Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuropharmacology ; 185: 108399, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33400937

RESUMEN

Midbrain dopamine neurons (DANs) regulate various brain functions such as motor control and motivation. Alteration of spiking activities of these neurons could contribute to severe brain disorders including Parkinson's disease and depression. Previous studies showed important roles of somatodendritic voltage-gated K+ channels (Kv) of DANs in governing neuronal excitability and dopamine release. However, it remains largely unclear about the biophysical properties and the function of Kv channels distributed at DAN axons. We performed whole-cell recordings from the axons of DANs in acute mouse midbrain and striatal slices. We detected both rapidly activating/inactivating Kv current (i.e. A-current) and rapidly activating but slowly inactivating current (i.e. D-current) in DAN axons. Pharmacological experiments with channel blockers revealed that these currents are predominantly mediated by Kv1.4 and Kv1.2 subunits, respectively. Blocking these currents could substantially prolong axonal action potentials (APs) via a reduction of their repolarization slope. Together, our results show that Kv channels mediating A- and D-currents shape AP waveforms in midbrain DAN axons, through this regulation they may control dopamine release at the axonal terminals. Therefore, these axonal Kv channels could be drug targets for brain disorders with abnormal dopamine release.


Asunto(s)
Potenciales de Acción/fisiología , Axones/fisiología , Neuronas Dopaminérgicas/fisiología , Canal de Potasio Kv1.3/fisiología , Canal de Potasio Kv1.4/fisiología , Mesencéfalo/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Femenino , Proteínas de Interacción con los Canales Kv/antagonistas & inhibidores , Proteínas de Interacción con los Canales Kv/fisiología , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.4/antagonistas & inhibidores , Masculino , Mesencéfalo/efectos de los fármacos , Ratones , Ratones Transgénicos , Bloqueadores de los Canales de Potasio/farmacología
2.
Nat Rev Dis Primers ; 5(1): 30, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31048702

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease caused by antibodies against the acetylcholine receptor (AChR), muscle-specific kinase (MuSK) or other AChR-related proteins in the postsynaptic muscle membrane. Localized or general muscle weakness is the predominant symptom and is induced by the antibodies. Patients are grouped according to the presence of antibodies, symptoms, age at onset and thymus pathology. Diagnosis is straightforward in most patients with typical symptoms and a positive antibody test, although a detailed clinical and neurophysiological examination is important in antibody-negative patients. MG therapy should be ambitious and aim for clinical remission or only mild symptoms with near-normal function and quality of life. Treatment should be based on MG subgroup and includes symptomatic treatment using acetylcholinesterase inhibitors, thymectomy and immunotherapy. Intravenous immunoglobulin and plasma exchange are fast-acting treatments used for disease exacerbations, and intensive care is necessary during exacerbations with respiratory failure. Comorbidity is frequent, particularly in elderly patients. Active physical training should be encouraged.


Asunto(s)
Miastenia Gravis/diagnóstico , Miastenia Gravis/terapia , Acetilcolinesterasa/genética , Acetilcolinesterasa/fisiología , Corticoesteroides/uso terapéutico , Agrina/genética , Agrina/fisiología , Antiinflamatorios no Esteroideos/uso terapéutico , Autoanticuerpos/análisis , Autoanticuerpos/sangre , Biomarcadores/análisis , Biomarcadores/sangre , Blefaroptosis/etiología , Colágeno/genética , Colágeno/fisiología , Cortactina/genética , Cortactina/fisiología , Electromiografía/métodos , Humanos , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Proteínas Relacionadas con Receptor de LDL/genética , Proteínas Relacionadas con Receptor de LDL/fisiología , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Miastenia Gravis/fisiopatología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Colinérgicos/genética , Receptores Colinérgicos/fisiología , Receptores Nicotínicos/genética , Factores de Riesgo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/fisiología
3.
Neuropharmacology ; 146: 65-73, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30465811

RESUMEN

Voltage-gated potassium channels (VGKCs) are transmembrane ion channels specific for potassium. Currently there are nine kinds of VGKCs. Kv1.4 is one of shaker-related potassium channels. It is a representative alpha subunit of potassium channels that can inactivate A type-currents, leading to N pattern inactivation. Inactivation of Kv channels plays an important role in shaping electrical signaling properties of neuronal and muscular cells. The shape of N pattern inactivation can be modified by removing the N-terminal (NT) domain which results in non-inactivated currents and C pattern inactivation. In a previous work, we have reported the regulatory effect of metergoline on Kv1.4 and Nav1.2 channel activity. In the present study, we constructed a mutant of deleted 61 residues from NT of Kv1.4 channels (Kv1.4 Δ2-61) and found that it induced an outward peak and steady-state currents We also studied the modulation effect of metergoline on the activity of this Kv1.4 Δ2-61 mutant channel without having the N-terminal quick inactivation domain. Our results revealed that treatment with metergoline inhibited NT deleted Kv1.4 mutant channel activity in a concentration-dependent manner which was reversible. Interestingly, metergoline treatment induced little effects on the outward peak current in the deleted Kv1.4 mutant channel. However, metergoline treatment conspicuously inhibited steady state currents of Kv1.4 Δ2-61 channels with acceleration current mode. The acceleration of steady-state current of deleted Kv1.4 mutant channel occurred in a concentration-dependent manner. This means that metergoline can accelerate C pattern inactivation of Kv1.4 Δ2-61 channel by acting as an open state dependent channel blocker. We also performed site-directed mutations in V561A and K532Y, also known as C-type inactivation sites. V561A, K532Y, and V561A + K532Y substitution mutants significantly attenuated the acceleration effect of metergoline on C pattern inactivation of hKv1.4 channel currents. In docking modeling study, predicted binding residues for metergoline were analyzed for six amino acids. Among them, the K532 residue known as the C-type inactivation site was analyzed to be a major site of action. Then various mutants were constructed. K532 substitution mutant significantly abolished the effect of metergoline on Kv1.4 currents among various mutants whereas other changes had slight inhibitory effects. Furthermore, we found that metergoline had specificity for Kv1.4, but not for Kv1.5 currents. In addition, the A type current in rat neuronal cell was inhibited and accelerated of inactivation. This result further shows that metergoline might interact with Lys532 residue and then accelerate C pattern inactivation of Kv1.4 channels with channel type specificity. Taken together, these results demonstrate the molecular basis involved in the effect of metergoline, an ergot alkaloid, on human Kv1.4 channel, providing a novel interaction ligand.


Asunto(s)
Antidepresivos/farmacología , Canal de Potasio Kv1.4/antagonistas & inhibidores , Metergolina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Animales , Sitios de Unión , Cinética , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Lectinas Tipo C , Modelos Moleculares , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Neuronas/fisiología , Oocitos , Canales de Potasio con Entrada de Voltaje , Ratas , Relación Estructura-Actividad , Xenopus laevis
4.
Biol Pharm Bull ; 39(6): 1069-72, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27251511

RESUMEN

Metergoline is an ergot-derived psychoactive drug that is a ligand for various serotonin and dopamine receptors. Little is known about the effect of metergoline on different types of receptors and ion channels. Potassium channels are the most diverse group of ion channels. Kv1.4, a shaker family K channel alpha subunit, is one of a family of voltage gated K channels that mediates transient and rapid inactivating A-type currents and N-type inactivation. We demonstrated previously that metergoline inhibited the activity of neuronal voltage-dependent Na(+) channels in Xenopus laevis oocytes (Acta Pharmacol. Sin., 35, 2014, Lee et al.). In this study, we sought to elucidate the regulatory effects underlying metergoline-induced human Kv1.4 channel inhibition. We used the two electrode voltage-clamp (TEVC) technique to investigate the effect of metergoline on human Kv1.4 channel currents in Xenopus laevis oocytes expressing human Kv1.4 alpha subunits. Interestingly, metergoline treatment also induced inhibition of peak currents in human Kv1.4 channels in a concentration-dependent manner. The IC50 of peak currents of hKv1.4 currents was 3.6±0.6 µM. These results indicate that metergoline might regulate the human Kv1.4 channel activity that is expressed in X. laevis oocytes. Further, this regulation of potassium currents by metergoline might be one of the pharmacological actions of metergoline-mediated psychoactivity.


Asunto(s)
Antidepresivos/farmacología , Canal de Potasio Kv1.4/antagonistas & inhibidores , Metergolina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Animales , Femenino , Humanos , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Oocitos/fisiología , Xenopus laevis
5.
PLoS One ; 11(5): e0154077, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27149380

RESUMEN

BACKGROUND: The left atrial posterior wall (LAPW) is potentially an important area for the development and maintenance of atrial fibrillation. We assessed whether there are regional electrical differences throughout the murine left atrial myocardium that could underlie regional differences in arrhythmia susceptibility. METHODS: We used high-resolution optical mapping and sharp microelectrode recordings to quantify regional differences in electrical activation and repolarisation within the intact, superfused murine left atrium and quantified regional ion channel mRNA expression by Taqman Low Density Array. We also performed selected cellular electrophysiology experiments to validate regional differences in ion channel function. RESULTS: Spontaneous ectopic activity was observed during sustained 1Hz pacing in 10/19 intact LA and this was abolished following resection of LAPW (0/19 resected LA, P<0.001). The source of the ectopic activity was the LAPW myocardium, distinct from the pulmonary vein sleeve and LAA, determined by optical mapping. Overall, LAPW action potentials (APs) were ca. 40% longer than the LAA and this region displayed more APD heterogeneity. mRNA expression of Kcna4, Kcnj3 and Kcnj5 was lower in the LAPW myocardium than in the LAA. Cardiomyocytes isolated from the LAPW had decreased Ito and a reduced IKACh current density at both positive and negative test potentials. CONCLUSIONS: The murine LAPW myocardium has a different electrical phenotype and ion channel mRNA expression profile compared with other regions of the LA, and this is associated with increased ectopic activity. If similar regional electrical differences are present in the human LA, then the LAPW may be a potential future target for treatment of atrial fibrillation.


Asunto(s)
Complejos Atriales Prematuros/fisiopatología , Atrios Cardíacos/fisiopatología , Canales Iónicos/fisiología , Potenciales de Acción/fisiología , Animales , Función Atrial/fisiología , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/análisis , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/fisiología , Atrios Cardíacos/química , Canales Iónicos/análisis , Canal de Potasio Kv1.4/análisis , Canal de Potasio Kv1.4/fisiología , Masculino , Ratones , Miocitos Cardíacos/química , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp
6.
J Biol Rhythms ; 30(5): 396-407, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26152125

RESUMEN

Neurons in the suprachiasmatic nucleus (SCN), the master circadian pacemaker in mammals, display daily rhythms in electrical activity with more depolarized resting potentials and higher firing rates during the day than at night. Although these daily variations in the electrical properties of SCN neurons are required for circadian rhythms in physiology and behavior, the mechanisms linking changes in neuronal excitability to the molecular clock are not known. Recently, we reported that mice deficient for either Kcna4 (Kv1.4(-/-)) or Kcnd2 (Kv4.2(-/-); but not Kcnd3, Kv4.3(-/-)), voltage-gated K(+) (Kv) channel pore-forming subunits that encode subthreshold, rapidly activating, and inactivating K(+) currents (IA), have shortened (0.5 h) circadian periods in SCN firing and in locomotor activity compared with wild-type (WT) mice. In the experiments here, we used a mouse (Per2(Luc)) line engineered with a bioluminescent reporter construct, PERIOD2::LUCIFERASE (PER2::LUC), replacing the endogenous Per2 locus, to test the hypothesis that the loss of Kv1.4- or Kv4.2-encoded IA channels also modifies circadian rhythms in the expression of the clock protein PERIOD2 (PER2). We found that SCN explants from Kv1.4(-/-)Per2(Luc) and Kv4.2(-/-) Per2(Luc), but not Kv4.3(-/-)Per2(Luc), mice have significantly shorter (by approximately 0.5 h) circadian periods in PER2 rhythms, compared with explants from Per2(Luc) mice, revealing that the membrane properties of SCN neurons feedback to regulate clock (PER2) expression. The combined loss of both Kv1.4- and Kv4.2-encoded IA channels in Kv1.4(-/-)/Kv4.2(-/-)Per2(Luc) SCN explants did not result in any further alterations in PER2 rhythms. Interestingly, however, mice lacking both Kv1.4 and Kv4.2 show a striking (approximately 1.8 h) advance in their daily activity onset in a light cycle compared with WT mice, suggesting additional roles for Kv1.4- and Kv4.2-encoded IA channels in controlling the light-dependent responses of neurons within and/or outside of the SCN to regulate circadian phase of daily activity.


Asunto(s)
Ritmo Circadiano/fisiología , Canal de Potasio Kv1.4/fisiología , Proteínas Circadianas Period/metabolismo , Canales de Potasio Shal/fisiología , Núcleo Supraquiasmático/fisiología , Animales , Ritmo Circadiano/genética , Activación del Canal Iónico/genética , Activación del Canal Iónico/fisiología , Canal de Potasio Kv1.4/genética , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Masculino , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Actividad Motora/genética , Actividad Motora/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Técnicas de Placa-Clamp , Proteínas Circadianas Period/genética , Canales de Potasio Shal/genética , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/metabolismo , Técnicas de Cultivo de Tejidos
7.
J Urol ; 190(6): 2296-304, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23896350

RESUMEN

PURPOSE: To clarify the functional and molecular mechanisms inducing hyperexcitability of C-fiber bladder afferent pathways after spinal cord injury we examined changes in the electrophysiological properties of bladder afferent neurons, focusing especially on voltage-gated K channels. MATERIALS AND METHODS: Freshly dissociated L6-S1 dorsal root ganglion neurons were prepared from female spinal intact and spinal transected (T9-T10 transection) Sprague Dawley® rats. Whole cell patch clamp recordings were performed on individual bladder afferent neurons. Kv1.2 and Kv1.4 α-subunit expression levels were also evaluated by immunohistochemical and real-time polymerase chain reaction methods. RESULTS: Capsaicin sensitive bladder afferent neurons from spinal transected rats showed increased cell excitability, as evidenced by lower spike activation thresholds and a tonic firing pattern. The peak density of transient A-type K+ currents in capsaicin sensitive bladder afferent neurons from spinal transected rats was significantly less than that from spinal intact rats. Also, the KA current inactivation curve was displaced to more hyperpolarized levels after spinal transection. The protein and mRNA expression of Kv1.4 α-subunits, which can form transient A-type K+ channels, was decreased in bladder afferent neurons after spinal transection. CONCLUSIONS: Results indicate that the excitability of capsaicin sensitive C-fiber bladder afferent neurons is increased in association with reductions in transient A-type K+ current density and Kv1.4 α-subunit expression in injured rats. Thus, the Kv1.4 α-subunit could be a molecular target for treating overactive bladder due to neurogenic detrusor overactivity.


Asunto(s)
Canal de Potasio Kv1.4/fisiología , Neuronas Aferentes/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Vejiga Urinaria/inervación , Vejiga Urinaria/fisiopatología , Vías Aferentes/fisiopatología , Animales , Femenino , Ratas , Ratas Sprague-Dawley
8.
Eur J Neurosci ; 36(12): 3698-708, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23009328

RESUMEN

Neocortical networks produce oscillations that often correspond to characteristic physiological or pathological patterns. However, the mechanisms underlying the generation of and the transitions between such oscillatory states remain poorly understood. In this study, we examined resonance in mouse layer V neocortical pyramidal neurons. To accomplish this, we employed standard electrophysiology to describe cellular resonance parameters. Bode plot analysis revealed a range of resonance magnitude values in layer V neurons and demonstrated that both magnitude and phase response characteristics of layer V neocortical pyramidal neurons are modulated by changes in the extracellular environment. Specifically, increased resonant frequencies and total inductive areas were observed at higher extracellular potassium concentrations and more hyperpolarised membrane potentials. Experiments using pharmacological agents suggested that current through hyperpolarization-activated cyclic nucleotide-gated channels (I(h) ) acts as the primary driver of resonance in these neurons, with other potassium currents, such as A-type potassium current and delayed-rectifier potassium current (Kv1.4 and Kv1.1, respectively), contributing auxiliary roles. The persistent sodium current was also shown to play a role in amplifying the magnitude of resonance without contributing significantly to the phase response. Although resonance effects in individual neurons are small, their properties embedded in large networks may significantly affect network behavior and may have potential implications for pathological processes.


Asunto(s)
Potenciales de la Membrana , Neocórtex/fisiología , Red Nerviosa/fisiología , Células Piramidales/fisiología , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos/antagonistas & inhibidores , Canales Catiónicos Regulados por Nucleótidos Cíclicos/fisiología , Canal de Potasio Kv.1.1/antagonistas & inhibidores , Canal de Potasio Kv.1.1/fisiología , Canal de Potasio Kv1.4/antagonistas & inhibidores , Canal de Potasio Kv1.4/fisiología , Ratones , Ratones Endogámicos , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Sodio/metabolismo
9.
Biochim Biophys Acta ; 1798(11): 2076-83, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20674541

RESUMEN

Inactivation of potassium channels plays an important role in shaping the electrical signalling properties of nerve and muscle cells. While it has been assumed that the rapid inactivation of the Kv1.4 channel is controlled by a "ball and chain" inactivation mechanism, the chain structure of the channel has not been well defined. Here, by conducting electrophysiological studies on variants containing mutations of the positively charged and negatively charged segments of the NH(2)-terminal of the channel protein, we show that neutralization or deletion of the positively charged segment (residues 83-98) significantly slowed the inactivation process. Replacement of this positively charged segment with the negatively charged segment (residues 123-137), and vice versa, so that both segments were simultaneously positively or negatively charged, also slowed the inactivation process. Furthermore, the inactivation process was not changed when the positively charged and the negatively charged segments were interchanged. In contrast, the voltage dependence of activation and inactivation of the channels was not significantly altered by these mutants. These results indicate that the electrostatic interaction between the positively and negatively charged segments plays a critical role in the inactivation process of the Kv1.4 channel. Taken together, we propose that the electrostatic interaction accelerates the inactivation of the Kv1.4 channel by making it easier for the inactivation ball to access its binding site.


Asunto(s)
Canal de Potasio Kv1.4/química , Secuencia de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , Interacciones Hidrofóbicas e Hidrofílicas , Canal de Potasio Kv1.4/fisiología , Magnesio/farmacología , Electricidad Estática , Relación Estructura-Actividad
10.
J Mol Cell Cardiol ; 48(1): 12-25, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19619557

RESUMEN

Rapidly activating and inactivating cardiac transient outward K(+) currents, I(to), are expressed in most mammalian cardiomyocytes, and contribute importantly to the early phase of action potential repolarization and to plateau potentials. The rapidly recovering (I(t)(o,f)) and slowly recovering (I(t)(o,s)) components are differentially expressed in the myocardium, contributing to regional heterogeneities in action potential waveforms. Consistent with the marked differences in biophysical properties, distinct pore-forming (alpha) subunits underlie the two I(t)(o) components: Kv4.3/Kv4.2 subunits encode I(t)(o,f), whereas Kv1.4 encodes I(t)(o,s), channels. It has also become increasingly clear that cardiac I(t)(o) channels function as components of macromolecular protein complexes, comprising (four) Kvalpha subunits and a variety of accessory subunits and regulatory proteins that influence channel expression, biophysical properties and interactions with the actin cytoskeleton, and contribute to the generation of normal cardiac rhythms. Derangements in the expression or the regulation of I(t)(o) channels in inherited or acquired cardiac diseases would be expected to increase the risk of potentially life-threatening cardiac arrhythmias. Indeed, a recently identified Brugada syndrome mutation in KCNE3 (MiRP2) has been suggested to result in increased I(t)(o,f) densities. Continued focus in this area seems certain to provide new and fundamentally important insights into the molecular determinants of functional I(t)(o) channels and into the molecular mechanisms involved in the dynamic regulation of I(t)(o) channel functioning in the normal and diseased myocardium.


Asunto(s)
Regulación de la Expresión Génica , Miocardio/metabolismo , Miocardio/patología , Canales de Potasio con Entrada de Voltaje/fisiología , Potasio/metabolismo , Animales , Humanos , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/metabolismo , Canal de Potasio Kv1.4/fisiología , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo , Canales de Potasio Shal/fisiología
11.
Circ Arrhythm Electrophysiol ; 2(3): 285-94, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19808479

RESUMEN

BACKGROUND: Potassium currents contribute to action potential duration (APD) and arrhythmogenesis. In heart failure, Ca/calmodulin-dependent protein kinase II (CaMKII) is upregulated and can alter ion channel regulation and expression. METHODS AND RESULTS: We examine the influence of overexpressing cytoplasmic CaMKIIdelta(C), both acutely in rabbit ventricular myocytes (24-hour adenoviral gene transfer) and chronically in CaMKIIdelta(C)-transgenic mice, on transient outward potassium current (I(to)), and inward rectifying current (I(K1)). Acute and chronic CaMKII overexpression increases I(to,slow) amplitude and expression of the underlying channel protein K(V)1.4. Chronic but not acute CaMKII overexpression causes downregulation of I(to,fast), as well as K(V)4.2 and KChIP2, suggesting that K(V)1.4 expression responds faster and oppositely to K(V)4.2 on CaMKII activation. These amplitude changes were not reversed by CaMKII inhibition, consistent with CaMKII-dependent regulation of channel expression and/or trafficking. CaMKII (acute and chronic) greatly accelerated recovery from inactivation for both I(to) components, but these effects were acutely reversed by AIP (CaMKII inhibitor), suggesting that CaMKII activity directly accelerates I(to) recovery. Expression levels of I(K1) and Kir2.1 mRNA were downregulated by CaMKII overexpression. CaMKII acutely increased I(K1), based on inhibition by AIP (in both models). CaMKII overexpression in mouse prolonged APD (consistent with reduced I(to,fast) and I(K1)), whereas CaMKII overexpression in rabbit shortened APD (consistent with enhanced I(K1) and I(to,slow) and faster I(to) recovery). Computational models allowed discrimination of contributions of different channel effects on APD. CONCLUSIONS: CaMKII has both acute regulatory effects and chronic expression level effects on I(to) and I(K1) with complex consequences on APD.


Asunto(s)
Potenciales de Acción/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Miocitos Cardíacos/fisiología , Canales de Potasio/fisiología , Potasio/metabolismo , Adenoviridae/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Femenino , Insuficiencia Cardíaca/fisiopatología , Cinética , Canal de Potasio Kv1.4/fisiología , Masculino , Ratones , Ratones Transgénicos , Modelos Cardiovasculares , Miocitos Cardíacos/citología , Conejos , Canales de Potasio Shal/fisiología , Transfección , Regulación hacia Arriba/fisiología
12.
Nat Neurosci ; 10(8): 1063-72, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17603477

RESUMEN

Proteins participate in various biological processes and can be harnessed to probe and control biological events selectively and reproducibly, but the genetic code limits the building block to 20 common amino acids for protein manipulation in living cells. The genetic encoding of unnatural amino acids will remove this restriction and enable new chemical and physical properties to be precisely introduced into proteins. Here we present new strategies for generating orthogonal tRNA-synthetase pairs, which made possible the genetic encoding of diverse unnatural amino acids in different mammalian cells and primary neurons. Using this new methodology, we incorporated unnatural amino acids with extended side chains into the K+ channel Kv1.4, and found that the bulkiness of residues in the inactivation peptide is essential for fast channel inactivation, a finding that had not been possible using conventional mutagenesis. This technique will stimulate and facilitate new molecular studies using tailored unnatural amino acids for cell biology and neurobiology.


Asunto(s)
Aminoácidos/genética , Código Genético , Mutagénesis Sitio-Dirigida/métodos , Neuronas/fisiología , Biosíntesis de Proteínas/genética , Aminoácidos/metabolismo , Aminoacil-ARNt Sintetasas/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Hipocampo/citología , Humanos , Canal de Potasio Kv1.4/química , Canal de Potasio Kv1.4/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Modelos Biológicos , Técnicas de Placa-Clamp/métodos , Ratas , Ratas Sprague-Dawley , Transfección
13.
Am J Physiol Cell Physiol ; 292(2): C778-87, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16956965

RESUMEN

Episodic ataxia type 1 (EA1) is a Shaker-like channelopathy characterized by continuous myokymia and attacks of imbalance with jerking movements of the head, arms, and legs. Although altered expression and gating properties of Kv1.1 channels underlie EA1, several disease-causing mechanisms remain poorly understood. It is likely that Kv1.1, Kv1.4, and Kvbeta1.1 subunits form heteromeric channels at hippocampal mossy fiber boutons from which Zn(2+) ions are released into the synaptic cleft in a Ca(2+)-dependent fashion. The sensitivity of this macromolecular channel complex to Zn(2+) is unknown. Here, we show that this heteromeric channel possesses a high-affinity (<10 muM) and a low-affinity (<0.5 mM) site for Zn(2+), which are likely to regulate channel availability at distinct presynaptic membranes. Furthermore, the EA1 mutation F184C, located within the S1 segment of the Kv1.1 subunit, markedly decreased the equilibrium dissociation constants for Zn(2+) binding to the high- and low-affinity sites. The functional characterization of the Zn(2+) effects on heteromeric channels harboring the F184C mutation also showed that this ion significantly 1) slowed the activation rate of the channel, 2) increased the time to reach peak current amplitude, 3) decreased the rate and amount of current undergoing N-type inactivation, and 4) slowed the repriming of the channel compared with wild-type channels. These results demonstrate that the EA1 mutation F184C will not only sensitize the homomeric Kv1.1 channel to extracellular Zn(2+), but it will also endow heteromeric channels with a higher sensitivity to this metal ion. During the vesicular release of Zn(2+), its effects will be in addition to the intrinsic gating defects caused by the mutation, which is likely to exacerbate the symptoms by impairing the integration and transmission of signals within specific brain areas.


Asunto(s)
Canal de Potasio Kv.1.1/fisiología , Canal de Potasio Kv1.4/fisiología , Zinc/farmacología , Animales , Calcio/fisiología , Cationes Bivalentes , Femenino , Humanos , Técnicas In Vitro , Activación del Canal Iónico , Canal de Potasio Kv.1.1/genética , Canal de Potasio Kv1.4/genética , Mutación , Oocitos/fisiología , Terminales Presinápticos/fisiología , Degeneraciones Espinocerebelosas/genética , Xenopus laevis
14.
Circulation ; 114(16): 1682-6, 2006 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-17030681

RESUMEN

BACKGROUND: One key element of natural pacemakers is the pacemaker current encoded by the hyperpolarization-activated nucleotide-gated channel (HCN) gene family. Although HCN gene transfer has been used to engineer biological pacemakers, this strategy may be confounded by unpredictable consequences of heteromultimerization with endogenous HCN family members and limited flexibility with regard to frequency tuning of the engineered pacemaker. METHODS AND RESULTS: To circumvent these limitations, we converted a depolarization-activated potassium-selective channel, Kv1.4, into a hyperpolarization-activated nonselective channel by site-directed mutagenesis (R447N, L448A, and R453I in S4 and G528S in the pore). Gene transfer into ventricular myocardium demonstrated the ability of this construct to induce pacemaker activity with spontaneous action potential oscillations in adult ventricular myocytes and idioventricular rhythms by in vivo electrocardiography. CONCLUSIONS: Given the sparse expression of Kv1 family channels in the human ventricle, gene transfer of a synthetic pacemaker channel based on the Kv1 family has novel therapeutic potential as a biological alternative to electronic pacemakers.


Asunto(s)
Relojes Biológicos/fisiología , Técnicas de Transferencia de Gen , Corazón/fisiología , Canales Iónicos/fisiología , Animales , Relojes Biológicos/genética , Línea Celular , Femenino , Cobayas , Humanos , Activación del Canal Iónico/genética , Activación del Canal Iónico/fisiología , Canales Iónicos/genética , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Miocitos Cardíacos/fisiología
15.
J Gen Physiol ; 127(4): 391-400, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16533897

RESUMEN

The intracellular tetramerization domain (T1) of most eukaryotic voltage-gated potassium channels (Kv channels) exists as a "hanging gondola" below the transmembrane regions that directly control activation gating via the electromechanical coupling between the S4 voltage sensor and the main S6 gate. However, much less is known about the putative contribution of the T1 domain to Kv channel gating. This possibility is mechanistically intriguing because the T1-S1 linker connects the T1 domain to the voltage-sensing domain. Previously, we demonstrated that thiol-specific reagents inhibit Kv4.1 channels by reacting in a state-dependent manner with native Zn(2+) site thiolate groups in the T1-T1 interface; therefore, we concluded that the T1-T1 interface is functionally active and not protected by Zn(2+) (Wang, G., M. Shahidullah, C.A. Rocha, C. Strang, P.J. Pfaffinger, and M. Covarrubias. 2005. J. Gen. Physiol. 126:55-69). Here, we co-expressed Kv4.1 channels and auxiliary subunits (KChIP-1 and DPPX-S) to investigate the state and voltage dependence of the accessibility of MTSET to the three interfacial cysteines in the T1 domain. The results showed that the average MTSET modification rate constant (k(MTSET)) is dramatically enhanced in the activated state relative to the resting and inactivated states (approximately 260- and approximately 47-fold, respectively). Crucially, under three separate conditions that produce distinct activation profiles, k(MTSET) is steeply voltage dependent in a manner that is precisely correlated with the peak conductance-voltage relations. These observations strongly suggest that Kv4 channel gating is tightly coupled to voltage-dependent accessibility changes of native T1 cysteines in the intersubunit Zn(2+) site. Furthermore, cross-linking of cysteine pairs across the T1-T1 interface induced substantial inhibition of the channel, which supports the functionally dynamic role of T1 in channel gating. Therefore, we conclude that the complex voltage-dependent gating rearrangements of eukaryotic Kv channels are not limited to the membrane-spanning core but must include the intracellular T1-T1 interface. Oxidative stress in excitable tissues may perturb this interface to modulate Kv4 channel function.


Asunto(s)
Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Canales de Potasio con Entrada de Voltaje/genética , Compuestos de Sulfhidrilo/metabolismo , Animales , Sitios de Unión , Cisteína/química , Dimerización , Proteínas de Interacción con los Canales Kv/química , Canal de Potasio Kv1.4/química , Potenciales de la Membrana , Mesilatos/farmacología , Técnicas de Placa-Clamp , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Compuestos de Sulfhidrilo/química , Xenopus laevis
16.
Circulation ; 113(3): 345-55, 2006 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-16432066

RESUMEN

BACKGROUND: Sustained heart rate abnormalities produce electrical remodeling and susceptibility to arrhythmia. Uncontrolled tachycardia produces heart failure and ventricular tachyarrhythmia susceptibility, whereas bradycardia promotes spontaneous torsade de pointes (TdP). This study compared arrhythmic phenotypes and molecular electrophysiological remodeling produced by tachycardia versus bradycardia in rabbits. METHODS AND RESULTS: We evaluated mRNA and protein expression of subunits underlying rapid (IKr) and slow (IKs) delayed-rectifier and transient-outward K+ currents in ventricular tissues from sinus rhythm control rabbits and rabbits with AV block submitted to 3-week ventricular pacing either at 60 to 90 bpm (bradypaced) or at 350 to 370 bpm (tachypaced). QT intervals at matched ventricular pacing rates were longer in bradypaced than tachypaced rabbits (eg, by approximately 50% at 60 bpm; P<0.01). KvLQT1 and minK mRNA and protein levels were downregulated in both bradypaced and tachypaced rabbits, whereas ERG was significantly downregulated in bradypaced rabbits only. Kv4.3 and Kv1.4 were downregulated by tachypacing only. Patch-clamp experiments showed that IKs was reduced in both but IKr was decreased in bradypaced rabbits only. Continuous monitoring revealed spontaneous TdP in 75% of bradypaced but only isolated ventricular ectopy in tachypaced rabbits. Administration of dofetilide (0.02 mg/kg) to mimic IKr downregulation produced ultimately lethal TdP in all tachypaced rabbits. CONCLUSIONS: Sustained tachycardia and bradycardia downregulate IKs subunits, but bradycardia also suppresses ERG/IKr, causing prominent repolarization delays and spontaneous TdP. Susceptibility of tachycardia/heart failure rabbits to malignant tachyarrhythmias is induced by exposure to IKr blockers. These results point to a crucial role for delayed-rectifier subunit remodeling in TdP susceptibility associated with rate-related cardiac remodeling.


Asunto(s)
Bradicardia/fisiopatología , Canales de Potasio de Tipo Rectificador Tardío/genética , Canales de Potasio de Tipo Rectificador Tardío/fisiología , Taquicardia Ventricular/fisiopatología , Animales , Bradicardia/diagnóstico , Canales de Potasio de Tipo Rectificador Tardío/química , Regulación hacia Abajo , Electrocardiografía , Canal de Potasio KCNQ1/química , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/fisiología , Canal de Potasio Kv1.4/química , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/fisiología , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/fisiopatología , Marcapaso Artificial , Técnicas de Placa-Clamp , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Conejos , Canales de Potasio Shal/química , Canales de Potasio Shal/genética , Canales de Potasio Shal/fisiología , Taquicardia Ventricular/diagnóstico , Torsades de Pointes/diagnóstico , Torsades de Pointes/fisiopatología
17.
J Physiol Biochem ; 62(4): 263-70, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17615952

RESUMEN

Interactions between antiarrhythmic drugs and ion channels are important subjects in the field of cardiovascular electro-pharmacology. This study explores the relationship between propafenone and C-type inactivation of Kv1.4 channel. fKvl.4deltaN, a ferret Kv1.4 N-terminal deleted mutant, was employed in this study. fKvl.4deltaN cRNA was injected into Xenopus oocytes to express fKvl.4deltaN channel and two electrode voltage clamp technique was used to record the current. We found that fKvl.4deltaN channel current was rapidly depressed in a frequency-dependent manner and meanwhile, C-type inactivation in this channel was increased more than 7 folds in the presence of 100 microM propafenone. While propafenone has no effect on Kv1.4deltaN recovery. All the results indicate that propafenone blocks Kvl.4deltaN channel through intracellular bindings and that binding of propafenone with Kvl.4deltaN channel leads to a conformational change on the extracellular site which accelerates C-type inactivation, suggesting that propafenone, as an open channel blocker, may affect the mechanism of C-type inactivation.


Asunto(s)
Antiarrítmicos/farmacología , Canal de Potasio Kv1.4/antagonistas & inhibidores , Propafenona/farmacología , Animales , Femenino , Activación del Canal Iónico , Cinética , Canal de Potasio Kv1.4/fisiología , Xenopus
18.
Mol Hum Reprod ; 11(12): 891-7, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16421215

RESUMEN

Fertility depends in part on the ability of the spermatozoon to respond to osmotic challenges by regulating its volume, which may rely on the movement of K+. These experiments were designed to characterize the K+ channels possibly involved in volume regulation of human ejaculated spermatozoa by simultaneously exposing them to a physiological hypo-osmotic challenge and a wide range of K+ channel inhibitors. Regulation of cellular volume, as measured by flow cytometry, was inhibited when spermatozoa were exposed to quinine (QUI; 0.3 mM), 4-aminopyridine (4AP; 4 mM) and clofilium (CLO; 10 microM) which suggests the involvement of voltage-gated K+ channels Kv1.4, Kv1.5 and Kv1.7, acid-sensitive channel TASK2 and the beta-subunit minK (IsK) in regulatory volume decrease (RVD). QUI and 4AP and, to some extent, CLO also induced hyper activation-like motility. A sensitivity of RVD to pH could not be demonstrated in spermatozoa to support the involvement of TASK2 channels. Western blotting indicated the presence of Kv1.5, TASK2, TASK3 and minK channel proteins, but not Kv1.4. Furthermore, Kv1.5, minK and TASK2 were localized to various regions of the spermatozoa. Although Kv1.4, Kv1.7, TASK2 and TASK3 channels may have important roles in human spermatozoa, Kv1.5 and minK appear to be the most likely candidates for human sperm RVD, serving as targets for non-hormonal contraception.


Asunto(s)
Canales de Potasio/fisiología , Espermatozoides/citología , Espermatozoides/fisiología , 4-Aminopiridina/farmacología , Tamaño de la Célula , Citometría de Flujo , Humanos , Canal de Potasio Kv1.4/efectos de los fármacos , Canal de Potasio Kv1.4/fisiología , Canal de Potasio Kv1.5/efectos de los fármacos , Canal de Potasio Kv1.5/fisiología , Masculino , Canales de Potasio/efectos de los fármacos , Compuestos de Amonio Cuaternario/farmacología , Quinina/farmacología , Canales de Potasio de la Superfamilia Shaker/efectos de los fármacos , Canales de Potasio de la Superfamilia Shaker/fisiología , Espermatozoides/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...