Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Neurochem Int ; 150: 105155, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34384853

RESUMO

As well as their ion transportation function, the voltage-dependent potassium channels could act as the cell signal inducer in a variety of pathogenic processes. However, their roles in neurogenesis after stroke insults have not been clearly illustrated. In our preliminary study, the expressions of voltage-dependent potassium channels Kv4.2 was significantly decreased after stroke in cortex, striatum and hippocampus by real-time quantitative PCR assay. To underlie the neuroprotection of Kv4.2 in stroke rehabilitation, recombinant plasmids encoding the cDNAs of mouse Kv4.2 was constructed. Behavioral tests showed that the increased Kv4.2 could be beneficial to the recovery of the sensory, the motor functions and the cognitive deficits after stroke. Temozolomide (TMZ), an inhibitor of neurogenesis, could partially abolish the mentioned protections of Kv4.2. The immunocytochemical staining showed that Kv4.2 could promote the proliferations of neural stem cells and induce the neural stem cells to differentiate into neurons in vitro and in vivo. And Kv4.2 could up-regulate the expressions of ERK1/2, p-ERK1/2, p-STAT3, NGF, p-TrkA, and BDNF, CAMKII and the concentration of intracellular Ca2+. Namely, we concluded that Kv4.2 promoted neurogenesis through ERK1/2/STAT3, NGF/TrkA, Ca2+/CAMKII signal pathways and rescued the ischemic impairments. Kv4.2 might be a potential drug target for ischemic stroke intervention.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , AVC Isquêmico/metabolismo , AVC Isquêmico/prevenção & controle , Neurogênese/fisiologia , Canais de Potássio Shal/biossíntese , Animais , Isquemia Encefálica/genética , Linhagem Celular Transformada , AVC Isquêmico/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio Shal/análise , Canais de Potássio Shal/genética
2.
Mol Brain ; 14(1): 62, 2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33785038

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that causes memory loss. Most AD researches have focused on neurodegeneration mechanisms. Considering that neurodegenerative changes are not reversible, understanding early functional changes before neurodegeneration is critical to develop new strategies for early detection and treatment of AD. We found that Tg2576 mice exhibited impaired pattern separation at the early preclinical stage. Based on previous studies suggesting a critical role of dentate gyrus (DG) in pattern separation, we investigated functional changes in DG of Tg2576 mice. We found that granule cells in DG (DG-GCs) in Tg2576 mice showed increased action potential firing in response to long depolarizations and reduced 4-AP sensitive K+-currents compared to DG-GCs in wild-type (WT) mice. Among Kv4 family channels, Kv4.1 mRNA expression in DG was significantly lower in Tg2576 mice. We confirmed that Kv4.1 protein expression was reduced in Tg2576, and this reduction was restored by antioxidant treatment. Hyperexcitable DG and impaired pattern separation in Tg2576 mice were also recovered by antioxidant treatment. These results highlight the hyperexcitability of DG-GCs as a pathophysiologic mechanism underlying early cognitive deficits in AD and Kv4.1 as a new target for AD pathogenesis in relation to increased oxidative stress.


Assuntos
Giro Denteado/fisiopatologia , Memória/fisiologia , Canais de Potássio Shal/biossíntese , Potenciais de Ação , Doença de Alzheimer , Peptídeos beta-Amiloides/genética , Animais , Antioxidantes/farmacologia , Condicionamento Clássico/fisiologia , Giro Denteado/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Eletrochoque , Medo , Reação de Congelamento Cataléptica , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Estresse Oxidativo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Recombinantes/genética , Canais de Potássio Shal/genética
3.
Biomed Pharmacother ; 132: 110896, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33254430

RESUMO

OBJECTIVE: Previous study has shown that Kv4.3, a main coding subunit generating cardiac transient-outward K+ current (Ito), can inhibit Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity. Based on these observations, we speculate that over-expression of Kv4.3 gene could reverse not only Ito reduction but also cardiac remodeling in the rat myocardial infarction (MI) model. METHODS AND RESULTS: Healthy male Sprague-Dawley (SD) rats were used to establish MI model by ligation of left anterior descending coronary artery, and adenovirus integrated with Kv4.3 gene (AD-Kv4.3) was delivered in infarct border zone by intramyocardial injection. The hearts were harvested for histological analysis (HE or Masson trichrome staining), western blot or patch clamp 4 weeks after MI. Our data showed that the application of AD-Kv4.3 could reduce myocardial infarct size and fibrosis, and its cardioprotective effects were similar with medicine therapy (combination of metoprolol and captopril). Moreover, Kv4.3 over-expression significantly improved MI-induced cardiac dysfunction and enhanced Ito density while decreasing corrected QT (QTc) intervals and cardiac electrophysiological instability. Western blot showed that Kv4.3 transfection reduced CaMKII, PLB-17 and ryanodine receptor2 (RyR2 Ser2814) phosphorylation level, at same time increased SERCA2 expression dramatically. CONCLUSION: Over-expression of Kv4.3 can not only attenuate cardiac electrophysiological instability and cardiac performance, but also reduce myocardial infarct area and cardiac fibrosis. Like traditional anti-remodeling therapy-angiotensin converting enzyme inhibitor (ACEI) combined with ß-adrenergic receptor blocker, over-expression of Kv4.3 seems to be an effective and safe therapy for both structural and electrical remodeling induced by MI via CaMKII inhibition.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/antagonistas & inibidores , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Potássio Shal/biossíntese , Remodelação Ventricular/fisiologia , Animais , Antiarrítmicos/administração & dosagem , Anti-Hipertensivos/administração & dosagem , Expressão Gênica , Masculino , Infarto do Miocárdio/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Canais de Potássio Shal/genética , Remodelação Ventricular/efeitos dos fármacos
4.
Exp Neurol ; 334: 113437, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32822706

RESUMO

The voltage-gated potassium channel Kv4.2 is a critical regulator of dendritic excitability in the hippocampus and is crucial for dendritic signal integration. Kv4.2 mRNA and protein expression as well as function are reduced in several genetic and pharmacologically induced rodent models of epilepsy and autism. It is not known, however, whether reduced Kv4.2 is just an epiphenomenon or a disease-contributing cause of neuronal hyperexcitability and behavioral impairments in these neurological disorders. To address this question, we used male and female mice heterozygous for a Kv.2 deletion and adult-onset manipulation of hippocampal Kv4.2 expression in male mice to assess the role of Kv4.2 in regulating neuronal network excitability, morphology and anxiety-related behaviors. We observed a reduction in dendritic spine density and reduced proportions of thin and stubby spines but no changes in anxiety, overall activity, or retention of conditioned freezing memory in Kv4.2 heterozygous mice compared with wildtype littermates. Using EEG analyses, we showed elevated theta power and increased spike frequency in Kv4.2 heterozygous mice under basal conditions. In addition, the latency to onset of kainic acid-induced seizures was significantly shortened in Kv4.2 heterozygous mice compared with wildtype littermates, which was accompanied by a significant increase in theta power. By contrast, overexpressing Kv4.2 in wildtype mice through intrahippocampal injection of Kv4.2-expressing lentivirus delayed seizure onset and reduced EEG power. These results suggest that Kv4.2 is an important regulator of neuronal network excitability and dendritic spine morphology, but not anxiety-related behaviors. In the future, manipulation of Kv4.2 expression could be used to alter seizure susceptibility in epilepsy.


Assuntos
Espinhas Dendríticas/metabolismo , Eletroencefalografia/métodos , Hipocampo/metabolismo , Convulsões/metabolismo , Canais de Potássio Shal/biossíntese , Animais , Feminino , Predisposição Genética para Doença , Células HEK293 , Hipocampo/citologia , Humanos , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Convulsões/genética , Convulsões/fisiopatologia , Canais de Potássio Shal/genética
5.
J Biol Chem ; 295(34): 12099-12110, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32641494

RESUMO

Voltage-gated potassium (Kv) channels of the Kv4 subfamily associate with Kv channel-interacting proteins (KChIPs), which leads to enhanced surface expression and shapes the inactivation gating of these channels. KChIP3 has been reported to also interact with the late endosomal/lysosomal membrane glycoprotein CLN3 (ceroid lipofuscinosis neuronal 3), which is modified because of gene mutation in juvenile neuronal ceroid lipofuscinosis (JNCL). The present study was undertaken to find out whether and how CLN3, by its interaction with KChIP3, may indirectly modulate Kv4.2 channel expression and function. To this end, we expressed KChIP3 and CLN3, either individually or simultaneously, together with Kv4.2 in HEK 293 cells. We performed co-immunoprecipitation experiments and found a lower amount of KChIP3 bound to Kv4.2 in the presence of CLN3. In whole-cell patch-clamp experiments, we examined the effects of CLN3 co-expression on the KChIP3-mediated modulation of Kv4.2 channels. Simultaneous co-expression of CLN3 and KChIP3 with Kv4.2 resulted in a suppression of the typical KChIP3-mediated modulation; i.e. we observed less increase in current density, less slowing of macroscopic current decay, less acceleration of recovery from inactivation, and a less positively shifted voltage dependence of steady-state inactivation. The suppression of the KChIP3-mediated modulation of Kv4.2 channels was weaker for the JNCL-related missense mutant CLN3R334C and for a JNCL-related C-terminal deletion mutant (CLN3ΔC). Our data support the notion that CLN3 is involved in Kv4.2/KChIP3 somatodendritic A-type channel formation, trafficking, and function, a feature that may be lost in JNCL.


Assuntos
Regulação da Expressão Gênica/genética , Proteínas Interatuantes com Canais de Kv , Glicoproteínas de Membrana , Chaperonas Moleculares , Mutação de Sentido Incorreto , Lipofuscinoses Ceroides Neuronais , Proteínas Repressoras , Canais de Potássio Shal , Substituição de Aminoácidos , Células HEK293 , Humanos , Proteínas Interatuantes com Canais de Kv/genética , Proteínas Interatuantes com Canais de Kv/metabolismo , Lipofuscinoses Ceroides Neuronais/genética , Lipofuscinoses Ceroides Neuronais/metabolismo , Lipofuscinoses Ceroides Neuronais/patologia , Ligação Proteica , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Canais de Potássio Shal/biossíntese , Canais de Potássio Shal/genética
6.
J Neurosci ; 40(11): 2200-2214, 2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32047055

RESUMO

The dentate gyrus (DG) in the hippocampus may play key roles in remembering distinct episodes through pattern separation, which may be subserved by the sparse firing properties of granule cells (GCs) in the DG. Low intrinsic excitability is characteristic of mature GCs, but ion channel mechanisms are not fully understood. Here, we investigated ionic channel mechanisms for firing frequency regulation in hippocampal GCs using male and female mice, and identified Kv4.1 as a key player. Immunofluorescence analysis showed that Kv4.1 was preferentially expressed in the DG, and its expression level determined by Western blot analysis was higher at 8-week than 3-week-old mice, suggesting a developmental regulation of Kv4.1 expression. With respect to firing frequency, GCs are categorized into two distinctive groups: low-frequency (LF) and high-frequency (HF) firing GCs. Input resistance (Rin) of most LF-GCs is lower than 200 MΩ, suggesting that LF-GCs are fully mature GCs. Kv4.1 channel inhibition by intracellular perfusion of Kv4.1 antibody increased firing rates and gain of the input-output relationship selectively in LF-GCs with no significant effect on resting membrane potential and Rin, but had no effect in HF-GCs. Importantly, mature GCs from mice depleted of Kv4.1 transcripts in the DG showed increased firing frequency, and these mice showed an impairment in contextual discrimination task. Our findings suggest that Kv4.1 expression occurring at late stage of GC maturation is essential for low excitability of DG networks and thereby contributes to pattern separation.SIGNIFICANCE STATEMENT The sparse activity of dentate granule cells (GCs), which is essential for pattern separation, is supported by high inhibitory inputs and low intrinsic excitability of GCs. Low excitability of GCs is thought to be attributable to a high K+ conductance at resting membrane potentials, but this study identifies Kv4.1, a depolarization-activated K+ channel, as a key ion channel that regulates firing of GCs without affecting resting membrane potentials. Kv4.1 expression is developmentally regulated and Kv4.1 currents are detected only in mature GCs that show low-frequency firing, but not in less mature high-frequency firing GCs. Furthermore, mice depleted of Kv4.1 transcripts in the dentate gyrus show impaired pattern separation, suggesting that Kv4.1 is crucial for sparse coding and pattern separation.


Assuntos
Aprendizagem da Esquiva/fisiologia , Giro Denteado/citologia , Discriminação Psicológica/fisiologia , Neurônios/fisiologia , Canais de Potássio Shal/fisiologia , Potenciais de Ação , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/fisiologia , Condicionamento Clássico , Giro Denteado/fisiologia , Eletrochoque , Feminino , Reação de Congelamento Cataléptica/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Genes Reporter , Humanos , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/classificação , Técnicas de Patch-Clamp , Células Piramidais/fisiologia , Interferência de RNA , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Interferente Pequeno/farmacologia , Canais de Potássio Shal/biossíntese , Canais de Potássio Shal/genética , Organismos Livres de Patógenos Específicos
7.
Curr Neurovasc Res ; 14(4): 397-405, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29119925

RESUMO

BACKGROUND: Although the neuroprotective effect of sodium hydrosulfide (NaHS, a hydrogen sulfide donor) pretreatment has been revealed, the effect of NaHS post-conditioning remains largely unknown. OBJECTIVE: We aimed to investigate the neuroprotective effect of NaHS post-conditioning against transient Global Cerebral Ischemia (tGCI)-induced hippocampal CA1 injury and its underlying molecular mechanism. METHODS: A tGCI rat model was established using the four-vessel occlusion method for 15 min of ischemia. The survival of hippocampal neurons was determined by Nissl staining and NeuN immunostaining. Protein expression of potassium voltage-gated channel subfamily D member 2 (Kv4.2) and potassium channel interacting protein 3 (KChIP3) was assessed by Immunohistochemistry (IHC) and Western blot. RESULTS: Decreased concentrations (12 and 24 µmol/kg) of NaHS post-conditioning significantly increased the numbers of survival neurons and NeuN-positive neurons in the hippocampal CA1 region at 7 days post-tGCI (all P<0.05). NaHS post-conditioning (24 µmol/kg) at 12 and 24 hr posttGCI can achieve the best protective effect (both P<0.05). IHC data demonstrated that NaHS postconditioning (24 µmol/kg) markedly attenuated tGCI-induced down-regulation of Kv4.2 protein in the hippocampal CA1 region at 26 hr post-tGCI. Confocal images showed that Kv4.2 did not express in the neuronal nuclei but predominantly express in the neuronal dendrites. In addition, NaHS post-conditioning significantly up-regulated Kv4.2 and down-regulated KChIP3 in tGCI rats at 26 and 168 hr post- tGCI (all P<0.05). CONCLUSION: Decreased concentrations of NaHS post-conditioning at 12-24 hr post-tGCI effectively protected hippocampal CA1 neurons from tGCI-induced injury, which may be through regulating the expression of Kv4.2 and KChIP3.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Ataque Isquêmico Transitório/metabolismo , Proteínas Interatuantes com Canais de Kv/biossíntese , Neurônios/metabolismo , Canais de Potássio Shal/biossíntese , Sulfetos/uso terapêutico , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Ataque Isquêmico Transitório/tratamento farmacológico , Proteínas Interatuantes com Canais de Kv/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Wistar , Canais de Potássio Shal/agonistas , Sulfetos/farmacologia , Resultado do Tratamento
8.
Biomed Pharmacother ; 92: 196-206, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28544933

