Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
PLoS One ; 17(1): e0261960, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35030226

RESUMEN

Inhibitory regulation of the heart is determined by both cholinergic M2 receptors (M2R) and adenosine A1 receptors (A1R) that activate the same signaling pathway, the ACh-gated inward rectifier K+ (KACh) channels via Gi/o proteins. Previously, we have shown that the agonist-specific voltage sensitivity of M2R underlies several voltage-dependent features of IKACh, including the 'relaxation' property, which is characterized by a gradual increase or decrease of the current when cardiomyocytes are stepped to hyperpolarized or depolarized voltages, respectively. However, it is unknown whether membrane potential also affects A1R and how this could impact IKACh. Upon recording whole-cell currents of guinea-pig cardiomyocytes, we found that stimulation of the A1R-Gi/o-IKACh pathway with adenosine only caused a very slight voltage dependence in concentration-response relationships (~1.2-fold EC50 increase with depolarization) that was not manifested in the relative affinity, as estimated by the current deactivation kinetics (τ = 4074 ± 214 ms at -100 mV and τ = 4331 ± 341 ms at +30 mV; P = 0.31). Moreover, IKACh did not exhibit relaxation. Contrarily, activation of the M2R-Gi/o-IKACh pathway with acetylcholine induced the typical relaxation of the current, which correlated with the clear voltage-dependent effect observed in the concentration-response curves (~2.8-fold EC50 increase with depolarization) and in the IKACh deactivation kinetics (τ = 1762 ± 119 ms at -100 mV and τ = 1503 ± 160 ms at +30 mV; P = 0.01). Our findings further substantiate the hypothesis of the agonist-specific voltage dependence of GPCRs and that the IKACh relaxation is consequence of this property.


Asunto(s)
Acetilcolina/farmacología , Agonistas del Receptor de Adenosina A1/farmacología , Adenosina/farmacología , Activación del Canal Iónico/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Canales de Potasio/metabolismo , Receptor de Adenosina A1/metabolismo , Animales , Femenino , Cobayas , Masculino , Receptor Muscarínico M2/agonistas , Receptor Muscarínico M2/metabolismo
2.
J Gen Physiol ; 153(5)2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33822868

RESUMEN

Inwardly rectifying potassium (Kir) channels are broadly expressed in both excitable and nonexcitable tissues, where they contribute to a wide variety of cellular functions. Numerous studies have established that rectification of Kir channels is not an inherent property of the channel protein itself, but rather reflects strong voltage dependence of channel block by intracellular cations, such as polyamines and Mg2+. Here, we identify a previously unknown mechanism of inward rectification in Kir4.1/Kir5.1 channels in the absence of these endogenous blockers. This novel intrinsic rectification originates from the voltage-dependent behavior of Kir4.1/Kir5.1, which is generated by the flux of potassium ions through the channel pore; the inward K+-flux induces the opening of the gate, whereas the outward flux is unable to maintain the gate open. This gating mechanism powered by the K+-flux is convergent with the gating of PIP2 because, at a saturating concentration, PIP2 greatly reduces the inward rectification. Our findings provide evidence of the coexistence of two rectification mechanisms in Kir4.1/Kir5.1 channels: the classical inward rectification induced by blocking cations and an intrinsic voltage-dependent mechanism generated by the K+-flux gating.


Asunto(s)
Canales de Potasio de Rectificación Interna , Iones , Potasio , Bloqueadores de los Canales de Potasio
3.
Eur J Pharmacol ; 899: 174026, 2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-33722592

RESUMEN

Riluzole is an anticonvulsant drug also used to treat the amyotrophic lateral sclerosis and major depressive disorder. This compound has antiglutamatergic activity and is an important multichannel blocker. However, little is known about its actions on the Kv4.2 channels, the molecular correlate of the A-type K+ current (IA) and the fast transient outward current (Itof). Here, we investigated the effects of riluzole on Kv4.2 channels transiently expressed in HEK-293 cells. Riluzole inhibited Kv4.2 channels with an IC50 of 190 ± 14 µM and the effect was voltage- and frequency-independent. The activation rate of the current (at +50 mV) was not affected by the drug, nor the voltage dependence of channel activation, but the inactivation rate was accelerated by 100 and 300 µM riluzole. When Kv4.2 channels were maintained at the closed state, riluzole incubation induced a tonic current inhibition. In addition, riluzole significantly shifted the voltage dependence of inactivation to hyperpolarized potentials without affecting the recovery from inactivation. In the presence of the drug, the closed-state inactivation was significantly accelerated, and the percentage of inactivated channels was increased. Altogether, our findings indicate that riluzole inhibits Kv4.2 channels mainly affecting the closed and closed-inactivated states.


Asunto(s)
Bloqueadores de los Canales de Potasio/farmacología , Riluzol/farmacología , Canales de Potasio Shal/antagonistas & inhibidores , Células HEK293 , Humanos , Activación del Canal Iónico , Potenciales de la Membrana , Canales de Potasio Shal/genética , Canales de Potasio Shal/metabolismo , Factores de Tiempo
4.
Pflugers Arch ; 470(12): 1765-1776, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30155776

RESUMEN

The acetylcholine (ACh)-gated inwardly rectifying K+ current (IKACh) plays a vital role in cardiac excitability by regulating heart rate variability and vulnerability to atrial arrhythmias. These crucial physiological contributions are determined principally by the inwardly rectifying nature of IKACh. Here, we investigated the relative contribution of two distinct mechanisms of IKACh inward rectification measured in atrial myocytes: a rapid component due to KACh channel block by intracellular Mg2+ and polyamines; and a time- and concentration-dependent mechanism. The time- and ACh concentration-dependent inward rectification component was eliminated when IKACh was activated by GTPγS, a compound that bypasses the muscarinic-2 receptor (M2R) and directly stimulates trimeric G proteins to open KACh channels. Moreover, the time-dependent component of IKACh inward rectification was also eliminated at ACh concentrations that saturate the receptor. These observations indicate that the time- and concentration-dependent rectification mechanism is an intrinsic property of the receptor, M2R; consistent with our previous work demonstrating that voltage-dependent conformational changes in the M2R alter the receptor affinity for ACh. Our analysis of the initial and time-dependent components of IKACh indicate that rapid Mg2+-polyamine block accounts for 60-70% of inward rectification, with M2R voltage sensitivity contributing 30-40% at sub-saturating ACh concentrations. Thus, while both inward rectification mechanisms are extrinsic to the KACh channel, to our knowledge, this is the first description of extrinsic inward rectification of ionic current attributable to an intrinsic voltage-sensitive property of a G protein-coupled receptor.


Asunto(s)
Potenciales de Acción , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Miocitos Cardíacos/metabolismo , Receptor Muscarínico M2/metabolismo , Acetilcolina/metabolismo , Animales , Gatos , Células Cultivadas , Femenino , Atrios Cardíacos/citología , Magnesio/metabolismo , Masculino , Miocitos Cardíacos/fisiología , Poliaminas/metabolismo
5.
Biochem Pharmacol ; 152: 264-271, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29621539

RESUMEN

Amitriptyline (AMIT) is a compound widely prescribed for psychiatric and non-psychiatric conditions including depression, migraine, chronic pain, and anorexia. However, AMIT has been associated with risks of cardiac arrhythmia and sudden death since it can induce prolongation of the QT interval on the surface electrocardiogram and torsade de pointes ventricular arrhythmia. These complications have been attributed to the inhibition of the rapid delayed rectifier potassium current (IKr). The slow delayed rectifier potassium current (IKs) is the main repolarizing cardiac current when IKr is compromised and it has an important role in cardiac repolarization at fast heart rates induced by an elevated sympathetic tone. Therefore, we sought to characterize the effects of AMIT on Kv7.1/KCNE1 and homomeric Kv7.1 channels expressed in HEK-293H cells. Homomeric Kv7.1 and Kv7.1/KCNE1 channels were inhibited by AMIT in a concentration-dependent manner with IC50 values of 8.8 ±â€¯2.1 µM and 2.5 ±â€¯0.8 µM, respectively. This effect was voltage-independent for both homomeric Kv7.1 and Kv7.1/KCNE1 channels. Moreover, mutation of residues located on the P-loop and S6 domain along with molecular docking, suggest that T312, I337 and F340 are the most important molecular determinants for AMIT-Kv7.1 channel interaction. Our experimental findings and modeling suggest that AMIT preferentially blocks the open state of Kv7.1/KCNE1 channels by interacting with specific residues that were previously reported to be important for binding of other compounds, such as chromanol 293B and the benzodiazepine L7.


Asunto(s)
Amitriptilina/farmacología , Canal de Potasio KCNQ1/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Potenciales de Acción , Amitriptilina/química , Antidepresivos Tricíclicos/química , Antidepresivos Tricíclicos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Canal de Potasio KCNQ1/metabolismo , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Canales de Potasio con Entrada de Voltaje/metabolismo , Conformación Proteica
6.
Eur J Pharmacol ; 815: 56-63, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28993158

RESUMEN

Inward rectifier potassium (Kir) channels are expressed in almost all mammalian tissues and contribute to a wide range of physiological processes. Kir4.1 channel expression is found in the brain, inner ear, eye, and kidney. Loss-of-function mutations in the pore-forming Kir4.1 subunit cause an autosomal recessive disorder characterized by epilepsy, ataxia, sensorineural deafness and tubulopathy (SeSAME/EST syndrome). Despite its importance in physiological and pathological conditions, pharmacological research of Kir4.1 is limited. Here, we characterized the effect of pentamidine on Kir4.1 channels using electrophysiology, mutagenesis and computational methods. Pentamidine potently inhibited Kir4.1 channels when applied to the cytoplasmic side under inside-out patch clamp configuration (IC50 = 97nM). The block was voltage dependent. Molecular modeling predicted the binding of pentamidine to the transmembrane pore region of Kir4.1 at aminoacids T127, T128 and E158. Mutation of each of these residues reduced the potency of pentamidine to block Kir4.1 channels. A pentamidine analog (PA-6) inhibited Kir4.1 with similar potency (IC50 = 132nM). Overall, this study shows that pentamidine blocks Kir4.1 channels interacting with threonine and glutamate residues in the transmembrane pore region. These results can be useful to design novel compounds with major potency and specificity over Kir4.1 channels.


Asunto(s)
Pentamidina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Sitios de Unión , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Simulación del Acoplamiento Molecular , Pentamidina/metabolismo , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/metabolismo , Conformación Proteica
7.
Brain Res ; 1663: 87-94, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28288868

RESUMEN

Inwardly rectifying potassium (Kir) channels are expressed in many cell types and contribute to a wide range of physiological processes. Particularly, Kir4.1 channels are involved in the astroglial spatial potassium buffering. In this work, we examined the effects of the cationic amphiphilic drug quinacrine on Kir4.1 channels heterologously expressed in HEK293 cells, employing the patch clamp technique. Quinacrine inhibited the currents of Kir4.1 channels in a concentration and voltage dependent manner. In inside-out patches, quinacrine inhibited Kir4.1 channels with an IC50 value of 1.8±0.3µM and with extremely slow blocking and unblocking kinetics. Molecular modeling combined with mutagenesis studies suggested that quinacrine blocks Kir4.1 by plugging the central cavity of the channels, stabilized by the residues E158 and T128. Overall, this study shows that quinacrine blocks Kir4.1 channels, which would be expected to impact the potassium transport in several tissues.


Asunto(s)
Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio de Rectificación Interna/metabolismo , Quinacrina/farmacología , Animales , Astrocitos/metabolismo , Células HEK293 , Humanos , Activación del Canal Iónico/fisiología , Técnicas de Placa-Clamp/métodos , Potasio/metabolismo , Canales de Potasio/metabolismo , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Quinacrina/metabolismo , Ratas
8.
Eur J Pharmacol ; 800: 40-47, 2017 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-28216048

RESUMEN

Kir4.1 channels have been implicated in various physiological processes, mainly in the K+ homeostasis of the central nervous system and in the control of glial function and neuronal excitability. Even though, pharmacological research of these channels is very limited. Chloroquine (CQ) is an amino quinolone derivative known to inhibit Kir2.1 and Kir6.2 channels with different action mechanism and binding site. Here, we employed patch-clamp methods, mutagenesis analysis, and molecular modeling to characterize the molecular pharmacology of Kir4.1 inhibition by CQ. We found that this drug inhibits Kir4.1 channels heterologously expressed in HEK-293 cells. CQ produced a fast-onset voltage-dependent pore-blocking effect on these channels. In inside-out patches, CQ showed notable higher potency (IC50 ≈0.5µM at +50mV) and faster onset of block when compared to whole-cell configuration (IC50 ≈7µM at +60mV). Also, CQ showed a voltage-dependent unblock with repolarization. These results suggest that the drug directly blocks Kir4.1 channels by a pore-plugging mechanism. Moreover, we found that two residues (Thr128 and Glu158), facing the central cavity and located within the transmembrane pore, are particularly important structural determinants of CQ block. This evidence was similar to what was previously reported with Kir6.2, but distinct from the interaction site (cytoplasmic pore) CQ-Kir2.1. Thus, our findings highlight the diversity of interaction sites and mechanisms that underlie amino quinolone inhibition of Kir channels.


Asunto(s)
Cloroquina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/química , Sitios de Unión , Cloroquina/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Cinética , Simulación del Acoplamiento Molecular , Porosidad , Bloqueadores de los Canales de Potasio/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Conformación Proteica
9.
Mol Pharmacol ; 90(3): 334-40, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27247338

RESUMEN

Potassium (K(+)) channels are crucial for determining the shape, duration, and frequency of action-potential firing in excitable cells. Broadly speaking, K(+) channels can be classified based on whether their macroscopic current outwardly or inwardly rectifies, whereby rectification refers to a change in conductance with voltage. Outwardly rectifying K(+) channels conduct greater current at depolarized membrane potentials, whereas inward rectifier channels conduct greater current at hyperpolarized membrane potentials. Under most circumstances, outward currents through inwardly rectifying K(+) channels are reduced at more depolarized potentials. However, the acetylcholine-gated K(+) channel (KACh) conducts current that inwardly rectifies when activated by some ligands (such as acetylcholine), and yet conducts current that outwardly rectifies when activated by other ligands (for example, pilocarpine and choline). The perplexing and paradoxical behavior of KACh channels is due to the intrinsic voltage sensitivity of the receptor that activates KACh channels, the M2 muscarinic receptor (M2R). Emerging evidence reveals that the affinity of M2R for distinct ligands varies in a voltage-dependent and ligand-specific manner. These intrinsic receptor properties determine whether current conducted by KACh channels inwardly or outwardly rectifies. This review summarizes the most recent concepts regarding the intrinsic voltage sensitivity of muscarinic receptors and the consequences of this intriguing behavior on cardiac physiology and pharmacology of KACh channels.


Asunto(s)
Canales de Potasio de Rectificación Interna/metabolismo , Canales de Potasio/metabolismo , Receptores Muscarínicos/metabolismo , Animales , Corazón/fisiología , Humanos , Activación del Canal Iónico , Sistema Nervioso Parasimpático/fisiología
10.
Pflugers Arch ; 468(7): 1207-1214, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27023349

RESUMEN

Recently, it has been shown that G protein-coupled receptors (GPCRs) display intrinsic voltage sensitivity. We reported that the voltage sensitivity of M2 muscarinic receptor (M2R) is also ligand specific. Here, we provide additional evidence to understand the mechanism underlying the ligand-specific voltage sensitivity of the M2R. Using ACh, pilocarpine (Pilo), and bethanechol (Beth), we evaluated the agonist-specific effects of voltage by measuring the ACh-activated K(+) current (I KACh) in feline and rabbit atrial myocytes and in HEK-293 cells expressing M2R-Kir3.1/Kir3.4. The activation of I KACh by the muscarinic agonist Beth was voltage insensitive, suggesting that the voltage-induced conformational changes in M2R do not modify its affinity for this agonist. Moreover, deactivation of the Beth-evoked I KACh was voltage insensitive. By contrast, deactivation of the ACh-induced I KACh was significantly slower at -100 mV than at +50 mV, while an opposite effect was observed when I KACh was activated by Pilo. These findings are consistent with the voltage affinity pattern observed for these three agonists. Our findings suggest that independent of how voltage disturbs the receptor binding site, the voltage dependence of the signaling pathway is ultimately determined by the agonist. These observations emphasize the pharmacological potential to regulate the M2R-parasympathetic associated cardiac function and also other cellular signaling pathways by exploiting the voltage-dependent properties of GPCRs.


Asunto(s)
Acetilcolina/farmacología , Activación del Canal Iónico/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Canales de Potasio/metabolismo , Potasio/metabolismo , Receptor Muscarínico M2/metabolismo , Animales , Sitios de Unión/efectos de los fármacos , Gatos , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Células HEK293 , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Conejos , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos
11.
Pharmacol Rep ; 68(2): 383-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26922543

RESUMEN

BACKGROUND: Inwardly rectifying potassium (Kir) channels are expressed in many cell types and contribute to a wide range of physiological processes. Kir channels dysfunction cause several diseases in brain, ear, heart, muscle, kidney and pancreas, and developmental abnormalities. Therefore, a better understanding of Kir channels pharmacology is desirable. In this study we characterized the electrophysiological and molecular basis of the inhibition produced by the α-adrenergic agonist/antagonist chloroethylclonidine of the currents generated by wild type and mutant Kir2.1 and Kir4.1 channels heterologously expressed in HEK293 cells. METHODS: Macroscopic currents were recorded using the patch clamp technique in the inside out configuration. RESULTS: We found that chloroethylclonidine inhibits the Kir2.1 and Kir4.1 channels in a voltage-dependent manner by interacting with pore facing residues in the cytoplasmic and transmembrane domains, respectively. Site-directed mutagenesis experiments demonstrate that chloroethylclonidine interact with Kir2.1 channels in the cytoplasmic pore involving the E224, E299, D255 and D259 residues, whereas in Kir4.1channels T128 and E158 residues located in the transmembrane pore are important for the chloroethylclonidine effect. CONCLUSIONS: Overall, our results suggest that differences in the cavity of Kir channels are determinants in its interactions with chloroethylclonidine.


Asunto(s)
Clonidina/análogos & derivados , Canales de Potasio de Rectificación Interna/metabolismo , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Clonidina/farmacología , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana , Mutagénesis Sitio-Dirigida/métodos , Potasio/metabolismo
12.
J Pharmacol Toxicol Methods ; 69(3): 237-44, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24412489

RESUMEN

INTRODUCTION: Voltage- and state-dependent blocks are important mechanisms by which drugs affect voltage-gated ionic channels. However, spontaneous (i.e. drug-free) time-dependent changes in the activation and inactivation of hERG and Na(+) channels have been reported when using conventional whole-cell patch-clamp in HEK-293 cells. METHODS: hERG channels were heterologously expressed in HEK-293 cells and in Xenopus laevis oocytes. hERG current (IhERG) was recorded using both conventional and perforated whole-cell patch-clamp (HEK-293 cells), and two microelectrode voltage-clamp (Xenopus oocytes) in drug-free solution, and in the presence of the drug trazodone. RESULTS: In conventional whole-cell setup, we observed a spontaneous time-dependent hyperpolarizing shift in the activation curve of IhERG. Conversely, in perforated patch whole-cell (HEK-293 cells) or in two microelectrode voltage-clamp (Xenopus oocytes) activation curves of IhERG were very stable for periods ~50min. Voltage-dependent inactivation of IhERG was not significantly altered in the three voltage clamp configurations tested. When comparing voltage- and state-dependent effects of the antidepressant drug trazodone on IhERG, similar changes between the three voltage clamp configurations were observed as under drug-free conditions. DISCUSSION: The comparative analysis performed in this work showed that only under conventional whole-cell voltage-clamp conditions, a leftward shift in the activation curve of IhERG occurred, both in the presence and absence of drugs. These spontaneous time-dependent changes in the voltage activation gate of IhERG are a potential confounder in pharmacological studies on hERG channels expressed in HEK-293 cells.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Trazodona/farmacología , Animales , Antidepresivos de Segunda Generación , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/metabolismo , Células HEK293 , Humanos , Oocitos , Factores de Tiempo , Xenopus laevis
13.
Pflugers Arch ; 466(5): 915-24, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24043571

RESUMEN

The human intestinal pathogen Giardia lamblia is a flagellated unicellular protozoan with pronounced medical and biological relevance. However, the basic physiology of Giardia trophozoites has been sparsely studied, especially the electrical and ionic properties of their cellular membrane which are virtually unknown. In this study, we were able to record and characterize the macroscopic ionic currents of Giardia trophozoite membrane by electrophysiological methods of the patch clamp technique. Giardia trophozoites showed a high current density (∼600 pA/pF at -140 mV) that was activated upon hyperpolarization. This current was carried by a chloride-selective channel (I Cl-G) and it was the most important determinant of the membrane potential in Giardia trophozoites. Moreover, this conductance was able to carry other halide anions and the sequence of permeability was Br(-) > Cl(-) ≈ I(-) ≫ F(-). Besides the voltage-dependent inward-rectifying nature of I Cl-G, its activation and deactivation kinetics were comparable to those observed in ClC-2 channels. Extracellular pH modified the voltage-dependent properties of I Cl-G, shifting the activation curve from a V 1/2 = -79 ± 1 mV (pH 7.4) to -93 ± 2 mV (pH 8.4) and -112 ± 2 mV (pH 5.4). Furthermore, the maximal amplitude of I Cl-G measured at -100 mV showed dependence to external pH in a bell-shaped fashion reported only for ClC-2 channels. Therefore, our results suggest that I Cl-G possesses several functional properties similar to the mammalian ClC-2 channels.


Asunto(s)
Potenciales de Acción , Canales de Cloruro/metabolismo , Giardia lamblia/metabolismo , Proteínas Protozoarias/metabolismo , Trofozoítos/metabolismo , Canales de Cloruro CLC-2 , Cloruros/metabolismo , Giardia lamblia/crecimiento & desarrollo , Giardia lamblia/fisiología , Potenciales de la Membrana , Trofozoítos/fisiología
14.
PLoS One ; 8(9): e76085, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086693

RESUMEN

KCNQ genes encode five Kv7 K(+) channel subunits (Kv7.1-Kv7.5). Four of these (Kv7.2-Kv7.5) are expressed in the nervous system. Kv7.2 and Kv7.3 are the principal molecular components of the slow voltage-gated M-channel, which regulates neuronal excitability. In this study, we demonstrate that tamoxifen, an estrogen receptor antagonist used in the treatment of breast cancer, inhibits Kv7.2/Kv7.3 currents heterologously expressed in human embryonic kidney HEK-293 cells. Current inhibition by tamoxifen was voltage independent but concentration-dependent. The IC50 for current inhibition was 1.68 ± 0.44 µM. The voltage-dependent activation of the channel was not modified. Tamoxifen inhibited Kv7.2 homomeric channels with a higher potency (IC50 = 0.74 ± 0.16 µM). The mutation Kv7.2 R463E increases phosphatidylinositol- 4,5-bisphosphate (PIP2) - channel interaction and diminished dramatically the inhibitory effect of tamoxifen compared with that for wild type Kv7.2. Conversely, the mutation Kv7.2 R463Q, which decreases PIP2 -channel interaction, increased tamoxifen potency. Similar results were obtained on the heteromeric Kv7.2 R463Q/Kv7.3 and Kv7.2 R463E/Kv7.3 channels, compared to Kv7.2/Kv7.3 WT. Overexpression of type 2A PI(4)P5-kinase (PIP5K 2A) significantly reduced tamoxifen inhibition of Kv7.2/Kv7.3 and Kv7.2 R463Q channels. Our results suggest that tamoxifen inhibited Kv7.2/Kv7.3 channels by interfering with PIP2-channel interaction because of its documented interaction with PIP2 and the similar effect of tamoxifen on various PIP2 sensitive channels.


Asunto(s)
Canal de Potasio KCNQ2/antagonistas & inhibidores , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/antagonistas & inhibidores , Canal de Potasio KCNQ3/metabolismo , Tamoxifeno/farmacología , Células HEK293 , Humanos , Concentración 50 Inhibidora , Canal de Potasio KCNQ2/genética , Mutación Missense/genética , Técnicas de Placa-Clamp , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo
15.
J Physiol ; 591(17): 4273-86, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23652593

RESUMEN

Choline (Ch) is a precursor and metabolite of the neurotransmitter acetylcholine (ACh). In canine and guinea pig atrial myocytes, Ch was shown to activate an outward K(+) current in a delayed rectifier fashion. This current has been suggested to modulate cardiac electrical activity and to play a role in atrial fibrillation pathophysiology. However, the exact nature and identity of this current has not been convincingly established. We recently described the unique ligand- and voltage-dependent properties of muscarinic activation of ACh-activated K(+) current (IKACh) and showed that, in contrast to ACh, pilocarpine induces a current with delayed rectifier-like properties with membrane depolarization. Here, we tested the hypothesis that Ch activates IKACh in feline atrial myocytes in a voltage-dependent manner similar to pilocarpine. Single-channel recordings, biophysical profiles, specific pharmacological inhibition and computational data indicate that the current activated by Ch is IKACh. Moreover, we show that membrane depolarization increases the potency and efficacy of IKACh activation by Ch and thus gives the appearance of a delayed rectifier activating K(+) current at depolarized potentials. Our findings support the emerging concept that IKACh modulation is both voltage- and ligand-specific and reinforce the importance of these properties in understanding cardiac physiology.


Asunto(s)
Potenciales de Acción , Canales de Potasio de Tipo Rectificador Tardío/metabolismo , Atrios Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Receptor Muscarínico M2/metabolismo , Animales , Gatos , Colina/farmacología , Femenino , Atrios Cardíacos/citología , Masculino , Potenciales de la Membrana , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Pilocarpina/farmacología
16.
Mol Pharmacol ; 82(5): 803-13, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22851715

RESUMEN

Chloroquine and related compounds can inhibit inwardly rectifying potassium channels by multiple potential mechanisms, including pore block and allosteric effects on channel gating. Motivated by reports that chloroquine inhibition of cardiac ATP-sensitive inward rectifier K(+) current (I(KATP)) is antifibrillatory in rabbit ventricle, we investigated the mechanism of chloroquine inhibition of ATP-sensitive potassium (K(ATP)) channels (Kir6.2/SUR2A) expressed in human embryonic kidney 293 cells, using inside-out patch-clamp recordings. We found that chloroquine inhibits the Kir6.2/SUR2A channel by interacting with at least two different sites and by two mechanisms of action. A fast-onset effect is observed at depolarized membrane voltages and enhanced by the N160D mutation in the central cavity, probably reflecting direct channel block resulting from the drug entering the channel pore from the cytoplasmic side. Conversely, a slow-onset, voltage-independent inhibition of I(KATP) is regulated by chloroquine interaction with a different site and probably involves disruption of interactions between Kir6.2/SUR2A and phosphatidylinositol 4,5-bisphosphate. Our findings reveal multiple mechanisms of K(ATP) channel inhibition by chloroquine, highlighting the numerous convergent regulatory mechanisms of these ligand-dependent ion channels.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Antimaláricos/farmacología , Cloroquina/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Receptores de Droga/antagonistas & inhibidores , Animales , Sitios de Unión , Células HEK293 , Humanos , Ratones , Mutación , Técnicas de Placa-Clamp , Fosfatidilinositol 4,5-Difosfato/farmacología , Canales de Potasio de Rectificación Interna/genética , Espermina/farmacología , Receptores de Sulfonilureas , Transfección
17.
Pflugers Arch ; 462(4): 505-17, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21779761

RESUMEN

Cardiac inward rectifier potassium currents determine the resting membrane potential and contribute repolarization capacity during phase 3 repolarization. Quinacrine is a cationic amphiphilic drug. In this work, the effects of quinacrine were studied on cardiac Kir channels expressed in HEK 293 cells and on the inward rectifier potassium currents, I(K1) and I(KATP), in cardiac myocytes. We found that quinacrine differentially inhibited Kir channels, Kir6.2 ∼ Kir2.3 > Kir2.1. In addition, we found in cardiac myocytes that quinacrine inhibited I(KATP) > I(K1). We presented evidence that quinacrine displays a double action towards strong inward rectifier Kir2.x channels, i.e., direct pore block and interference in phosphatidylinositol 4,5-bisphosphate, PIP(2)-Kir channel interaction. Pore block is evident in Kir2.1 and 2.3 channels as rapid block; channel block involves residues E224 and E299 facing the cytoplasmic pore of Kir2.1. The interference of the drug with the interaction of Kir2.x and Kir6.2/SUR2A channels and PIP(2) is suggested from four sources of evidence: (1) Slow onset of current block when quinacrine is applied from either the inside or the outside of the channel. (2) Mutation of Kir2.3(I213L) and mutation of Kir6.2(C166S) increase their affinity for PIP(2) and lowers its sensitivity for quinacrine. (3) Mutations of Kir2.1(L222I and K182Q) which decreased its affinity for PIP(2) increased its sensitivity for quinacrine. (4) Co-application of quinacrine with PIP(2) lowers quinacrine-mediated current inhibition. In conclusion, our data demonstrate how an old drug provides insight into a dual a blocking mechanism of Kir carried inward rectifier channels.


Asunto(s)
Miocitos Cardíacos/fisiología , Fosfatidilinositol 4,5-Difosfato/fisiología , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio de Rectificación Interna/fisiología , Células HEK293 , Humanos , Quinacrina/farmacología
18.
Pflugers Arch ; 462(3): 385-96, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21698391

RESUMEN

We characterized the properties of the voltage-dependent K(+) currents I (to), I (Kr), and I (Ks) in isolated feline sino-atrial node (SAN) myocytes. I (to) activated rapidly and then inactivated with a single exponential and voltage-independent time course. Recovery from inactivation of I (to) followed a single exponential time course with τ = 21.1 ± 2.5 ms, at -80 mV. Steady-state inactivation relationship showed a V½ of inactivation at -47.9 ± 2.3 mV. These biophysical properties are similar to the fast I (to) phenotype of other mammals. I (Kr) exhibited typical negative slope conductance at test potentials > 0 mV and slow deactivation. I (Ks) activated very slowly. The functional contribution of I (to), I (Kr), and I (Ks) to the sustained pacemaking activity of feline SAN myocytes was analyzed. Similar to other mammals, I (to) underlies the initial repolarization phase of the SAN action potential, whereas I (Kr) and I (Ks) mediate repolarization back to the maximal diastolic potential. I (Kr) and I (Ks) also contribute to diastolic depolarization because of their slow deactivation kinetics. The I (Kr) specific blocker E-4031 and the I (Ks) blocker HMR 1556 significantly increased action potential duration, but had negligible effects on the maximum diastolic potential and only modest effects on the frequency of spontaneous activity, suggesting that each one of these two currents itself is capable of supporting action potential repolarization in the feline sinus node.


Asunto(s)
Células Musculares/metabolismo , Miocardio/citología , Canales de Potasio/fisiología , Nodo Sinoatrial/citología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Gatos , Células Cultivadas , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Células Musculares/citología , Células Musculares/efectos de los fármacos , Técnicas de Placa-Clamp , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Nodo Sinoatrial/fisiología
19.
Eur J Pharmacol ; 668(1-2): 72-7, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21663737

RESUMEN

Carvedilol, a ß- and α-adrenoceptor blocker, is used to treat congestive heart failure, mild to moderate hypertension, and myocardial infarction. It has been proposed to block K(ATP) channels by binding to the bundle crossing region at a domain including cysteine at position 166, and thereby plugging the pore region. However, carvedilol was reported not to affect Kir2.1 channels, which lack 166 Cys. Here, we demonstrate that carvedilol inhibits Kir2.3 carried current by an alternative mechanism. Carvedilol inhibited Kir2.3 channels with at least 100 fold higher potency (IC(50)=0.49 µM) compared to that for Kir2.1 (IC(50)>50 µM). Kir2.3 channel inhibition was concentration-dependent and voltage-independent. Increasing Kir2.3 channel affinity for PIP(2), by a I213L point mutation, decreased the inhibitory effect of carvedilol more than twentyfold (IC(50)=11.1 µM). In the presence of exogenous PIP(2), Kir2.3 channel inhibition by carvedilol was strongly reduced (80 vs. 2% current inhibition). These results suggest that carvedilol, as other cationic amphiphilic drugs, inhibits Kir2.3 channels by interfering with the PIP(2)-channel interaction.


Asunto(s)
Carbazoles/farmacología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Rectificación Interna/antagonistas & inhibidores , Canales de Potasio de Rectificación Interna/metabolismo , Propanolaminas/farmacología , Carvedilol , Células HEK293 , Humanos , Mutación Puntual , Canales de Potasio de Rectificación Interna/genética , Unión Proteica/efectos de los fármacos
20.
Pflugers Arch ; 462(2): 235-43, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21487692

RESUMEN

Pilocarpine is a nonspecific agonist of muscarinic receptors which was recently found to activate the M(2) receptor subtype in a voltage-dependent manner. The purpose of our study was to investigate the role of the acetylcholine (muscarinic)-activated K(+) current (I (KACh)) on the negative chronotropic effect of pilocarpine in rabbit sinoatrial node. In multicellular preparations, we studied the effect of pilocarpine on spontaneous action potentials. In isolated myocytes, using the patch clamp technique, we studied the effects of pilocarpine on I (KACh). Pilocarpine produced a decrease in spontaneous frequency, hyperpolarization of the maximum diastolic potential, and a decrease in the diastolic depolarization rate. These effects were partially antagonized by tertiapin Q. Cesium and calyculin A in the presence of tertiapin Q partially prevented the effects of pilocarpine. In isolated myocytes, pilocarpine activated the muscarinic potassium current, I (KACh) in a voltage-dependent manner. In conclusion, the negative chronotropic effects of pilocarpine on the sinatrial node could be mainly explained by activation of I (KACh).


Asunto(s)
Acetilcolina/metabolismo , Potenciales de Acción/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Pilocarpina/farmacología , Canales de Potasio/metabolismo , Potasio/metabolismo , Nodo Sinoatrial/efectos de los fármacos , Animales , Venenos de Abeja/farmacología , Cesio/metabolismo , Inhibidores Enzimáticos/farmacología , Activación del Canal Iónico/efectos de los fármacos , Toxinas Marinas , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Oxazoles/farmacología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Conejos , Nodo Sinoatrial/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...