Gástricas/metabolismo , Células Parietais Gástricas/patologia , Células Parietais Gástricas/ultraestrutura , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Estômago/patologia , Estômago/ultraestrutura , Regulação para Cima/genética
18.
J Neurophysiol ; 94(3): 1770-80, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15917322

RESUMO

Using patch-clamp techniques, we investigated the characteristics of the spontaneous oscillatory activity displayed by starburst amacrine cells in the mouse retina. At a holding potential of -70 mV, oscillations appeared as spontaneous, rhythmic inward currents with a frequency of approximately 3.5 Hz and an average maximal amplitude of approximately 120 pA. Application of TEA, a potassium channel blocker, increased the amplitude of oscillatory currents by >70% but reduced their frequency by approximately 17%. The TEA effects did not appear to result from direct actions on starburst cells, but rather a modulation of their synaptic inputs. Oscillatory currents were inhibited by 6-cyano-7-nitroquinoxalene-2,3-dione (CNQX), an antagonist of AMPA/kainate receptors, indicating that they were dependent on a periodic glutamatergic input likely from presynaptic bipolar cells. The oscillations were also inhibited by the calcium channel blockers cadmium and nifedipine, suggesting that the glutamate release was calcium dependent. Application of AP4, an agonist of mGluR6 receptors on on-center bipolar cells, blocked the oscillatory currents in starburst cells. However, application of TEA overcame the AP4 blockade, suggesting that the periodic glutamate release from bipolar cells is intrinsic to the inner plexiform layer in that, under experimental conditions, it can occur independent of photoreceptor input. The GABA receptor antagonists picrotoxin and bicuculline enhanced the amplitude of oscillations in starburst cells prestimulated with TEA. Our results suggest that this enhancement was due to a reduction of a GABAergic feedback inhibition from amacrine cells to bipolar cells and the resultant increased glutamate release. Finally, we found that some ganglion cells and other types of amacrine cell also displayed rhythmic activity, suggesting that oscillatory behavior is expressed by a number of inner retinal neurons.


Assuntos
Células Amácrinas/fisiologia , Relógios Biológicos/fisiologia , Potenciais da Membrana/fisiologia , Retina/citologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Células Amácrinas/efeitos dos fármacos , Células Amácrinas/efeitos da radiação , Aminobutiratos/farmacologia , Animais , Bicuculina/farmacologia , Relógios Biológicos/efeitos dos fármacos , Relógios Biológicos/efeitos da radiação , Cloreto de Cádmio/farmacologia , Relação Dose-Resposta à Radiação , Interações Medicamentosas , Estimulação Elétrica/métodos , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas GABAérgicos/farmacologia , Técnicas In Vitro , Lisina/análogos & derivados , Lisina/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/efeitos da radiação , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Técnicas de Patch-Clamp/métodos , Picrotoxina/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/fisiologia , Células Ganglionares da Retina/efeitos da radiação , Canais de Potássio Shaw , Tetraetilamônio/farmacologia , Fatores de Tempo
19.
Proc Natl Acad Sci U S A ; 102(7): 2596-601, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15695336

RESUMO

Egr2 is a transcription factor required for peripheral nerve myelination in rodents, and mutations in Egr2 are associated with congenital hypomyelinating neuropathy (CHN) in humans. To further study its role in myelination, we generated mice harboring a hypomorphic Egr2 allele (Egr2Lo) that survive for up to 3 weeks postnatally, a period of active myelination in rodents. These Egr2Lo/Lo mice provided the opportunity to study the molecular effects of Egr2 deficiency on Schwann cell biology, an analysis that was not possible previously, because of the perinatal lethality of Egr2-null mice. Egr2Lo/Lo mice phenocopy CHN, as evidenced by the severe hypomyelination and increased numbers of proliferating Schwann cells of the peripheral nerves. Comparison of sciatic nerve gene expression profiles during development and after crush injury with those of Egr2Lo/Lo Schwann cells revealed that they are developmentally arrested, with down-regulation of myelination-related genes and up-regulation of genes associated with immature and promyelinating Schwann cells. One of the abnormally elevated genes in Egr2Lo/Lo Schwann cells, Sox2, encodes a transcription factor that is crucial for maintenance of neural stem cell pluripotency. Wild-type Schwann cells infected with Sox2 adenovirus or lentivirus inhibited expression of myelination-associated genes (e.g., myelin protein zero; Mpz), and failed to myelinate axons in vitro, but had an enhanced proliferative response to beta-neuregulin. The characterization of a mouse model of CHN has provided insight into Schwann cell differentiation and allowed the identification of Sox2 as a negative regulator of myelination.


Assuntos
Proteínas de Ligação a DNA/genética , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/patologia , Proteínas Nucleares/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Células de Schwann/patologia , Animais , Diferenciação Celular , Doenças Desmielinizantes/metabolismo , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go , Perfilação da Expressão Gênica , Proteínas HMGB , Camundongos , Camundongos Mutantes Neurológicos , Bainha de Mielina/metabolismo , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/patologia , Fatores de Transcrição SOXB1 , Células de Schwann/metabolismo , Fatores de Transcrição
20.
Nat Neurosci ; 8(1): 51-60, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15608631

RESUMO

In humans, mutations in the KCNQ2 or KCNQ3 potassium-channel genes are associated with an inherited epilepsy syndrome. We have studied the contribution of KCNQ/M-channels to the control of neuronal excitability by using transgenic mice that conditionally express dominant-negative KCNQ2 subunits in brain. We show that suppression of the neuronal M current in mice is associated with spontaneous seizures, behavioral hyperactivity and morphological changes in the hippocampus. Restriction of transgene expression to defined developmental periods revealed that M-channel activity is critical to the development of normal hippocampal morphology during the first postnatal weeks. Suppression of the M current after this critical period resulted in mice with signs of increased neuronal excitability and deficits in hippocampus-dependent spatial memory. M-current-deficient hippocampal CA1 pyramidal neurons showed increased excitability, reduced spike-frequency adaptation, attenuated medium afterhyperpolarization and reduced intrinsic subthreshold theta resonance. M channels are thus critical determinants of cellular and neuronal network excitability, postnatal brain development and cognitive performance.


Assuntos
Comportamento Animal , Encéfalo/fisiopatologia , Epilepsia/fisiopatologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Animais , Fenômenos Biofísicos , Biofísica , Encéfalo/metabolismo , Encéfalo/patologia , Eletrofisiologia , Epilepsia/genética , Epilepsia/patologia , Epilepsia/psicologia , Feminino , Genes Dominantes , Hipercinese/genética , Canal de Potássio KCNQ2 , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Oócitos , Fenótipo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Canais de Potássio de Abertura Dependente da Tensão da Membrana/deficiência , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Células Piramidais , Natação , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...