RESUMO

Prolongation of QT interval is possible with fluoroquinolones, yet the underlying contributing factors have not been elucidated. Two widely used fluoroquinolone drugs were at the focus of this study in rats with/without acute myocardial dysfunction (AMI) induced by isoproterenol. The effects of levofloxacin and ciprofloxacin on the cardiac mRNA expression of rat Kv4.3, Kv1.2 and Nav1.5 mRNAs were determined. Administration of the two antibiotics produced dose-dependent changes in ECG parameters that were more prominent in rats with AMI than healthy rats; this was accompanied by elevations in serum lactate dehydrogenase and creatine kinase-MB. Histopathological examination indicated some loss of striations, edema and fibrotic changes in rats with AMI; however the two antibiotics did not further exacerbate the cardiac histopathology. mRNA expression of the ion channels was altered in rats with AMI and healthy rats. In conclusion, long-term administration of levofloxacin and ciprofloxacin produced deleterious effects on the ECG pattern of rats with/without AMI. The effect was generally baseline-dependent and therefore, rats with AMI showed greater ECG disturbances and increases in cardiac enzymes. Taken together, these data make it advisable to monitor patients with a history of acute AMI requiring treatment with these antibiotics until data from human studies are available.


Assuntos
Cardiotoxinas/toxicidade , Ciprofloxacina/toxicidade , Canal de Potássio Kv1.2/biossíntese , Levofloxacino/toxicidade , Infarto do Miocárdio/fisiopatologia , Canal de Sódio Disparado por Voltagem NAV1.5/biossíntese , Canais de Potássio Shal/biossíntese , Animais , Antibacterianos/toxicidade , Relação Dose-Resposta a Droga , Eletrocardiografia/efeitos dos fármacos , Expressão Gênica , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Canal de Potássio Kv1.2/genética , Masculino , Infarto do Miocárdio/induzido quimicamente , Infarto do Miocárdio/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Ratos , Ratos Wistar , Canais de Potássio Shal/genética
9.
J Biol Chem ; 291(33): 17369-81, 2016 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-27307045

RESUMO

Neuritin is an important neurotrophin that regulates neural development, synaptic plasticity, and neuronal survival. Elucidating the downstream molecular signaling is important for potential therapeutic applications of neuritin in neuronal dysfunctions. We previously showed that neuritin up-regulates transient potassium outward current (IA) subunit Kv4.2 expression and increases IA densities, in part by activating the insulin receptor signaling pathway. Molecular mechanisms of neuritin-induced Kv4.2 expression remain elusive. Here, we report that the Ca(2+)/calcineurin (CaN)/nuclear factor of activated T-cells (NFAT) c4 axis is required for neuritin-induced Kv4.2 transcriptional expression and potentiation of IA densities in cerebellum granule neurons. We found that neuritin elevates intracellular Ca(2+) and increases Kv4.2 expression and IA densities; this effect was sensitive to CaN inhibition and was eliminated in Nfatc4(-/-) mice but not in Nfatc2(-/-) mice. Stimulation with neuritin significantly increased nuclear accumulation of NFATc4 in cerebellum granule cells and HeLa cells, which expressed IR. Furthermore, NFATc4 was recruited to the Kv4.2 gene promoter loci detected by luciferase reporter and chromatin immunoprecipitation assays. More importantly, data obtained from cortical neurons following adeno-associated virus-mediated overexpression of neuritin indicated that reduced neuronal excitability and increased formation of dendritic spines were abrogated in the Nfatc4(-/-) mice. Together, these data demonstrate an indispensable role for the CaN/NFATc4 signaling pathway in neuritin-regulated neuronal functions.


Assuntos
Calcineurina/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Espinhas Dendríticas/metabolismo , Regulação da Expressão Gênica/fisiologia , Fatores de Transcrição NFATC/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neuropeptídeos/metabolismo , Canais de Potássio Shal/biossíntese , Animais , Calcineurina/genética , Cerebelo/metabolismo , Espinhas Dendríticas/genética , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Fatores de Transcrição NFATC/genética , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/genética , Canais de Potássio Shal/genética
10.
Alcohol Clin Exp Res ; 40(6): 1251-61, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27147118

RESUMO

BACKGROUND: Exposure to chronic ethanol (EtOH) results in changes in the expression of proteins that regulate neuronal excitability. This study examined whether chronic EtOH alters the hippocampal expression and function of fragile X mental retardation protein (FMRP) and the role of FMRP in the modulation of chronic EtOH-induced changes in the expression of NMDA receptors and Kv4.2 channels. METHODS: For in vivo studies, C57BL/6J mice underwent a chronic intermittent EtOH (CIE) vapor exposure procedure. After CIE, hippocampal tissue was collected and subjected to immunoblot blot analysis of NMDA receptor subunits (GluN1, GluN2B), Kv4.2, and its accessory protein KChIP3. For in vitro studies, hippocampal slice cultures were exposed to 75 mM EtOH for 8 days. Following EtOH exposure, mRNAs bound to FMRP was measured. In a separate set of studies, cultures were exposed to an inhibitor of S6K1 (PF-4708671 [PF], 6 µM) in order to assess whether EtOH-induced homeostatic changes in protein expression depend upon changes in FMRP activity. RESULTS: Immunoblot blot analysis revealed increases in GluN1 and GluN2B but reductions in Kv4.2 and KChIP3. Analysis of mRNAs bound to FMRP revealed a similar bidirectional change observed as reduction of GluN2B and increase in Kv4.2 and KChIP3 mRNA transcripts. Analysis of FMRP further revealed that while chronic EtOH did not alter the expression of FMRP, it significantly increased phosphorylation of FMRP at the S499 residue that is known to critically regulate its activity. Inhibition of S6K1 prevented the chronic EtOH-induced increase in phospho-FMRP and changes in NMDA subunits, Kv4.2, and KChIP3. In contrast, PF had no effect in the absence of alcohol, indicating it was specific for the chronic EtOH-induced changes. CONCLUSIONS: These findings demonstrate that chronic EtOH exposure enhances translational control of plasticity-related proteins by FMRP, and that S6K1 and FMRP activities are required for expression of chronic EtOH-induced homeostatic plasticity at glutamatergic synapses in the hippocampus.


Assuntos
Etanol/farmacologia , Proteína do X Frágil da Deficiência Intelectual/fisiologia , Hipocampo/metabolismo , Proteínas Interatuantes com Canais de Kv/biossíntese , Receptores de N-Metil-D-Aspartato/biossíntese , Canais de Potássio Shal/biossíntese , Administração por Inalação , Animais , Etanol/administração & dosagem , Etanol/antagonistas & inibidores , Proteína do X Frágil da Deficiência Intelectual/efeitos dos fármacos , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Imidazóis/farmacologia , Masculino , Camundongos , Fosforilação/efeitos dos fármacos , Piperazinas/farmacologia , Ratos , Proteínas Quinases S6 Ribossômicas 90-kDa/antagonistas & inibidores
11.
J Mol Cell Cardiol ; 86: 85-94, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26205295

RESUMO

BACKGROUND AND OBJECTIVE: We have previously shown that androgens upregulate cardiac K(+) channels and shorten repolarization. However, the effects that estrogens (E2) and estrogen receptors (ER) might have on the various repolarizing K(+) currents and underlying ion channels remain incompletely understood. Accordingly, our objective was to verify whether and how E2 and its ERs subtypes influence these K(+) currents. METHODS AND RESULTS: In order to examine the influence of E2 and ERs on K(+) currents we drastically lowered the E2 level through ovariectomy (OVX; 74% reduction vs CTL) and in parallel, we used female mice lacking either ERα (ERαKO) or ERß (ERßKO). In OVX mice, results showed a specific increase of 35% in the density of the Ca(2+)-independent transient outward K(+) current (Ito) compared to CTL. Western blots showed increase in Kv4.2 and Kv4.3 sarcolemmal protein expression while qPCR revealed higher mRNA expression of only Kv4.3 in OVX mice. This upregulation of Ito was correlated with a shorter ventricular action potential duration and QTc interval. In ERαKO but not ERßKO mice, the mRNA of Kv4.3 was selectively increased. Furthermore, when ventricular myocytes obtained from ERαKO and ERßKO were cultured in the presence of E2, results showed that E2 reduced Ito density only in ERßKO myocytes confirming the repressive role of E2-ERα in regulating Ito. CONCLUSION: Altogether, these results suggest that E2 negatively regulates the density of Ito through ERα, this highlights a potential role for this female hormone and its α-subtype receptor in modulating cardiac electrical activity.


Assuntos
Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Canais de Potássio Shal/biossíntese , Potenciais de Ação , Animais , Cálcio/metabolismo , Receptor beta de Estrogênio/genética , Estrogênios/genética , Feminino , Ventrículos do Coração/patologia , Humanos , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Ovariectomia , Técnicas de Patch-Clamp , RNA Mensageiro/biossíntese , Canais de Potássio Shal/genética
12.
Int J Mol Med ; 36(1): 309-15, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26016905

RESUMO

The human cardiac fast transient outward K+ channel is composed of the KV4.3 α subunit encoded by KCND3 and the K+ channel­interacting protein 2 (KChIP2) ß subunit, and determines the early repolarization of the action potential (AP). Two human mutations (G600R and L450F) in KV4.3 are associated with Brugada syndrome and they increase the KV4.3/KChIP2­encoded fast transient outward K+ current (Ito,f) and cause the stable loss of the AP dome. However, the detailed mechanisms underlying the gain of Ito,f function by these two mutations are largely unknown. The experiments in the present study were undertaken to investigate the effect of these mutations and the underlying mechanism. Whole cell patch­clamp recording was performed in HEK­293 cells expressing KV4.3­wild­type (WT) and KV4.3 mutants with KChIP2. The two individual mutant­encoded currents were significantly increased but the kinetics of the channels affected by the two mutations were different. The two mutations slowed KV4.3/KChIP2­encoded channel inactivation; they did not increase the recovery from the KV4.3/KChIP2­encoded channel inactivation. Western blotting showed that total KV4.3 protein was significantly augmented in HEK­293 cells expressing the two individual mutants with KChIP2. Furthermore, immunofluorescence confocal microscopy demonstrated that the KV4.3 channel protein was expressed more in the cell membrane compared to the cytoplasm in cells that expressed individual mutants with KChIP2. Also, KChIP2 increased the amount of channel protein in the cell membrane of KV4.3 mutants significantly more than KV4.3­WT. Reverse transcription­polymerase chain reaction showed that KV4.3 mRNA was not significantly changed by individual mutations in the presence of KChIP2. Taken together, the present study revealed that the mutations cause a gain­of­function of KV4.3/KChIP2­encoded channels by increasing membrane protein expression and slowing channel inactivation.


Assuntos
Síndrome de Brugada/genética , Proteínas Interatuantes com Canais de Kv/genética , Potenciais da Membrana/fisiologia , Canais de Potássio Shal/genética , Linhagem Celular , Membrana Celular/fisiologia , Células HEK293 , Coração/fisiopatologia , Humanos , Mutação/genética , Miocárdio/metabolismo , Técnicas de Patch-Clamp , RNA Mensageiro/genética , Canais de Potássio Shal/biossíntese , Canais de Potássio Shal/metabolismo
14.
Circ J ; 78(8): 1950-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24859499

RESUMO

BACKGROUND: This study was designed to investigate the role of a primary hyperoxidative stress in myocardial electrical remodeling using heterozygous heart/muscle-specific manganese superoxide dismutase-deficient (H/M-Sod2(+/-)) mice treated with L-buthionine-sulfoximine (BSO). METHODS AND RESULTS: Both H/M-Sod2(+/-)and wild-type (WT) mice were treated with intra-peritoneal BSO or saline for 7 days, and divided into 4 groups: H/M-Sod2(+/-)+BSO, WT+BSO, H/M-Sod2(+/-)control, and WT control. The ventricular effective refractory period (ERP) and the monophasic action potential duration (MAPD) were determined. Levels of oxidative stress, potassium channel-related molecules, and K(+)channel-interacting protein-2 (KChIP2) were also evaluated. The H/M-Sod2(+/-)+BSO group exhibited markedly prolonged MAPD20, MAPD90 and ERP in comparison with the other groups (MAPD20: 14 ± 1 vs. 11 ± 1 ms, MAPD90: 77 ± 7 vs. 58 ± 4 ms, ERP: 61 ± 6 vs. 41 ± 3 ms, H/M-Sod2(+/-)+BSO vs. WT control; P<0.05). Mitochondrial superoxide and hydrogen peroxide formation in the myocardium increased in the H/M-Sod2(+/-)+BSO group in comparison with the WT+BSO group (P<0.05). Real-time RT-PCR and Western blotting revealed that Kv4.2 expression was downregulated in both BSO-treated groups, whereas KChIP2 expression was downregulated only in the H/M-Sod2(+/-)+BSO group (P<0.05). CONCLUSIONS: BSO treatment caused hyperoxidative stress in the myocardium of H/M-Sod2(+/-)mice. Changes in the expression and function of potassium channels were considered to be involved in the mechanism of electrical remodeling in this model.


Assuntos
Regulação para Baixo , Proteínas Interatuantes com Canais de Kv/biossíntese , Mitocôndrias Cardíacas/metabolismo , Miocárdio/metabolismo , Estresse Oxidativo , Canais de Potássio Shal/biossíntese , Superóxidos/metabolismo , Animais , Antimetabólitos/farmacologia , Butionina Sulfoximina/farmacologia , Peróxido de Hidrogênio/metabolismo , Proteínas Interatuantes com Canais de Kv/genética , Camundongos , Camundongos Knockout , Miócitos Cardíacos , Canais de Potássio Shal/genética , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
15.
Neuron ; 82(3): 537-44, 2014 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24811378

RESUMO

Ion channel gene expression can vary substantially among neurons of a given type, even though neuron-type-specific firing properties remain stable and reproducible. The mechanisms that modulate ion channel gene expression and stabilize neural firing properties are unknown. In Drosophila, we demonstrate that loss of the Shal potassium channel induces the compensatory rebalancing of ion channel expression including, but not limited to, the enhanced expression and function of Shaker and slowpoke. Using genomic and network modeling approaches combined with genetic and electrophysiological assays, we demonstrate that the transcription factor Krüppel is necessary for the homeostatic modulation of Shaker and slowpoke expression. Remarkably, Krüppel induction is specific to the loss of Shal, not being observed in five other potassium channel mutants that cause enhanced neuronal excitability. Thus, homeostatic signaling systems responsible for rebalancing ion channel expression can be selectively induced after the loss or impairment of a specific ion channel.


Assuntos
Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/fisiologia , Ativação do Canal Iônico/fisiologia , Canais Iônicos/biossíntese , Fatores de Transcrição Kruppel-Like/fisiologia , Canais de Potássio Shal/biossíntese , Animais , Animais Geneticamente Modificados , Drosophila , Proteínas de Drosophila/genética , Ativação do Canal Iônico/genética , Canais Iônicos/genética , Fatores de Transcrição Kruppel-Like/genética , Mutação/genética , Canais de Potássio Shal/genética
16.
J Cardiothorac Surg ; 8: 194, 2013 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-24074263

RESUMO

BACKGROUND: Recent studies have demonstrated that atrial electrical remodeling was an important contributing factor for the occurrence, persistence and maintenance of atrial fibrillation. The expression changes of ionic channels, especially L-type calcium channel and potassium channel Kv4.3, were the important molecular mechanism of atrial electrical remodeling. This study aimed to observe the expression changes of ionic channels in a rapid paced cell model with primary cultured atrial myocytes. METHODS: The primary rat atrial myocytes were cultured, characteristics of the cultured myocytes were observed with light microscope and the cell phenotype was harvested by immunocytochemical stain to detect α-actin. The cellular model of rapid pacing was established with primary cultured atrial myocytes. The expressions of L-type calcium channel α1c and potassium channel Kv4.3 in cultured atrial myocytes were detected by immunocytochemistry, reverse transcription polymerase chain reaction and Western blot after rapid pacing. RESULTS: The primary rat atrial myocytes were isolated and cultured successfully, and used for following experiment by identification of activity and purity. Cellular model of rapid electrical field pacing was established successfully. There is no significant difference in cell activity after pacing compared to that before pacing by 3-[4, 5-dimethylthiazol-2-y1]-2, 5-diphenytetrazolium bromide assay, and cell degeneration can be observed by transmission electron microscope. The mRNA expression of L-type calcium channel α1c started to reduce after 6 h of rapid pacing and continued to decline as pacing continued. Protein expression changes were paralleled with decreased mRNA expression of the L-type calcium channel α1c. The mRNA expressions of potassium channel Kv4.3 were not altered within the first 6 h, but after 12 h, mRNA expressions were reduced. Longer pacing periods did not further decrease mRNA expression of potassium channel Kv4.3. Protein expression changes were paralleled with decreased mRNA expression of potassium channel Kv4.3. CONCLUSIONS: Rapid paced cultured atrial myocyte model was established utilized primary cultured atrial myocytes and this model can be used for studying the early electrical remodeling in atrial fibrillation. Expressions of L-type calcium channel α1c and potassium channel Kv4.3 were both reduced at different levels in early phase of rapid pacing atrial myocytes. It implicates the occurrence of ionic channel remodeling of atrial myocytes.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Estimulação Cardíaca Artificial/métodos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Canais de Potássio Shal/biossíntese , Actinas/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Células Cultivadas , Estimulação Elétrica , Átrios do Coração/citologia , Átrios do Coração/metabolismo , Imuno-Histoquímica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Canais de Potássio Shal/genética
17.
J Biol Chem ; 287(21): 17656-17661, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22511771

RESUMO

The dorsal and ventral regions of the hippocampus perform different functions. Whether the integrative properties of hippocampal cells reflect this heterogeneity is unknown. We focused on dendrites where most synaptic input integration takes place. We report enhanced backpropagation and theta resonance and decreased summation of synaptic inputs in ventral versus dorsal CA1 pyramidal cell distal dendrites. Transcriptional Kv4.2 down-regulation and post-transcriptional hyperpolarization-activated cyclic AMP-gated channel (HCN1/2) up-regulation may underlie these differences, respectively. Our results reveal differential dendritic integrative properties along the dorso-ventral axis, reflecting diverse computational needs.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/biossíntese , Dendritos/metabolismo , Regulação para Baixo/fisiologia , Canais Iônicos/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Canais de Potássio/biossíntese , Células Piramidais/metabolismo , Canais de Potássio Shal/biossíntese , Regulação para Cima/fisiologia , Animais , Dendritos/genética , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Especificidade de Órgãos , Células Piramidais/citologia , Ratos , Transcrição Gênica/fisiologia
18.
J Neurosci ; 31(15): 5693-8, 2011 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-21490210

RESUMO

A prominent characteristic of the inherited intellectual impairment disease fragile X syndrome (FXS) is neuronal hyperexcitability, resulting in a variety of symptoms, such as hyperactivity, increased sensitivity to sensory stimuli, and a high incidence of epileptic seizures. These symptoms account for a significant part of the disease pattern, but the underlying molecular mechanisms of neuronal hyperexcitability in FXS remain poorly understood. FXS is caused by loss of expression of fragile X mental retardation protein (FMRP), which regulates synaptic protein synthesis and is a key player to limit signaling pathways downstream of metabotropic glutamate receptors 1/5 (mGlu1/5). Recent findings suggest that FMRP might also directly regulate voltage-gated potassium channels. Here, we show that total and plasma membrane protein levels of Kv4.2, the major potassium channel regulating hippocampal neuronal excitability, are reduced in the brain of an FXS mouse model. Antagonizing mGlu5 activity with 2-methyl-6-(phenylethynyl)-pyridine (MPEP) partially rescues reduced surface Kv4.2 levels in Fmr1 knock-out (KO) mice, suggesting that excess mGlu1/5 signal activity contributes to Kv4.2 dysregulation. As an additional mechanism, we show that FMRP is a positive regulator of Kv4.2 mRNA translation and protein expression and associates with Kv4.2 mRNA in vivo and in vitro. Our results suggest that absence of FMRP-mediated positive control of Kv4.2 mRNA translation, protein expression, and plasma membrane levels might contribute to excess neuronal excitability in Fmr1 KO mice, and thus imply a potential mechanism underlying FXS-associated epilepsy.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/fisiologia , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Canais de Potássio Shal/biossíntese , Canais de Potássio Shal/genética , Regiões 3' não Traduzidas/genética , Regiões 5' não Traduzidas/genética , Actinas/biossíntese , Actinas/genética , Animais , Biotinilação , Western Blotting , Primers do DNA , Dendritos/metabolismo , Imunofluorescência , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Polirribossomos/genética , Polirribossomos/metabolismo , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/metabolismo , Convulsões/fisiopatologia , Sinapses/metabolismo
19.
Biochem Biophys Res Commun ; 404(2): 678-83, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21147063

RESUMO

Normal vision depends on the correct function of retinal neurons and glia and it is impaired in the course of diabetic retinopathy. Müller cells, the main glial cells of the retina, suffer morphological and functional alterations during diabetes participating in the pathological retinal dysfunction. Recently, we showed that Müller cells express the pleiotropic protein potassium channel interacting protein 3 (KChIP3), an integral component of the voltage-gated K(+) channels K(V)4. Here, we sought to analyze the role of KChIP3 in the molecular mechanisms underlying hyperglycemia-induced phenotypic changes in the glial elements of the retina. The expression and function of KChIp3 was analyzed in vitro in rat Müller primary cultures grown under control (5.6 mM) or high glucose (25 mM) (diabetic-like) conditions. We show the up-regulation of KChIP3 expression in Müller cell cultures under high glucose conditions and demonstrate a previously unknown interaction between the K(V)4 channel and KChIP3 in Müller cells. We show evidence for the expression of a 4-AP-sensitive transient outward voltage-gated K(+) current and an alteration in the inactivation of the macroscopic outward K(+) currents expressed in high glucose-cultured Müller cells. Our data support the notion that induction of KChIP3 and functional changes of K(V)4 channels in Müller cells could exert a physiological role in the onset of diabetic retinopathy.


Assuntos
Glucose/metabolismo , Hiperglicemia/metabolismo , Proteínas Interatuantes com Canais de Kv/biossíntese , Neuroglia/metabolismo , Proteínas Repressoras/biossíntese , Retina/metabolismo , Canais de Potássio Shal/biossíntese , Animais , Células Cultivadas , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Glucose/fisiologia , Hiperglicemia/patologia , Proteínas Interatuantes com Canais de Kv/metabolismo , Proteínas Interatuantes com Canais de Kv/fisiologia , Neuroglia/efeitos dos fármacos , Neuroglia/patologia , Ratos , Ratos Long-Evans , Retina/efeitos dos fármacos , Retina/patologia , Canais de Potássio Shal/fisiologia
20.
Neuroscience ; 171(3): 721-33, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20849929

RESUMO

The Kv4 potassium channel α subunits, Kv4.1, Kv4.2, and Kv4.3, determine some of the fundamental physiological properties of neurons in the CNS. Kv4 subunits are associated with auxiliary ß-subunits, such as the potassium channel interacting proteins (KChIP1 - 4), which are thought to regulate the trafficking and gating of native Kv4 potassium channels. Intriguingly, KChIP1 is thought to show cell type-selective expression in GABA-ergic inhibitory interneurons, while other ß-subunits (KChIP2-4) are associated with principal glutamatergic neurons. However, nothing is known about the expression of Kv4 family α- and ß-subunits in specific interneurons populations in the BLA. Here, we have used immunofluorescence, co-immunoprecipitation, and Western Blotting to determine the relative expression of KChIP1 in the different interneuron subtypes within the BLA, and its co-localization with one or more of the Kv4 α subunits. We show that all three α-subunits of Kv4 potassium channel are found in rat BLA neurons, and that the immunoreactivity of KChIP1 closely resembles that of Kv4.3. Indeed, Kv4.3 showed almost complete co-localization with KChIP1 in the soma and dendrites of a distinct subpopulation of BLA neurons. Dual-immunofluorescence studies revealed this to be in BLA interneurons immunoreactive for parvalbumin, cholecystokin-8, and somatostatin. Finally, co-immunoprecipitation studies showed that KChIP1 was associated with all three Kv4 α subunits. Together our results suggest that KChIP1 is selectively expressed in BLA interneurons where it may function to regulate the activity of A-type potassium channels. Hence, KChIP1 might be considered as a cell type-specific regulator of GABAergic inhibitory circuits in the BLA.


Assuntos
Tonsila do Cerebelo/metabolismo , Interneurônios/metabolismo , Proteínas Interatuantes com Canais de Kv/biossíntese , Subunidades Proteicas/biossíntese , Canais de Potássio Shal/biossíntese , Tonsila do Cerebelo/citologia , Animais , Interneurônios/citologia , Proteínas Interatuantes com Canais de Kv/genética , Proteínas Interatuantes com Canais de Kv/metabolismo , Masculino , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Ratos , Ratos Sprague-Dawley , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA