Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
J Neurosci ; 40(11): 2200-2214, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32047055

RESUMEN

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.


Asunto(s)
Reacción de Prevención/fisiología , Giro Dentado/citología , Discriminación en Psicología/fisiología , Neuronas/fisiología , Canales de Potasio Shal/fisiología , Potenciales de Acción , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/fisiología , Condicionamiento Clásico , Giro Dentado/fisiología , Electrochoque , Femenino , Reacción Cataléptica de Congelación/fisiología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Genes Reporteros , Humanos , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Neuronas/clasificación , Técnicas de Placa-Clamp , Células Piramidales/fisiología , Interferencia de ARN , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Canales de Potasio Shal/biosíntesis , Canales de Potasio Shal/genética , Organismos Libres de Patógenos Específicos
2.
J Neurosci ; 39(4): 596-611, 2019 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-30504282

RESUMEN

Neurons operate within defined activity limits, and feedback control mechanisms dynamically tune ionic currents to maintain this optimal range. This study describes a novel, rapid feedback mechanism that uses SUMOylation to continuously adjust ionic current densities according to changes in activity. Small ubiquitin-like modifier (SUMO) is a peptide that can be post-translationally conjugated to ion channels to influence their surface expression and biophysical properties. Neuronal activity can regulate the extent of protein SUMOylation. This study on the single, unambiguously identifiable lateral pyloric neuron (LP), a component of the pyloric network in the stomatogastric nervous system of male and female spiny lobsters (Panulirus interruptus), focused on dynamic SUMOylation in the context of activity homeostasis. There were four major findings: First, neuronal activity adjusted the balance between SUMO conjugation and deconjugation to continuously and bidirectionally fine-tune the densities of two opposing conductances: the hyperpolarization activated current (Ih) and the transient potassium current (IA). Second, tonic 5 nm dopamine (DA) gated activity-dependent SUMOylation to permit and prevent activity-dependent regulation of Ih and IA, respectively. Third, DA-gated, activity-dependent SUMOylation contributed to a feedback mechanism that restored the timing and duration of LP activity during prolonged modulation by 5 µm DA, which initially altered these and other activity features. Fourth, DA modulatory and metamoduatory (gating) effects were tailored to simultaneously alter and stabilize neuronal output. Our findings suggest that modulatory tone may select a subset of rapid activity-dependent mechanisms from a larger menu to achieve homeostasis under varying conditions.SIGNIFICANCE STATEMENT Post-translational SUMOylation of ion channel subunits controls their interactions. When subunit SUMOylation is dysregulated, conductance densities mediated by the channels are distorted, leading to nervous system disorders, such as seizures and chronic pain. Regulation of ion channel SUMOylation is poorly understood. This study demonstrated that neuronal activity can regulate SUMOylation to reconfigure ionic current densities over minutes, and this regulation was gated by tonic nanomolar dopamine. Dynamic SUMOylation was necessary to maintain specific aspects of neuronal output while the neuron was being modulated by high (5 µm) concentrations of dopamine, suggesting that the gating function may ensure neuronal homeostasis during extrinsic modulation of a circuit.


Asunto(s)
Homeostasis/fisiología , Canales Iónicos/fisiología , Palinuridae/fisiología , Sumoilación/fisiología , Animales , Dopamina/fisiología , Femenino , Ganglios de Invertebrados/fisiología , Células HEK293 , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Activación del Canal Iónico/fisiología , Masculino , Potenciales de la Membrana/fisiología , Neuronas/metabolismo , Procesamiento Proteico-Postraduccional , Canales de Potasio Shal/fisiología
3.
J Neurosci ; 38(42): 9059-9071, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30185460

RESUMEN

Sleep is highly conserved across animal species. Both wake- and sleep-promoting neurons are implicated in the regulation of wake-sleep transition at dusk in Drosophila However, little is known about how they cooperate and whether they act via different mechanisms. Here, we demonstrated that in female Drosophila, sleep onset was specifically delayed by blocking the Shaker cognate L channels [Shal; also known as voltage-gated K+ channel 4 (Kv4)] in wake-promoting cells, including large ventral lateral neurons (l-LNvs) and pars intercerebralis (PI), but not in sleep-promoting dorsal neurons (DN1s). Delayed sleep onset was also observed in males by blocking Kv4 activity in wake-promoting neurons. Electrophysiological recordings show that Kv4 channels contribute A-type currents in LNvs and PI cells, but are much less conspicuous in DN1s. Interestingly, blocking Kv4 in wake-promoting neurons preferentially increased firing rates at dusk ∼ZT13, when the resting membrane potentials and firing rates were at lower levels. Furthermore, pigment-dispersing factor (PDF) is essential for the regulation of sleep onset by Kv4 in l-LNvs, and downregulation of PDF receptor (PDFR) in PI neurons advanced sleep onset, indicating Kv4 controls sleep onset via regulating PDF/PDFR signaling in wake-promoting neurons. We propose that Kv4 acts as a sleep onset controller by suppressing membrane excitability in a clock-dependent manner to balance the wake-sleep transition at dusk. Our results have important implications for the understanding and treatment of sleep disorders such as insomnia.SIGNIFICANCE STATEMENT The mechanisms by which our brains reversibly switch from waking to sleep state remain an unanswered and intriguing question in biological research. In this study, we identified that Shal/Kv4, a well known voltage-gated K+ channel, acts as a controller of wake-sleep transition at dusk in Drosophila circadian neurons. We find that interference of Kv4 function with a dominant-negative form (DNKv4) in subsets of circadian neurons specifically disrupts sleep onset at dusk, although Kv4 itself does not exhibit circadian oscillation. Kv4 preferentially downregulates neuronal firings at ZT9-ZT17, supporting that it plays an essential role in wake-sleep transition at dusk. Our findings may help understand and eventually treat sleep disorders such as insomnia.


Asunto(s)
Encéfalo/fisiología , Ritmo Circadiano , Proteínas de Drosophila/fisiología , Neuronas/fisiología , Canales de Potasio Shal/fisiología , Sueño , Animales , Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Femenino , Masculino , ARN Mensajero/metabolismo , Canales de Potasio Shal/metabolismo
4.
J Physiol ; 597(23): 5707-5722, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31612994

RESUMEN

As in mammals, Drosophila circadian clock neurons display rhythms of activity with higher action potential firing rates and more positive resting membrane potentials during the day. This rhythmic excitability has been widely observed but, critically, its regulation remains unresolved. We have characterized and modelled the changes underlying these electrical activity rhythms in the lateral ventral clock neurons (LNvs). We show that currents mediated by the voltage-gated potassium channels Shaw (Kv3) and Shal (Kv4) oscillate in a circadian manner. Disruption of these channels, by expression of dominant negative (DN) subunits, leads to changes in circadian locomotor activity and shortens lifespan. LNv whole-cell recordings then show that changes in Shaw and Shal currents drive changes in action potential firing rate and that these rhythms are abolished when the circadian molecular clock is stopped. A whole-cell biophysical model using Hodgkin-Huxley equations can recapitulate these changes in electrical activity. Based on this model and by using dynamic clamp to manipulate clock neurons directly, we can rescue the pharmacological block of Shaw and Shal, restore the firing rhythm, and thus demonstrate the critical importance of Shaw and Shal. Together, these findings point to a key role for Shaw and Shal in controlling circadian firing of clock neurons and show that changes in clock neuron currents can account for this. Moreover, with dynamic clamp we can switch the LNvs between morning-like and evening-like states of electrical activity. We conclude that changes in Shaw and Shal underlie the daily oscillation in LNv firing rate.


Asunto(s)
Relojes Circadianos/fisiología , Proteínas de Drosophila/fisiología , Neuronas/fisiología , Canales de Potasio Shal/fisiología , Canales de Potasio Shaw/fisiología , Animales , Ritmo Circadiano , Drosophila , Femenino , Locomoción , Masculino , Modelos Biológicos
5.
J Pharmacol Exp Ther ; 363(2): 184-195, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28864468

RESUMEN

Voltage clamp recordings of macroscopic currents were made from rat potassium-gated potassium 4.2(Kv4.2) channels expressed in human embryonic kidney (HEK293) cells with the main goals of quantifying the concentration, time, and voltage dependence of the block and to generate a state model that replicates the features of the block. When applied either externally or internally, the block of Kv4.2 currents by 4-aminopyridine (4AP) occurs at the holding potential (-80 mV), is affected by the stimulus frequency, and is relieved by membrane depolarization. The Kd for the tonic block at -80 mV was 0.9 ± 0.07 mM and was consistent with 1:1 binding. Relief of block during a step to 50 mV was well fitted by a single exponential with a time constant of ∼40 milliseconds. At -80 mV, the association rate constant was 0.08 mM-1 s-1, and the off-rate was 0.08 s-1 The state model replicates the features of the experimental data reasonably well by assuming that 4AP binds only to closed states, that 4AP binding and inactivation are mutually exclusive processes, and that the activation of closed-bound channels is the same as for closed channels. Since the open channel has a very low or no affinity for 4AP, channel opening promotes the unbinding of 4AP, which accounts for the reverse use dependence of the block.


Asunto(s)
4-Aminopiridina/farmacología , Modelos Biológicos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Shal/antagonistas & inhibidores , Canales de Potasio Shal/fisiología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Xenopus
6.
Cell Physiol Biochem ; 39(1): 102-14, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27322747

RESUMEN

BACKGROUND/AIMS: Acute myocardial infarction (AMI) is a devastating cardiovascular disease with a high rate of morbidity and mortality, partly due to enhanced arrhythmogenicity. MicroRNAs (miRNAs) have been shown to participate in the regulation of cardiac ion channels and the associated arrhythmias. The purpose of this study was to test our hypothesis that miR-223-3p contributes to the electrical disorders in AMI via modulating KCND2, the gene encoding voltage-gated channel Kv4.2 that carries transient outward K+ current Ito. METHODS: AMI model was established in male Sprague-Dawley (SD) rats by left anterior descending artery (LAD) ligation. Evans blue and TTC staining was used to measure infarct area. Ito was recorded in isolated ventricular cardiomyocytes or cultured neonatal rat ventricular cells (NRVCs) by whole-cell patch-clamp techniques. Western blot analysis was employed to detect the protein level of Kv4.2 and real-time RT-PCR to determine the transcript level of miR-223-3p. Luciferase assay was used to examine the interaction between miR-223-3p and KCND2 in cultured NRVCs. RESULTS: Expression of miR-223-3p was remarkably upregulated in AMI relative to sham control rats. On the contrary, the protein level of Kv4.2 and Ito density were significantly decreased in AMI. Consistently, transfection of miR-223-3p mimic markedly reduced Kv4.2 protein level and Ito current in cultured NRVCs. Co-transfection of AMO-223-3p (an antisense inhibitor of miR-223-3p) reversed the repressive effect of miR-223-3p. Luciferase assay showed that miR-223-3p, but not the negative control, substantially suppressed the luciferase activity, confirming the direct binding of miR-223-3p to the seed site within the KCND2 sequence. Finally, direct intramuscular injection of AMO-223-3p into the ischemic myocardium to knockdown endogenous miR-223-3p decreased the propensity of ischemic arrhythmias. CONCLUSIONS: Upregulation of miR-223-3p in AMI repressed the expression of KCND2/Kv4.2 resulting in reduction of Ito density that can cause APD prolongation and promote arrhythmias in AMI, and therefore knockdown of endogenous miR-223-3p might be considered a new approach for antiarrhythmic therapy of ischemic arrhythmias.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , Infarto del Miocardio/genética , Canales de Potasio Shal/genética , Animales , Animales Recién Nacidos , Western Blotting , Células Cultivadas , Activación del Canal Iónico/genética , Activación del Canal Iónico/fisiología , Masculino , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Potasio Shal/metabolismo , Canales de Potasio Shal/fisiología
7.
J Neurosci ; 34(41): 13586-99, 2014 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-25297088

RESUMEN

Parkinson disease (PD) is an α-synucleinopathy resulting in the preferential loss of highly vulnerable dopamine (DA) substantia nigra (SN) neurons. Mutations (e.g., A53T) in the α-synuclein gene (SNCA) are sufficient to cause PD, but the mechanism of their selective action on vulnerable DA SN neurons is unknown. In a mouse model overexpressing mutant α-synuclein (A53T-SNCA), we identified a SN-selective increase of in vivo firing frequencies in DA midbrain neurons, which was not observed in DA neurons in the ventral tegmental area. The selective and age-dependent gain-of-function phenotype of A53T-SCNA overexpressing DA SN neurons was in part mediated by an increase of their intrinsic pacemaker frequency caused by a redox-dependent impairment of A-type Kv4.3 potassium channels. This selective enhancement of "stressful pacemaking" of DA SN neurons in vivo defines a functional response to mutant α-synuclein that might be useful as a novel biomarker for the "DA system at risk" before the onset of neurodegeneration in PD.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Mutación/fisiología , Estrés Oxidativo/fisiología , Canales de Potasio Shal/fisiología , Sustancia Negra/fisiología , alfa-Sinucleína/genética , Envejecimiento/fisiología , Animales , Fenómenos Electrofisiológicos , Glutatión/metabolismo , Glutatión/fisiología , Activación del Canal Iónico/fisiología , Masculino , Ratones , Mutación/genética , Sustancia Negra/citología , Sustancia Negra/crecimiento & desarrollo , Área Tegmental Ventral/crecimiento & desarrollo , Área Tegmental Ventral/fisiología
8.
J Neurosci ; 34(28): 9182-9, 2014 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-25009251

RESUMEN

SNX-482, a peptide toxin isolated from tarantula venom, has become widely used as an inhibitor of Cav2.3 voltage-gated calcium channels. Unexpectedly, we found that SNX-482 dramatically reduced the A-type potassium current in acutely dissociated dopamine neurons from mouse substantia nigra pars compacta. The inhibition persisted when calcium was replaced by cobalt, showing that it was not secondary to a reduction of calcium influx. Currents from cloned Kv4.3 channels expressed in HEK-293 cells were inhibited by SNX-482 with an IC50 of <3 nM, revealing substantially greater potency than for SNX-482 inhibition of Cav2.3 channels (IC50 20-60 nM). At sub-saturating concentrations, SNX-482 produced a depolarizing shift in the voltage dependence of activation of Kv4.3 channels and slowed activation kinetics. Similar effects were seen on gating of cloned Kv4.2 channels, but the inhibition was less pronounced and required higher toxin concentrations. These results reveal SNX-482 as the most potent inhibitor of Kv4.3 channels yet identified. Because of the effects on both Kv4.3 and Kv4.2 channels, caution is needed when interpreting the effects of SNX-482 on cells and circuits where these channels are present.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Shal/efectos de los fármacos , Venenos de Araña/farmacología , Animales , Células Cultivadas , Neuronas Dopaminérgicas/fisiología , Femenino , Células HEK293 , Humanos , Concentración 50 Inhibidora , Activación del Canal Iónico/fisiología , Masculino , Potenciales de la Membrana/fisiología , Ratones , Técnicas de Placa-Clamp , Potasio/metabolismo , Canales de Potasio Shal/fisiología
9.
Circ Res ; 112(10): 1310-22, 2013 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-23532596

RESUMEN

RATIONALE: A chromosomal haplotype producing cardiac overexpression of dipeptidyl peptidase-like protein-6 (DPP6) causes familial idiopathic ventricular fibrillation. The molecular basis of transient outward current (I(to)) in Purkinje fibers (PFs) is poorly understood. We hypothesized that DPP6 contributes to PF I(to) and that its overexpression might specifically alter PF I(to) properties and repolarization. OBJECTIVE: To assess the potential role of DPP6 in PF I(to). METHODS AND RESULTS: Clinical data in 5 idiopathic ventricular fibrillation patients suggested arrhythmia origin in the PF-conducting system. PF and ventricular muscle I(to) had similar density, but PF I(to) differed from ventricular muscle in having tetraethylammonium sensitivity and slower recovery. DPP6 overexpression significantly increased, whereas DPP6 knockdown reduced, I(to) density and tetraethylammonium sensitivity in canine PF but not in ventricular muscle cells. The K(+)-channel interacting ß-subunit K(+)-channel interacting protein type-2, essential for normal expression of I(to) in ventricular muscle, was weakly expressed in human PFs, whereas DPP6 and frequenin (neuronal calcium sensor-1) were enriched. Heterologous expression of Kv4.3 in Chinese hamster ovary cells produced small I(to); I(to) amplitude was greatly enhanced by coexpression with K(+)-channel interacting protein type-2 or DPP6. Coexpression of DPP6 with Kv4.3 and K(+)-channel interacting protein type-2 failed to alter I(to) compared with Kv4.3/K(+)-channel interacting protein type-2 alone, but DPP6 expression with Kv4.3 and neuronal calcium sensor-1 (to mimic PF I(to) composition) greatly enhanced I(to) compared with Kv4.3/neuronal calcium sensor-1 and recapitulated characteristic PF kinetic/pharmacological properties. A mathematical model of cardiac PF action potentials showed that I(to) enhancement can greatly accelerate PF repolarization. CONCLUSIONS: These results point to a previously unknown central role of DPP6 in PF I(to), with DPP6 gain of function selectively enhancing PF current, and suggest that a DPP6-mediated PF early-repolarization syndrome might be a novel molecular paradigm for some forms of idiopathic ventricular fibrillation.


Asunto(s)
Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/fisiología , Proteínas de Interacción con los Canales Kv/fisiología , Proteínas del Tejido Nervioso/fisiología , Canales de Potasio/fisiología , Ramos Subendocárdicos/fisiología , Canales de Potasio Shal/fisiología , Fibrilación Ventricular/fisiopatología , Adulto , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Modelos Animales de Enfermedad , Perros , Femenino , Técnicas de Silenciamiento del Gen , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Técnicas In Vitro , Proteínas de Interacción con los Canales Kv/efectos de los fármacos , Proteínas de Interacción con los Canales Kv/genética , Masculino , Persona de Mediana Edad , Modelos Teóricos , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Canales de Potasio/genética , Ramos Subendocárdicos/patología , Canales de Potasio Shal/efectos de los fármacos , Canales de Potasio Shal/genética , Tetraetilamonio/farmacología , Transfección
10.
J Neurosci ; 32(41): 14427-32, 2012 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23055512

RESUMEN

Neuronal activity is critical for the formation and modification of neural circuits during brain development. In hippocampal CA1 pyramidal dendrites, A-type voltage-gated K(+) currents, formed primarily by Kv4.2 subunits, control excitability. Here we used Kv4.2 knock-out (Kv4.2-KO) mice along with acute in vivo expression of Kv4.2 or its dominant-negative pore mutant to examine the role of Kv4.2 in the development of CA1 synapses. We found that Kv4.2 expression induces synaptic maturation in juvenile WT mice and rescues developmentally delayed synapses in adult Kv4.2-KO mice. In addition, we show that NMDAR subunit composition can be reverted back to the juvenile form in WT adult synapses by functionally downregulating Kv4.2 levels. These results suggest that Kv4.2 regulation of excitability determines synaptic maturation state, which can be bidirectionally adjusted into adulthood.


Asunto(s)
Región CA1 Hipocampal/fisiología , Neurogénesis/fisiología , Canales de Potasio Shal/fisiología , Sinapsis/fisiología , Animales , Región CA1 Hipocampal/citología , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación de Dinámica Molecular , Neurogénesis/genética , Canales de Potasio Shal/deficiencia , Canales de Potasio Shal/genética , Sinapsis/genética
11.
J Neurosci ; 32(44): 15511-20, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-23115188

RESUMEN

Chronically altered levels of network activity lead to changes in the morphology and functions of neurons. However, little is known of how changes in neuronal activity alter the intracellular signaling pathways mediating neuronal survival. Here, we use primary cultures of rat hippocampal neurons to show that elevated neuronal activity impairs phosphorylation of the serine/threonine kinase, Erk1/2, and the activation of signal transducer and activator of transcription 3 (STAT3) by phosphorylation of serine 727. Chronically stimulated neurons go through apoptosis when they fail to activate another serine/threonine kinase, Akt. Gain- and loss-of-function experiments show that STAT3 plays the key role directly downstream from Erk1/2 as the alternative survival pathway. Elevated neuronal activity resulted in increased expression of a tumor suppressor, p53, and its target gene, Bax. These changes are observed in Kv4.2 knock-out mouse hippocampal neurons, which are also sensitive to the blockade of TrkB signaling, confirming that the alteration occurs in vivo. Thus, this study provides new insight into a mechanism by which chronic elevation of activity may cause neurodegeneration.


Asunto(s)
Hipocampo/fisiología , Neuronas/fisiología , Factor de Transcripción STAT3/fisiología , Transducción de Señal/fisiología , Animales , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/fisiología , Calcio/metabolismo , Recuento de Células , Supervivencia Celular/fisiología , Inmunoprecipitación de Cromatina , Hipocampo/citología , Inmunohistoquímica , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/patología , Neuroimagen , Proteínas Proto-Oncogénicas c-akt/fisiología , Reacción en Cadena en Tiempo Real de la Polimerasa , Canales de Potasio Shal/genética , Canales de Potasio Shal/fisiología , Transfección
12.
J Physiol ; 591(10): 2419-27, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23440961

RESUMEN

K+ channels containing Kv4.2 and Kv4.3 pore-forming subunits mediate most of the subthreshold-operating somatodendritic A-type K+ current in CNS neurons. These channels are believed to be important in regulating the frequency of repetitive firing, the backpropagation of action potential into dendrites, and dendritic integration and plasticity. Moreover, they have been implicated in several diseases from pain to epilepsy and autism spectrum disorders. The lack of toxins that specifically and efficiently block these channels has hampered studies aimed at confirming their functional role and their involvement in disease. AmmTX3 and other related members of the α-KTX15 family of scorpion toxins have been shown to block the A-type K+ current in cultured neurons, but their specificity has been questioned because the toxins do not efficiently block the currents mediated by Kv4.2 or Kv4.3 subunits expressed in heterologous cells. Here we show that the high-affinity blockade of Kv4.2 and Kv4.3 channels by AmmTX3 depends on the presence of the auxiliary subunits DPP6 and DPP10. These proteins are thought to be components of the Kv4 channel complex in neurons and to be important for channel expression in dendrites. These studies validate the use of AmmTX3 as a blocker of the Kv4-mediated A-type K+ current in neurons.


Asunto(s)
Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/fisiología , Venenos de Escorpión/farmacología , Canales de Potasio Shal/fisiología , Animales , Células CHO , Células Cultivadas , Cerebelo/citología , Cerebelo/fisiología , Cricetinae , Cricetulus , Ratones , Ratones Noqueados , Neuronas/fisiología , Proteínas Recombinantes/farmacología
13.
Biochem Biophys Res Commun ; 436(4): 591-4, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23747723

RESUMEN

Kv4.3 K(+) channels contributing to Ito are involved in the repolarization of cardiac action potential. Kv4.3 K(+) channels decrease in pathological cardiac hypertrophy, but the mechanism remains unclear. Our previous study found that the expression of bone morphogenetic protein 4 (BMP4) increased in pressure-overload and Ang II constant infusion induced cardiac hypertrophy. Since the downregulation of Kv4.3 K(+) channels and the upregulation of BMP4 simultaneously occur in pathological cardiac hypertrophy, we hypothesize that the up-regulated BMP4 would contribute to the downregulation of Kv4.3 K(+) channels in cardiac hypertrophy. We found that BMP4 treatment reduced Kv4.3 but not Kv4.2 and Kv1.4 K(+) channel protein expression, and BMP4-induced decrease of Kv4.3 K(+) channel protein expression was reversed by BMP4 inhibitor noggin and DMH1 in cultured cardiomyocytes in vitro. BMP4-induced decrease of Kv4.3 K(+) channel protein expression was also reversed by the NADPH oxidase inhibitor apocynin and the radical scavenger tempol. In in vivo transverse aortic constriction (TAC)-induced cardiac hypertrophy, constant infusion of DMH1 completely rescued TAC-induced down-regulation of Kv4.3 K(+) channel protein expression. We conclude that BMP4 contributes to the downregulation of Kv4.3 K(+) channels in pathological cardiac hypertrophy and the underlying mechanism might be through increasing ROS production.


Asunto(s)
Proteína Morfogenética Ósea 4/fisiología , Cardiomegalia/fisiopatología , Regulación hacia Abajo/fisiología , Canales de Potasio Shal/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Humanos , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Biochem J ; 441(3): 859-67, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22023388

RESUMEN

Down-regulation of Kv4.3 K⁺ channels commonly occurs in multiple diseases, but the understanding of the regulation of Kv4.3 K⁺ channels and the role of Kv4.3 K⁺ channels in pathological conditions are limited. HEK (human embryonic kidney)-293T cells are derived from HEK-293 cells which are transformed by expression of the large T-antigen. In the present study, by comparing HEK-293 and HEK-293T cells, we find that HEK-293T cells express more Kv4.3 K⁺ channels and more transcription factor Sp1 (specificity protein 1) than HEK-293 cells. Inhibition of Sp1 with Sp1 decoy oligonucleotide reduces Kv4.3 K⁺ channel expression in HEK-293T cells. Transfection of pN3-Sp1FL vector increases Sp1 protein expression and results in increased Kv4.3 K⁺ expression in HEK-293 cells. Since the ultimate determinant of the phenotype difference between HEK-293 and HEK-293T cells is the large T-antigen, we conclude that the large T-antigen up-regulates Kv4.3 K⁺ channel expression through an increase in Sp1. In both HEK-293 and HEK-293T cells, inhibition of Kv4.3 K⁺ channels with 4-AP (4-aminopyridine) or Kv4.3 small interfering RNA induces cell apoptosis and necrosis, which are completely rescued by the specific CaMKII (calcium/calmodulin-dependent protein kinase II) inhibitor KN-93, suggesting that Kv4.3 K⁺ channels contribute to cell apoptosis and necrosis through CaMKII activation. In summary, we establish: (i) the HEK-293 and HEK-293T cell model for Kv4.3 K⁺ channel study; (ii) that large T-antigen up-regulates Kv4.3 K⁺ channels through increasing Sp1 levels; and (iii) that Kv4.3 K⁺ channels contribute to cell apoptosis and necrosis through activating CaMKII. The present study provides deep insights into the mechanism of the regulation of Kv4.3 K⁺ channels and the role of Kv4.3 K⁺ channels in cell death.


Asunto(s)
Antígenos Virales de Tumores/fisiología , Apoptosis/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Necrosis/genética , Canales de Potasio Shal/genética , Canales de Potasio Shal/fisiología , Factor de Transcripción Sp1/fisiología , Antígenos Virales de Tumores/genética , Antígenos Virales de Tumores/metabolismo , Apoptosis/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Modelos Biológicos , Necrosis/metabolismo , ARN Interferente Pequeño/farmacología , Canales de Potasio Shal/antagonistas & inhibidores , Canales de Potasio Shal/metabolismo , Regulación hacia Arriba/genética
15.
Am J Physiol Lung Cell Mol Physiol ; 303(7): L598-607, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22865553

RESUMEN

Pulmonary neuroepithelial bodies (NEBs), composed of clusters of amine [serotonin (5-HT)] and peptide-producing cells, are widely distributed within the airway mucosa of human and animal lungs. NEBs are thought to function as airway O(2)-sensors, since they are extensively innervated and release 5-HT upon hypoxia exposure. The small cell lung carcinoma cell line (H146) provides a useful model for native NEBs, since they contain (and secrete) 5-HT and share the expression of a membrane-delimited O(2) sensor [classical NADPH oxidase (NOX2) coupled to an O(2)-sensitive K(+) channel]. In addition, both native NEBs and H146 cells express different NADPH oxidase homologs (NOX1, NOX4) and its subunits together with a variety of O(2)-sensitive voltage-dependent K(+) channel proteins (K(v)) and tandem pore acid-sensing K(+) channels (TASK). Here we used H146 cells to investigate the role and interactions of various NADPH oxidase components in O(2)-sensing using a combination of coimmunoprecipitation, Western blot analysis (quantum dot labeling), and electrophysiology (patchclamp, amperometry) methods. Coimmunoprecipitation studies demonstrated formation of molecular complexes between NOX2 and K(v)3.3 and K(v)4.3 ion channels but not with TASK1 ion channels, while NOX4 associated with TASK1 but not with K(v) channel proteins. Downregulation of mRNA for NOX2, but not for NOX4, suppressed hypoxia-sensitive outward current and significantly reduced hypoxia -induced 5-HT release. Collectively, our studies suggest that NOX2/K(v) complexes are the predominant O(2) sensor in H146 cells and, by inference, in native NEBs. Present findings favor a NEB cell-specific plasma membrane model of O(2)-sensing and suggest that unique NOX/K(+) channel combinations may serve diverse physiological functions.


Asunto(s)
Células Quimiorreceptoras/fisiología , Glicoproteínas de Membrana/fisiología , NADPH Oxidasas/fisiología , Cuerpos Neuroepiteliales/fisiología , Oxígeno/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Glicoproteínas de Membrana/química , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/química , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/fisiología , Canales de Potasio de Dominio Poro en Tándem/química , Canales de Potasio de Dominio Poro en Tándem/fisiología , Serotonina/metabolismo , Canales de Potasio Shal/química , Canales de Potasio Shal/fisiología , Canales de Potasio Shaw/química , Canales de Potasio Shaw/fisiología
16.
Eur Heart J ; 32(3): 305-15, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21148163

RESUMEN

AIMS: Reduction of transient outward current (I(to)) and excessive activation of Ca(2+)/Calmodulin-dependent kinase II (CaMKII) are general features of ventricular myocytes in heart failure. We hypothesize that alterations of I(to) directly regulate CaMKII activation in cardiomyocytes. METHODS AND RESULTS: A dynamic coupling of I(to) channel subunit Kv4.3 and inactive CaMKII was discovered in cardiomyocytes with the membrane predominant distribution by co-immunoprecipitation and fluorescence resonance energy transfer techniques. CaMKII dissociation from Kv4.3-CaMKII units caused a significant increase in CaMKII autophosphorylation and L-type calcium current (I(Ca)) facilitation. I(Ca) facilitation was blunted by the compartmental Ca²(+) chelator BAPTA but unaffected by bulk Ca²(+) chelator EGTA, implicating membrane-localized CaMKII. Kv4.3 overexpression reduced basal CaMKII autophosphorylation in myocytes and eliminated Ca²(+)-induced CaMKII activation. Kv4.3 blocks CaMKII activation by binding to the calmodulin binding sites, whereas Kv4.3 uncoupling releases these sites and leads to a substantial CaMKII activation. CONCLUSION: Our results uncovered an important mechanism that regulates CaMKII activation in the heart and implicate I(to) channel alteration in pathological CaMKII activation.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Miocitos Cardíacos/metabolismo , Canales de Potasio Shal/fisiología , 4-Aminopiridina/farmacología , Animales , Calcio/metabolismo , Quelantes/farmacología , Ácido Edético/farmacología , Ácido Egtácico/farmacología , Células HEK293 , Cardiopatías/etiología , Cardiopatías/prevención & control , Humanos , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Canales de Potasio Shal/metabolismo
17.
Neuron ; 54(6): 850-2, 2007 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-17582324

RESUMEN

Everyone agrees about how long-term potentiation (LTP) is induced-NMDA receptor activation-but much remains to be learned about how the increase in the strength of a synaptic connection between two neurons is expressed. In this issue of Neuron, Kim et al. report a new form of NMDAR-dependent plasticity that may contribute to LTP: internalization of postsynaptic Kv4.2 potassium channels that mediate transient IA-type outward current in dendrites.


Asunto(s)
Dendritas/fisiología , Plasticidad Neuronal/fisiología , Canales de Potasio Shal/fisiología , Animales , Receptores de N-Metil-D-Aspartato/fisiología
18.
J Neurosci ; 30(25): 8551-65, 2010 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-20573902

RESUMEN

In cerebellar granule (CG) cells and many other neurons, A-type potassium currents play an important role in regulating neuronal excitability, firing patterns, and activity-dependent plasticity. Protein biochemistry has identified dipeptidyl peptidase-like protein 6 (DPP6) as an auxiliary subunit of Kv4-based A-type channels and thus a potentially important regulator of neuronal excitability. In this study, we used an RNA interference (RNAi) strategy to examine the role DPP6 plays in forming and shaping the electrophysiological properties of CG cells. DPP6 RNAi delivered by lentiviral vectors effectively disrupts DPP6 protein expression in CG cells. In response to the loss of DPP6, I(SA) peak conductance amplitude is reduced by >85% in parallel with a dramatic reduction in the level of I(SA) channel protein complex found in CG cells. The I(SA) channels remaining in CG cells after suppression of DPP6 show alterations in gating similar to Kv4 channels expressed in heterologous systems without DPP6. In addition to these effects on A-type current, we find that loss of DPP6 has additional effects on input resistance and Na(+) channel conductance that combine with the effects on I(SA) to produce a global change in excitability. Overall, DPP6 expression seems to be critical for the expression of a high-frequency electrophysiological phenotype in CG cells by increasing leak conductance, A-type current levels and kinetics, and Na(+) current amplitude.


Asunto(s)
Potenciales de Acción/fisiología , Cerebelo/fisiología , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Canales de Potasio/metabolismo , Análisis de Varianza , Western Blotting , Línea Celular , Cerebelo/citología , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Electrofisiología , Vectores Genéticos , Hipocampo/citología , Hipocampo/fisiología , Humanos , Lentivirus , Proteínas del Tejido Nervioso/genética , Canales de Potasio/genética , Interferencia de ARN , Canales de Potasio Shal/fisiología
19.
Mol Pharmacol ; 80(2): 345-55, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21540294

RESUMEN

Kv4 (Shal) potassium channels are responsible for the transient outward K(+) currents in mammalian hearts and central nervous systems. Heteropoda toxin 2 (HpTx2) is an inhibitor cysteine knot peptide toxin specific for Kv4 channels that inhibits gating of Kv4.3 in the voltage-dependent manner typical for this type of toxin. HpTx2 interacts with four independent binding sites containing two conserved hydrophobic amino acids in the S3b transmembrane segments of Kv4.3 and the closely related Kv4.1. Despite these similarities, HpTx2 interaction with Kv4.1 is considerably less voltage-dependent, has smaller shifts in the voltage-dependences of conductance and steady-state inactivation, and a 3-fold higher K(d) value. Swapping four nonconserved amino acids in S3b between the two channels exchanges the phenotypic response to HpTx2. To understand these differences in gating modification, we constructed Markov models of Kv4.3 and Kv4.1 activation gating in the presence of HpTx2. Both models feature a series of voltage-dependent steps leading to a final voltage-independent transition to the open state and closely replicate the experimental data. Interaction with HpTx2 increases the energy barrier for channel opening by slowing activation and accelerating deactivation. The greater degree of voltage-dependence in Kv4.3 occurs because it is the voltage-dependent transitions that are most affected by HpTx2; in contrast, it is the voltage-independent step in Kv4.1 that is most affected by the presence of toxin. These data demonstrate the basis for subtype-specificity of HpTx2 and point the way to a general model of gating modifier toxin interaction with voltage-gated ion channels.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Canales de Potasio Shal/metabolismo , Venenos de Araña/metabolismo , Secuencia de Aminoácidos , Animales , Femenino , Ratones , Datos de Secuencia Molecular , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Ratas , Canales de Potasio Shal/fisiología , Venenos de Araña/farmacología , Xenopus laevis
20.
J Physiol ; 589(Pt 3): 461-79, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21098008

RESUMEN

Inactivation of voltage-gated ion channels is an intrinsic auto-regulatory process necessary to govern the occurrence and shape of action potentials and establish firing patterns in excitable tissues. Inactivation may occur from the open state (open-state inactivation, OSI) at strongly depolarized membrane potentials, or from pre-open closed states (closed-state inactivation, CSI) at hyperpolarized and modestly depolarized membrane potentials. Voltage-gated Na(+), K(+), Ca(2+) and non-selective cationic channels utilize both OSI and CSI. Whereas there are detailed mechanistic descriptions of OSI, much less is known about the molecular basis of CSI. Here, we review evidence for CSI in voltage-gated cationic channels (VGCCs) and recent findings that shed light on the molecular mechanisms of CSI in voltage-gated K(+) (Kv) channels. Particularly, complementary observations suggest that the S4 voltage sensor, the S4S5 linker and the main S6 activation gate are instrumental in the installment of CSI in Kv4 channels. According to this hypothesis, the voltage sensor may adopt a distinct conformation to drive CSI and, depending on the stability of the interactions between the voltage sensor and the pore domain, a closed-inactivated state results from rearrangements in the selectivity filter or failure of the activation gate to open. Kv4 channel CSI may efficiently exploit the dynamics of the subthreshold membrane potential to regulate spiking properties in excitable tissues.


Asunto(s)
Activación del Canal Iónico/fisiología , Canales Iónicos/fisiología , Potenciales de la Membrana/fisiología , Animales , Humanos , Canales de Potasio Shal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA