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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 296: 100381, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33556372

RESUMEN

Calcium-/voltage-gated, large-conductance potassium channels (BKs) control critical physiological processes, including smooth muscle contraction. Numerous observations concur that elevated membrane cholesterol (CLR) inhibits the activity of homomeric BKs consisting of channel-forming alpha subunits. In mammalian smooth muscle, however, native BKs include accessory KCNMB1 (ß1) subunits, which enable BK activation at physiological intracellular calcium. Here, we studied the effect of CLR enrichment on BK currents from rat cerebral artery myocytes. Using inside-out patches from middle cerebral artery (MCA) myocytes at [Ca2+]free=30 µM, we detected BK activation in response to in vivo and in vitro CLR enrichment of myocytes. While a significant increase in myocyte CLR was achieved within 5 min of CLR in vitro loading, this brief CLR enrichment of membrane patches decreased BK currents, indicating that BK activation by CLR requires a protracted cellular process. Indeed, blocking intracellular protein trafficking with brefeldin A (BFA) not only prevented BK activation but led to channel inhibition upon CLR enrichment. Surface protein biotinylation followed by Western blotting showed that BFA blocked the increase in plasmalemmal KCNMB1 levels achieved via CLR enrichment. Moreover, CLR enrichment of arteries with naturally high KCNMB1 levels, such as basilar and coronary arteries, failed to activate BK currents. Finally, CLR enrichment failed to activate BK channels in MCA myocytes from KCNMB1-/- mouse while activation was detected in their wild-type (C57BL/6) counterparts. In conclusion, the switch in CLR regulation of BK from inhibition to activation is determined by a trafficking-dependent increase in membrane levels of KCNMB1 subunits.


Asunto(s)
Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Células Musculares/metabolismo , Canales de Potasio/metabolismo , Animales , Canales de Calcio/metabolismo , Membrana Celular/metabolismo , Arterias Cerebrales/citología , Arterias Cerebrales/metabolismo , Colesterol/metabolismo , Colesterol/fisiología , Vasos Coronarios/metabolismo , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Canales de Potasio/fisiología , Ratas , Ratas Sprague-Dawley , Vasoconstricción
2.
Proc Natl Acad Sci U S A ; 113(23): E3231-9, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27217576

RESUMEN

Large-conductance Ca(2+)- and voltage-activated K(+) (BK) channels are involved in a large variety of physiological processes. Regulatory ß-subunits are one of the mechanisms responsible for creating BK channel diversity fundamental to the adequate function of many tissues. However, little is known about the structure of its voltage sensor domain. Here, we present the external architectural details of BK channels using lanthanide-based resonance energy transfer (LRET). We used a genetically encoded lanthanide-binding tag (LBT) to bind terbium as a LRET donor and a fluorophore-labeled iberiotoxin as the LRET acceptor for measurements of distances within the BK channel structure in a living cell. By introducing LBTs in the extracellular region of the α- or ß1-subunit, we determined (i) a basic extracellular map of the BK channel, (ii) ß1-subunit-induced rearrangements of the voltage sensor in α-subunits, and (iii) the relative position of the ß1-subunit within the α/ß1-subunit complex.


Asunto(s)
Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Animales , Transferencia de Energía , Femenino , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/química , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Modelos Moleculares , Oocitos , Conformación Proteica , Dominios Proteicos , Xenopus laevis
3.
Acta Pharmacol Sin ; 39(3): 371-381, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29188803

RESUMEN

Thiazide-like diuretics are the most commonly used drugs to treat arterial hypertension, with their efficacy being linked to their chronic vasodilatory effect. Previous studies suggest that activation of the large conductance voltage- and Ca2+-dependent K+ (BK) channel (Slo 1, MaxiK channel) is responsible for the thiazide-induced vasodilatory effect. But the direct electrophysiological evidence supporting this claim is lacking. BK channels can be associated with one small accessory ß-subunit (ß1-ß4) that confers specific biophysical and pharmacological characteristics to the current phenotype. The ß1-subunit is primarily expressed in smooth muscle cells (SMCs). In this study we investigated the effect of hydrochlorothiazide (HCTZ) on BK channel activity in native SMCs from human umbilical artery (HUASMCs) and HEK293T cells expressing the BK channel (with and without the ß1-subunit). Bath application of HCTZ (10 µmol/L) significantly augmented the BK current in HUASMCs when recorded using the whole-cell configurations, but it did not affect the unitary conductance and open probability of the BK channel in HUASMCs evaluated in the inside-out configuration, suggesting an indirect mechanism requiring cell integrity. In HEK293T cells expressing BK channels, HCTZ-augmented BK channel activity was only observed when the ß1-subunit was co-expressed, being concentration-dependent with an EC50 of 28.4 µmol/L, whereas membrane potential did not influence the concentration relationship. Moreover, HCTZ did not affect the BK channel current in HEK293T cells evaluated in the inside-out configuration, but significantly increases the open probability in the cell-attached configuration. Our data demonstrate that a ß1-subunit-dependent mechanism that requires SMC integrity leads to HCTZ-induced BK channel activation.


Asunto(s)
Hidroclorotiazida/farmacología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Células Cultivadas , Humanos , Potenciales de la Membrana/efectos de los fármacos
4.
Proc Natl Acad Sci U S A ; 112(15): 4809-14, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25825713

RESUMEN

Being activated by depolarizing voltages and increases in cytoplasmic Ca(2+), voltage- and calcium-activated potassium (BK) channels and their modulatory ß-subunits are able to dampen or stop excitatory stimuli in a wide range of cellular types, including both neuronal and nonneuronal tissues. Minimal alterations in BK channel function may contribute to the pathophysiology of several diseases, including hypertension, asthma, cancer, epilepsy, and diabetes. Several gating processes, allosterically coupled to each other, control BK channel activity and are potential targets for regulation by auxiliary ß-subunits that are expressed together with the α (BK)-subunit in almost every tissue type where they are found. By measuring gating currents in BK channels coexpressed with chimeras between ß1 and ß3 or ß2 auxiliary subunits, we were able to identify that the cytoplasmic regions of ß1 are responsible for the modulation of the voltage sensors. In addition, we narrowed down the structural determinants to the N terminus of ß1, which contains two lysine residues (i.e., K3 and K4), which upon substitution virtually abolished the effects of ß1 on charge movement. The mechanism by which K3 and K4 stabilize the voltage sensor is not electrostatic but specific, and the α (BK)-residues involved remain to be identified. This is the first report, to our knowledge, where the regulatory effects of the ß1-subunit have been clearly assigned to a particular segment, with two pivotal amino acids being responsible for this modulation.


Asunto(s)
Calcio/metabolismo , Activación del Canal Iónico/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Potasio/metabolismo , Animales , Sitios de Unión/genética , Femenino , Humanos , Activación del Canal Iónico/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Lisina/química , Lisina/genética , Lisina/fisiología , Potenciales de la Membrana/genética , Potenciales de la Membrana/fisiología , Modelos Moleculares , Mutación , Oocitos/metabolismo , Oocitos/fisiología , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Xenopus laevis
5.
Pharmacol Res ; 107: 186-194, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26995303

RESUMEN

The present study was designed to investigate the effect of equol on cerebral blood flow and the underlying molecular mechanisms. The regional cerebral blood flow in parietal lobe of rats was measured by using a laser Doppler flowmetry. Isolated cerebral basilar artery and mesenteric artery rings from rats were used for vascular reactivity measurement with a multi wire myography system. Outward K(+) current in smooth muscle cells of cerebral basilar artery, large-conductance Ca(2+)-activated K(+) (BK) channel current in BK-HEK 293 cells stably expressing both human α (hSlo)- and ß1-subunits, and hSlo channel current in hSlo-HEK 293 cells expressing only the α-subunit of BK channels were recorded with whole cell patch-clamp technique. The results showed that equol significantly increased regional cerebral blood flow in rats, and produced a concentration-dependent but endothelium-independent relaxation in rat cerebral basilar arteries. Both paxilline and iberiotoxin, two selective BK channel blockers, significantly inhibited equol-induced vasodilation in cerebral arteries. Outward K(+) currents in smooth muscle cells of cerebral basilar artery were increased by equol and fully reversed by washout or blockade of BK channels with iberiotoxin. Equol remarkably enhanced human BK current in BK-HEK 293 cells, but not hSlo current in hSlo-HEK 293 cells, and the increase was completely abolished by co-application of paxilline. Our findings provide the first information that equol selectively stimulates BK channel current by acting on its ß1 subunit, which may in turn contribute to the equol-mediated vasodilation and cerebral blood flow increase.


Asunto(s)
Circulación Cerebrovascular/efectos de los fármacos , Equol/farmacología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/fisiología , Células HEK293 , Humanos , Técnicas In Vitro , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Ratas , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos
6.
J Neurosci ; 33(27): 11253-61, 2013 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-23825428

RESUMEN

Large-conductance, voltage-, and Ca²âº-dependent K⁺ (BK) channels are broadly expressed in various tissues to modulate neuronal activity, smooth muscle contraction, and secretion. BK channel activation depends on the interactions among the voltage sensing domain (VSD), the cytosolic domain (CTD), and the pore gate domain (PGD) of the Slo1 α-subunit, and is further regulated by accessory ß subunits (ß1-ß4). How ß subunits fine-tune BK channel activation is critical to understand the tissue-specific functions of BK channels. Multiple sites in both Slo1 and the ß subunits have been identified to contribute to the interaction between Slo1 and the ß subunits. However, it is unclear whether and how the interdomain interactions among the VSD, CTD, and PGD are altered by the ß subunits to affect channel activation. Here we show that human ß1 and ß2 subunits alter interactions between bound Mg²âº and gating charge R213 and disrupt the disulfide bond formation at the VSD-CTD interface of mouse Slo1, indicating that the ß subunits alter the VSD-CTD interface. Reciprocally, mutations in the Slo1 that alter the VSD-CTD interaction can specifically change the effects of the ß1 subunit on the Ca²âº activation and of the ß2 subunit on the voltage activation. Together, our data suggest a novel mechanism by which the ß subunits modulated BK channel activation such that a ß subunit may interact with the VSD or the CTD and alter the VSD-CTD interface of the Slo1, which enables the ß subunit to have effects broadly on both voltage and Ca²âº-dependent activation.


Asunto(s)
Membrana Celular/metabolismo , Citosol/metabolismo , Activación del Canal Iónico/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Animales , Membrana Celular/química , Citosol/química , Femenino , Humanos , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Ratones , Estructura Terciaria de Proteína , Xenopus laevis
7.
J Physiol ; 592(12): 2563-74, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24687584

RESUMEN

The large conductance voltage- and Ca(2+)-activated K(+) (BK) channel is an important determinant of vascular tone and contributes to blood pressure regulation. Both activities depend on the ancillary BKß1 subunit. To determine the significance of smooth muscle BK channel activity for blood pressure regulation, we investigated the potential link between changes in arterial tone and altered blood pressure in BKß1 knockout (BKß1(-/-)) mice from three different genetically defined strains. While vascular tone was consistently increased in all BKß1(-/-) mice independent of genetic background, BKß1(-/-) strains exhibited increased (strain A), unaltered (strain B) or decreased (strain C) mean arterial blood pressures compared to their corresponding BKß1(+/+) controls. In agreement with previous data on aldosterone regulation by renal/adrenal BK channel function, BKß1(-/-) strain A mice have increased plasma aldosterone and increased blood pressure. Consistently, blockade of mineralocorticoid receptors by spironolactone treatment reversibly restored the elevated blood pressure to the BKß1(+/+) strain A level. In contrast, loss of BKß1 did not affect plasma aldosterone in strain C mice. Smooth muscle-restricted restoration of BKß1 expression increased blood pressure in BKß1(-/-) strain C mice, implying that impaired smooth muscle BK channel activity lowers blood pressure in these animals. We conclude that BK channel activity directly affects vascular tone but influences blood pressure independent of this effect via different pathways.


Asunto(s)
Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Músculo Liso/fisiología , Aldosterona/sangre , Animales , Aorta Torácica/fisiología , Presión Sanguínea/fisiología , Técnicas In Vitro , Riñón/fisiología , Ratones Transgénicos , Células Musculares/fisiología , Oocitos/fisiología , Xenopus
8.
J Biol Chem ; 287(46): 38552-8, 2012 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-23002235

RESUMEN

The epithelial Na(+) channel (ENaC) in the aldosterone-sensitive distal nephron (ASDN) is under tonic inhibition by a local purinergic signaling system responding to changes in dietary sodium intake. Normal BK(Ca) channel function is required for flow-sensitive ATP secretion in the ASDN. We tested here whether ATP secreted through connexin channels in a coupled manner with K(+) efflux through BK(Ca) channels is required for inhibitory purinergic regulation of ENaC in response to increases in sodium intake. Inhibition of connexin channels relieves purinergic inhibition of ENaC. Deletion of the BK-ß4 regulatory subunit, which is required for normal BK(Ca) channel function and flow-sensitive ATP secretion in the ASDN, suppresses increases in urinary ATP in response to increases in sodium intake. As a consequence, ENaC activity, particularly in the presence of high sodium intake, is inappropriately elevated in BK-ß4 null mice. ENaC in BK-ß4 null mice, however, responds normally to exogenous ATP, indicating that increases in activity do not result from end-organ resistance but rather from lowered urinary ATP. Consistent with this, disruption of purinergic regulation increases ENaC activity in wild type but not BK-ß4 null mice. Consequently, sodium excretion is impaired in BK-ß4 null mice. These results demonstrate that the ATP secreted in the ASDN in a BK(Ca) channel-dependent manner is physiologically available for purinergic inhibition of ENaC in response to changes in sodium homeostasis. Impaired sodium excretion resulting form loss of normal purinergic regulation of ENaC in BK-ß4 null mice likely contributes to their elevated blood pressure.


Asunto(s)
Adenosina Trifosfato/química , Canales Epiteliales de Sodio/metabolismo , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Nefronas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Presión Sanguínea , Conexinas/química , Homeostasis , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Placa-Clamp , Sodio/química , Sodio/metabolismo , Sodio en la Dieta/metabolismo
9.
J Cardiovasc Pharmacol ; 59(1): 29-36, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21885988

RESUMEN

We determined the possible role of large-conductance Ca2+-activated K (BK) channels in regulation of venous tone in small capacitance veins and blood pressure. In rat mesenteric venous smooth muscle cells (MV SMC), BK channel α- and ß1-subunits were coexpressed, unitary BK currents were detected, and single-channel currents were sensitive to voltage and [Ca2+]i. Rat MV SMCs displayed Ca sparks and iberiotoxin-sensitive spontaneous transient outward currents. Under resting conditions in vitro, rat MV exhibited nifedipine-sensitive spontaneous oscillatory constrictions. Blockade of BK channels by paxilline and Ca2+ sparks by ryanodine constricted rat MV. Nifedipine caused venodilation and blocked paxilline-induced, KCl-induced (20 mM), and BayK8644-induced contraction. Acute inhibition of BK channels with iberiotoxin in vivo increased blood pressure and reduced venous capacitance, measured as an increase in mean circulatory filling pressure in conscious rats. BK channel α-subunits and L-type Ca2+ channel α1-C subunits are expressed in murine MV. However, these channels are not functional because murine MV lack nifedipine-sensitive basal tone and rhythmic constrictions. Murine MV were also insensitive to paxilline, ryanodine, KCl, and BayK8644, consistent with our previous studies showing that murine MV do not have BK ß1-subunits. These data show that not only there are species-dependent properties in ion channel control of venomotor tone but also BK channels are required for rhythmic oscillations in venous tone.


Asunto(s)
Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Venas Mesentéricas/metabolismo , Contracción Muscular/fisiología , Músculo Liso Vascular/metabolismo , Vasodilatación/fisiología , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Células Cultivadas , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/antagonistas & inhibidores , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/biosíntesis , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/biosíntesis , Masculino , Venas Mesentéricas/efectos de los fármacos , Venas Mesentéricas/fisiopatología , Ratones , Ratones Endogámicos C57BL , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Vasodilatación/efectos de los fármacos
10.
J Neurosci ; 30(48): 16170-9, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21123563

RESUMEN

Large-conductance, Ca(2+)- and voltage-sensitive K(+) (BK) channels regulate neuronal functions such as spike frequency adaptation and transmitter release. BK channels are composed of four Slo1 subunits, which contain the voltage-sensing and pore-gate domains in the membrane and Ca(2+) binding sites in the cytoplasmic domain, and accessory ß subunits. Four types of BK channel ß subunits (ß1-ß4) show differential tissue distribution and unique functional modulation, resulting in diverse phenotypes of BK channels. Previous studies show that both the ß1 and ß2 subunits increase Ca(2+) sensitivity, but different mechanisms may underline these modulations. However, the structural domains in Slo1 that are critical for Ca(2+)-dependent activation and targeted by these ß subunits are not known. Here, we report that the N termini of both the transmembrane (including S0) and cytoplasmic domains of Slo1 are critical for ß2 modulation based on the study of differential effects of the ß2 subunit on two orthologs, mouse Slo1 and Drosophila Slo1. The N terminus of the cytoplasmic domain of Slo1, including the AC region (ßA-αC) of the RCK1 (regulator of K(+) conductance) domain and the peptide linking it to S6, both of which have been shown previously to mediate the coupling between Ca(2+) binding and channel opening, is specifically required for the ß2 but not for the ß1 modulation. These results suggest that the ß2 subunit modulates the coupling between Ca(2+) binding and channel opening, and, although sharing structural homology, the BK channel ß subunits interact with structural domains in the Slo1 subunit differently to enhance channel activity.


Asunto(s)
Membrana Celular/química , Membrana Celular/fisiología , Citoplasma/fisiología , Activación del Canal Iónico/fisiología , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/química , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Secuencia de Aminoácidos , Animales , Membrana Celular/genética , Citoplasma/química , Drosophila , Femenino , Humanos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Ratones , Datos de Secuencia Molecular , Proteínas Mutantes Quiméricas/química , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/fisiología , Estructura Terciaria de Proteína/genética , Xenopus laevis
11.
J Physiol ; 589(Pt 7): 1803-17, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21300746

RESUMEN

The large conductance calcium- and voltage-activated potassium channel (BK channel) and its smooth muscle-specific ß1 subunit regulate excitation­contraction coupling in many types of smooth muscle cells. However, the relative contribution of BK channels to control of M2- or M3-muscarinic acetylcholine receptor mediated airway smooth muscle contraction is poorly understood. Previously, we showed that knockout of the BK channel ß1 subunit enhances cholinergic-evoked trachea contractions. Here, we demonstrate that the enhanced contraction of the BK ß1 knockout can be ascribed to a defect in BK channel opposition of M2 receptor-mediated contractions. Indeed, the enhanced contraction of ß1 knockout is eliminated by specific M2 receptor antagonism. The role of BK ß1 to oppose M2 signalling is evidenced by a greater than fourfold increase in the contribution of L-type voltage-dependent calcium channels to contraction that otherwise does not occur with M2 antagonist or with ß1 containing BK channels. The mechanism through which BK channels oppose M2-mediated recruitment of calcium channels is through a negative shift in resting voltage that offsets, rather than directly opposes, M2-mediated depolarization. The negative shift in resting voltage is reduced to similar extents by BK ß1 knockout or by paxilline block of BK channels. Normalization of ß1 knockout baseline voltage with low external potassium eliminated the enhanced M2-receptor mediated contraction. In summary, these findings indicate that an important function of BK/ß1 channels is to oppose cholinergic M2 receptor-mediated depolarization and activation of calcium channels by restricting excitation­contraction coupling to more negative voltage ranges.


Asunto(s)
Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Contracción Muscular/fisiología , Receptor Muscarínico M2/fisiología , Tráquea/fisiología , Animales , Canales de Calcio Tipo L/fisiología , Acoplamiento Excitación-Contracción/efectos de los fármacos , Acoplamiento Excitación-Contracción/fisiología , Técnicas In Vitro , Indoles/farmacología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/deficiencia , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Antagonistas Muscarínicos/farmacología , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Músculo Liso/fisiología , Pirenzepina/análogos & derivados , Pirenzepina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Receptor Muscarínico M2/antagonistas & inhibidores , Mecánica Respiratoria , Transducción de Señal , Tráquea/efectos de los fármacos
12.
Am J Physiol Heart Circ Physiol ; 300(2): H507-13, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21112945

RESUMEN

Chronic hypoxia protects the heart against injury caused by acute oxygen deprivation, but its salutary mechanism is poorly understood. The aim was to find out whether cardiomyocytes isolated from chronically hypoxic hearts retain the improved resistance to injury and whether the mitochondrial large-conductance Ca2+-activated K+ (BKCa) channels contribute to the protective effect. Adult male rats were adapted to continuous normobaric hypoxia (inspired O2 fraction 0.10) for 3 wk or kept at room air (normoxic controls). Myocytes, isolated separately from the left ventricle (LVM), septum (SEPM), and right ventricle, were exposed to 25-min metabolic inhibition with sodium cyanide, followed by 30-min reenergization (MI/R). Some LVM were treated with either 30 µM NS-1619 (BKCa opener), or 2 µM paxilline (BKCa blocker), starting 25 min before metabolic inhibition. Cell injury was detected by Trypan blue exclusion and lactate dehydrogenase (LDH) release. Chronic hypoxia doubled the number of rod-shaped LVM and SEPM surviving the MI/R insult and reduced LDH release. While NS-1619 protected cells from normoxic rats, it had no additive salutary effect in the hypoxic group. Paxilline attenuated the improved resistance of cells from hypoxic animals without affecting normoxic controls; it also abolished the protective effect of NS-1619 on LDH release in the normoxic group. While chronic hypoxia did not affect protein abundance of the BKCa channel regulatory ß1-subunit, it markedly decreased its glycosylation level. It is concluded that ventricular myocytes isolated from chronically hypoxic rats retain the improved resistance against injury caused by MI/R. Activation of the mitochondrial BKCa channel likely contributes to this protective effect.


Asunto(s)
Hipoxia/fisiopatología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Mitocondrias Cardíacas/fisiología , Miocitos Cardíacos/fisiología , Animales , Bencimidazoles/farmacología , Western Blotting , Separación Celular , Supervivencia Celular , Células Cultivadas , Enfermedad Crónica , Glicosilación , Indoles/farmacología , Precondicionamiento Isquémico Miocárdico , L-Lactato Deshidrogenasa/metabolismo , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Daño por Reperfusión Miocárdica/fisiopatología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Wistar
13.
Am J Physiol Heart Circ Physiol ; 300(2): H476-85, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21131476

RESUMEN

Large-conductance Ca2+-activated K+ (BK) channels are composed of pore-forming α-subunits and accessory ß1-subunits that modulate Ca2+ sensitivity. BK channels regulate arterial myogenic tone and renal Na+ clearance/K+ reabsorption. Previous studies using indirect or short-term blood pressure measurements found that BK channel ß1-subunit knockout (BK ß1-KO) mice were hypertensive. We evaluated 24-h mean arterial pressure (MAP) and heart rate in BK ß1-KO mice using radiotelemetry. BK ß1-KO mice did not have a higher 24-h average MAP when compared with wild-type (WT) mice, although MAP was ∼10 mmHg higher at night. The dose-dependent peak declines in MAP by nifedipine were only slightly larger in BK ß1-KO mice. In BK ß1-KO mice, giving 1% NaCl to mice to drink for 7 days caused a transient (5 days) elevation of MAP (∼5 mmHg); MAP returned to pre-saline levels by day 6. BK ß1-KO mesenteric arteries in vitro demonstrated diminished contractile responses to paxilline, increased reactivity to Bay K 8644 and norepinephrine (NE), and maintained relaxation to isoproterenol. Paxilline and Bay K 8644 did not constrict WT or BK ß1-KO mesenteric veins (MV). BK ß1-subunits are not expressed in MV. The results indicate that BK ß1-KO mice are not hypertensive on normal or high-salt intake. BK channel deficiency increases arterial reactivity to NE and L-type Ca2+ channel function in vitro, but the L-type Ca2+ channel modulation of MAP is not altered in BK ß1-KO mice. BK and L-type Ca(2+) channels do not modulate murine venous tone. It appears that selective loss of BK channel function in arteries only is not sufficient to cause sustained hypertension.


Asunto(s)
Hipertensión/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Presión Sanguínea/fisiología , Peso Corporal/fisiología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/fisiología , Corazón/anatomía & histología , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/genética , Frecuencia Cardíaca/fisiología , Hipertensión/fisiopatología , Operón Lac/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Arterias Mesentéricas/fisiología , Venas Mesentéricas/efectos de los fármacos , Venas Mesentéricas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Muscular/genética , Contracción Muscular/fisiología , Relajación Muscular/genética , Relajación Muscular/fisiología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Nifedipino/farmacología , Tamaño de los Órganos/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Vasodilatación/genética , Vasodilatación/fisiología
14.
J Am Soc Nephrol ; 21(4): 634-45, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20299355

RESUMEN

The large-conductance, calcium-activated potassium (BK) channels help eliminate potassium in mammals consuming potassium-rich diets. In the distal nephron, principal cells contain BK-alpha/beta1 channels and intercalated cells contain BK-alpha/beta4 channels. We studied whether BK-beta4-deficient mice (Kcnmb4(-/-)) have altered renal sodium and potassium clearances compared with wild-type mice when fed a regular or potassium-rich diet for ten days. We did not detect differences in urinary flow or fractional excretions of potassium (FE(K)) or sodium (FE(Na)) between Kcnmb4-deficient and wild-type mice fed a regular diet. However, a potassium-rich diet led to >4-fold increases in urinary flows for both groups of mice, although Kcnmb4-deficient mice exhibited less urinary flow, higher plasma potassium concentration, more fluid retention, and significantly lower FE(K) and FE(Na) than wild-type mice despite similar plasma aldosterone levels. Immunohistochemical analysis revealed increased basolateral Na-K-ATPase in principal cells of all potassium-adapted mice, but expression of Na-K-ATPase in intercalated cells was >10-fold lower. The size of intercalated cells reduced and luminal volume increased among potassium-adapted wild-type but not Kcnmb4-deficient mice. Paradoxically, this led to increased urinary fluid velocity in potassium-adapted Kcnmb4-deficient mice compared with wild-type mice. Taken together, these data suggest that BK-alpha/beta4 channels in intercalated cells reduce cell size, increasing luminal volume to accommodate higher distal flow rates during potassium adaptation. These changes streamline flow across the principal cells, producing gradients more favorable for potassium secretion and less favorable for sodium reabsorption.


Asunto(s)
Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Potasio/metabolismo , Sodio/metabolismo , Adaptación Fisiológica , Animales , Ratones
15.
J Physiol ; 587(Pt 10): 2225-31, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19289549

RESUMEN

A number of potassium channels including members of the KCNQ family and the Ca(2+) activated IK and SK, but not BK, are strongly and reversibly regulated by small changes in cell volume. It has been argued that this general regulation is mediated through sensitivity to changes in membrane stretch. To test this hypothesis we have studied the regulation of KCNQ1 and BK channels after expression in Xenopus oocytes. Results from cell-attached patch clamp studies (approximately 50 microm(2) macropatches) in oocytes expressing BK channels demonstrate that the macroscopic volume-insensitive BK current increases with increasing negative hydrostatic pressure (suction) applied to the pipette. Thus, at a pipette pressure of -5.0 +/- 0.1 mmHg the increase amounted to 381 +/- 146% (mean +/- S.E.M., n = 6, P < 0.025). In contrast, in oocytes expressing the strongly volume-sensitive KCNQ1 channel, the current was not affected by membrane stretch. The results indicate that (1) activation of BK channels by local membrane stretch is not mimicked by membrane stress induced by cell swelling, and (2) activation of KCNQ1 channels by cell volume increase is not mediated by local tension in the cell membrane. We conclude that stretch and volume sensitivity can be considered two independent regulatory mechanisms.


Asunto(s)
Membrana Celular/fisiología , Tamaño de la Célula , Mecanotransducción Celular/fisiología , Canales de Potasio/fisiología , Animales , Línea Celular , Estimulación Eléctrica , Fenómenos Electrofisiológicos/fisiología , Femenino , Humanos , Presión Hidrostática , Soluciones Hipertónicas/farmacología , Soluciones Isotónicas/farmacología , Canales de Potasio KCNQ/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Potenciales de la Membrana/fisiología , Oocitos/metabolismo , Transfección , Xenopus laevis
16.
J Pharmacol Exp Ther ; 329(3): 978-86, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19321803

RESUMEN

Neurons are highly differentiated and polarized cells, whose various functions depend upon the compartmentalization of ion channels. The rat hypothalamic-neurohypophysial system (HNS), in which cell bodies and dendrites reside in the hypothalamus, physically separated from their nerve terminals in the neurohypophysis, provides a particularly powerful preparation in which to study the distribution and regional properties of ion channel proteins. Using electrophysiological and immunohistochemical techniques, we characterized the large-conductance calcium-activated potassium (BK) channel in each of the three primary compartments (soma, dendrite, and terminal) of HNS neurons. We found that dendritic BK channels, in common with somatic channels but in contrast to nerve terminal channels, are insensitive to iberiotoxin. Furthermore, analysis of dendritic BK channel gating kinetics indicates that they, like somatic channels, have fast activation kinetics, in contrast to the slow gating of terminal channels. Dendritic and somatic channels are also more sensitive to calcium and have a greater conductance than terminal channels. Finally, although terminal BK channels are highly potentiated by ethanol, somatic and dendritic channels are insensitive to the drug. The biophysical and pharmacological properties of somatic and dendritic versus nerve terminal channels are consistent with the characteristics of exogenously expressed alphabeta1 versus alphabeta4 channels, respectively. Therefore, one possible explanation for our findings is a selective distribution of auxiliary beta1 subunits to the somatic and dendritic compartments and beta4 to the terminal compartment. This hypothesis is supported immunohistochemically by the appearance of distinct punctate beta1 or beta4 channel clusters in the membrane of somatic and dendritic or nerve terminal compartments, respectively.


Asunto(s)
Sistema Nervioso Central/metabolismo , Etanol/farmacología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Neuronas/metabolismo , Animales , Calcio/metabolismo , Calcio/farmacología , Dendritas/metabolismo , Sistema Hipotálamo-Hipofisario/citología , Sistema Hipotálamo-Hipofisario/metabolismo , Técnicas In Vitro , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Cinética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Gran Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Potenciales de la Membrana/fisiología , Neuronas/efectos de los fármacos , Péptidos/farmacología , Terminales Presinápticos/metabolismo , Ratas , Ratas Sprague-Dawley , Núcleo Supraóptico/citología , Núcleo Supraóptico/metabolismo , Toxinas Biológicas/farmacología
17.
Endocrinology ; 149(2): 774-82, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17962348

RESUMEN

Estrogens play essential roles in the neuroendocrine control of reproduction. In the present study, we focused on the effects of 17beta-estradiol (E2) on the K(+) currents that regulate neuronal cell excitability and carried out perforated patch-clamp experiments with the GnRH-secreting neuronal cell line GT1-7. We revealed that a 3-d incubation with E2 at physiological concentrations (100 pm to 1 nm) augmented Ca(2+)-activated K(+) [K(Ca)] currents without influencing Ca(2+)-insensitive voltage-gated K(+) currents in GT1-7 cells. Acute application of E2 (1 nm) had no effect on the either type of K(+) current. The augmentation was completely blocked by an estrogen receptor (ER) antagonist, ICI-182,780. An ERbeta-selective agonist, 2,3-bis(4-hydroxyphenyl)-propionitrile, augmented the K(Ca) currents, although an ERalpha-selective agonist, 4,4',4''-[4-propyl-(1H)-pyrazole-1,3,5-triyl]tris-phenol, had no effect. Knockdown of ERbeta by means of RNA interference blocked the effect of E2 on the K(Ca) currents. Furthermore, semiquantitative RT-PCR analysis revealed that the levels of BK channel subunit mRNAs for alpha and beta4 were significantly increased by incubating cells with 300 pm E2 for 3 d. In conclusion, E2 at physiological concentrations augments K(Ca) currents through ERbeta in the GT1-7 GnRH neuronal cell line and increases the expression of the BK channel subunit mRNAs, alpha and beta4.


Asunto(s)
Estradiol/farmacología , Receptor beta de Estrógeno/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Neuronas/fisiología , Animales , Calcio/farmacología , Línea Celular Transformada , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/fisiología , Receptor beta de Estrógeno/genética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Transgénicos , Neuronas/citología , Neuronas/efectos de los fármacos , Nitrilos/farmacología , Técnicas de Placa-Clamp , Potasio/metabolismo , Propionatos/farmacología , ARN Mensajero/metabolismo , ARN Interferente Pequeño
18.
J Gen Physiol ; 127(2): 119-31, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16418401

RESUMEN

Proteins arising from the Slo family assemble into homotetramers to form functional large-conductance, Ca2+- and voltage-activated K+ channels, or BK channels. These channels are also found in association with accessory beta subunits, which modulate several aspects of channel gating and expression. Coexpression with either of two such subunits, beta2 or beta3b, confers time-dependent inactivation onto BK currents. mSlo1+beta3b channels display inactivation that is very rapid but incomplete. Previous studies involving macroscopic recordings from these channels have argued for the existence of a second, short-lived conducting state in rapid equilibrium with the nonconducting, inactivated conformation. This state has been termed "pre-inactivated," or O*. beta2-mediated inactivation, in contrast, occurs more slowly but is virtually complete at steady state. Here we demonstrate, using both macroscopic and single channel current recordings, that a preinactivated state is also a property of mSlo1+beta2 channels. Detection of this state is enhanced by a mutation (W4E) within the initial beta2 NH2-terminal segment critical for inactivation. This mutation increases the rate of recovery to the preinactivated open state, yielding macroscopic inactivation properties qualitatively more similar to those of beta3b. Furthermore, short-lived openings corresponding to entry into the preinactivated state can be observed directly with single-channel recording. By examining the initial openings after depolarization of a channel containing beta2-W4E, we show that channels can arrive directly at the preinactivated state without passing through the usual long-lived open conformation. This final result suggests that channel opening and inactivation are at least partly separable in this channel. Mechanistically, the preinactivated and inactivated conformations may correspond to binding of the beta subunit NH2 terminus in the vicinity of the cytoplasmic pore mouth, followed by definitive movement of the NH2 terminus into a position of occlusion within the ion-conducting pathway.


Asunto(s)
Activación del Canal Iónico/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/química , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Animales , Conductividad Eléctrica , Femenino , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Mutación , Oocitos/fisiología , Técnicas de Placa-Clamp , Conformación Proteica , Canales de Potasio de la Superfamilia Shaker/química , Canales de Potasio de la Superfamilia Shaker/genética , Canales de Potasio de la Superfamilia Shaker/fisiología , Factores de Tiempo , Xenopus laevis
19.
J Gen Physiol ; 127(4): 449-65, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16567466

RESUMEN

Large-conductance (BK-type) Ca(2+)-activated potassium channels are activated by membrane depolarization and cytoplasmic Ca(2+). BK channels are expressed in a broad variety of cells and have a corresponding diversity in properties. Underlying much of the functional diversity is a family of four tissue-specific accessory subunits (beta1-beta4). Biophysical characterization has shown that the beta4 subunit confers properties of the so-called "type II" BK channel isotypes seen in brain. These properties include slow gating kinetics and resistance to iberiotoxin and charybdotoxin blockade. In addition, the beta4 subunit reduces the apparent voltage sensitivity of channel activation and has complex effects on apparent Ca(2+) sensitivity. Specifically, channel activity at low Ca(2+) is inhibited, while at high Ca(2+), activity is enhanced. The goal of this study is to understand the mechanism underlying beta4 subunit action in the context of a dual allosteric model for BK channel gating. We observed that beta4's most profound effect is a decrease in P(o) (at least 11-fold) in the absence of calcium binding and voltage sensor activation. However, beta4 promotes channel opening by increasing voltage dependence of P(o)-V relations at negative membrane potentials. In the context of the dual allosteric model for BK channels, we find these properties are explained by distinct and opposing actions of beta4 on BK channels. beta4 reduces channel opening by decreasing the intrinsic gating equilibrium (L(0)), and decreasing the allosteric coupling between calcium binding and voltage sensor activation (E). However, beta4 has a compensatory effect on channel opening following depolarization by shifting open channel voltage sensor activation (Vh(o)) to more negative membrane potentials. The consequence is that beta4 causes a net positive shift of the G-V relationship (relative to alpha subunit alone) at low calcium. At higher calcium, the contribution by Vh(o) and an increase in allosteric coupling to Ca(2+) binding (C) promotes a negative G-V shift of alpha+beta4 channels as compared to alpha subunits alone. This manner of modulation predicts that type II BK channels are downregulated by beta4 at resting voltages through effects on L(0). However, beta4 confers a compensatory effect on voltage sensor activation that increases channel opening during depolarization.


Asunto(s)
Calcio/metabolismo , Membrana Celular/fisiología , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Sitios de Unión , Calcio/farmacología , Línea Celular , Humanos , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp , Relación Estructura-Actividad , Transfección
20.
Neuroscience ; 149(4): 789-803, 2007 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-17945424

RESUMEN

Large conductance (BK-type) calcium-activated potassium channels utilize alternative splicing and association with accessory beta subunits to tailor BK channel properties to diverse cell types. Two important modulators of BK channel gating are the neuronal-specific beta4 accessory subunit (beta4) and alternative splicing at the stress axis hormone-regulated exon (STREX). Individually, these modulators affect the gating properties of the BK channel as well as its response to phosphorylation. In this study, the combined functional consequences of STREX and the mouse beta4 subunit on mouse BK channel biophysical properties were investigated in transfected HEK 293 cells. Surprisingly, we found that the combined effects of STREX and beta4 are non-additive and even opposite for some properties. At high calcium, beta4 and the STREX individually share properties that promote BK channel opening via slowing of deactivation. However, the combined effects are a speeding of deactivation and a decreased open probability. beta4 also inhibits BK channel opening by a slowing of activation. This effect occurs across calcium concentrations in the absence of STREX, but predominates only at low calcium for STREX containing channels. BK channel responses to phosphorylation status are also altered by the combination of the beta4 subunit and STREX. beta4/STREX channels show a slowing of activation kinetics following dephosphorylation whereas beta4 channels lacking STREX do not. In contrast, beta4 confers a speeding of activation in response to cyclic AMP-dependent phosphorylation in channels lacking STREX, but not in channels containing STREX. These results indicate that the combination of the beta4 subunit and STREX confers non-additive and unique properties to BK channels. Analysis of expression in brain slices suggests that STREX and beta4 mRNA overlap expression in the dentate gyrus of the hippocampus and the cerebellar Purkinje cells, suggesting that these unique properties of BK channels may underlie BK channel gating in these cells.


Asunto(s)
Calcio/metabolismo , Exones/fisiología , Activación del Canal Iónico/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Fosfatasa Alcalina/farmacología , Animales , Calcio/farmacología , Línea Celular Transformada , AMP Cíclico/farmacología , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/efectos de la radiación , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Potenciales de la Membrana/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Técnicas de Placa-Clamp/métodos , Fosforilación , Ratas , Transfección/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA