Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mar Drugs ; 19(11)2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34822501

RESUMEN

Potassium channel Kv1.5 has been considered a key target for new treatments of atrial tachyarrhythmias, with few side effects. Four new debromoaplysiatoxin analogues with a 6/6/12 fused ring system were isolated from marine cyanobacterium Lyngbya sp. Their planar structures were elucidated by HRESIMS, 1D and 2D NMR. The absolute configuration of oscillatoxin J (1) was determined by single-crystal X-ray diffraction, and the absolute configurations of oscillatoxin K (2), oscillatoxin L (3) and oscillatoxin M (4) were confirmed on the basis of GIAO NMR shift calculation followed by DP4 analysis. The current study confirmed the absolute configuration of the pivotal chiral positions (7S, 9S, 10S, 11R, 12S, 15S, 29R and 30R) at traditional ATXs with 6/12/6 tricyclic ring system. Compound 1, 2 and 4 exhibited blocking activities against Kv1.5 with IC50 values of 2.61 ± 0.91 µM, 3.86 ± 1.03 µM and 3.79 ± 1.01 µM, respectively. However, compound 3 exhibited a minimum effect on Kv1.5 at 10 µM. Furthermore, all of these new debromoaplysiatoxin analogs displayed no apparent activity in a brine shrimp toxicity assay.


Asunto(s)
Canal de Potasio Kv1.5/efectos de los fármacos , Toxinas de Lyngbya/farmacología , Lyngbya , Animales , Organismos Acuáticos , Artemia , Humanos , Concentración 50 Inhibidora , Canal de Potasio Kv1.5/antagonistas & inhibidores , Toxinas de Lyngbya/química , Ratones , Relación Estructura-Actividad
2.
Toxicol Appl Pharmacol ; 411: 115365, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33316272

RESUMEN

Polychlorinated biphenyls (PCBs) are persistent and serious organic pollutants and can theoretically form 209 congeners. PCBs can be divided into two categories: dioxin-like (DL) and non-DL (NDL). NDL-PCBs, which lack aryl hydrocarbon receptor affinity, have been shown to perturb the functions of Jurkat T cells, cerebellar granule cells, and uterine cells. Kv1.3 and Kv1.5 channels are important in immune and heart functions, respectively. We investigated the acute effects of 2,2',6-trichlorinated biphenyl (PCB19), an NDL-PCB, on the currents of human Kv1.3 and Kv1.5 channels. PCB19 acutely blocked the Kv1.3 peak currents concentration-dependently with an IC50 of ~2 µM, without changing the steady-state current. The PCB19-induced inhibition of the Kv1.3 peak current occurred rapidly and voltage-independently, and the effect was irreversible, excluding the possibility of genomic regulation. PCB19 increased the time constants of both activation and inactivation of Kv1.3 channels, resulting in the slowing down of both ultra-rapid activation and intrinsic inactivation. However, PCB19 failed to alter the steady-state curves of activation and inactivation. Regarding the Kv1.5 channel, PCB19 affected neither the peak current nor the steady-state current at the same concentrations tested in the Kv1.3 experiments, showing selective inhibition of PCB19 on the Kv1.3 than the Kv1.5. The presented data indicate that PCB19 could acutely affect the human Kv1.3 channel through a non-genomic mechanism, possibly causing toxic effects on various human physiological functions related to the Kv1.3 channel, such as immune and neural systems.


Asunto(s)
Contaminantes Ambientales/toxicidad , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.5/efectos de los fármacos , Bifenilos Policlorados/toxicidad , Bloqueadores de los Canales de Potasio/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Potenciales de la Membrana , Oocitos , Factores de Tiempo , Xenopus laevis
3.
Circ J ; 83(4): 718-726, 2019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30787218

RESUMEN

BACKGROUND: Intracellular uric acid is known to increase the protein level and channel current of atrial Kv1.5; however, mechanisms of the uric acid-induced enhancement of Kv1.5 expression remain unclear. Methods and Results: The effects of uric acid on mRNA and protein levels of Kv1.5, as well as those of Akt, HSF1 and Hsp70, in HL-1 cardiomyocytes were studied by using qRT-PCR and Western blotting. The uptake of uric acid was measured using radio-labeled uric acid. The Kv1.5-mediated channel current was also measured by using patch clamp techniques. Uric acid up-taken by HL-1 cells significantly increased the level of Kv1.5 proteins in a concentration-dependent manner, with this increase abolished by an uric acid transporter inhibitor. Uric acid slowed degradation of Kv1.5 proteins without altering its mRNA level. Uric acid enhanced phosphorylation of Akt and HSF1, and thereby increased both transcription and translation of Hsp70; these effects were abolished by a PI3K inhibitor. Hsp70 knockdown abolished the uric acid-induced increases of Kv1.5 proteins and channel currents. CONCLUSIONS: Intracellular uric acid could stabilize Kv1.5 proteins through phosphorylation of Akt and HSF1 leading to enhanced expression of Hsp70.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico/metabolismo , Canal de Potasio Kv1.5/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ácido Úrico/farmacología , Animales , Línea Celular , Canal de Potasio Kv1.5/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas , Transcripción Genética
4.
Pflugers Arch ; 468(11-12): 1885-1894, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27796577

RESUMEN

Human Kv1.5 channels (hKv1.5) conduct the ultra-rapid delayed rectifier potassium current (I Kur), which plays an important role in action potential repolarization of atrial myocytes. The present study was undertaken to examine the effects of acidic pH on hKv1.5 wild-type (WT) and its pore mutant channels heterologously expressed in Chinese hamster ovary (CHO) cells using site-directed mutagenesis combined with whole-cell patch-clamp technique. Both extracellular and intracellular acidifications equally and reversely reduced the amplitude of hKv1.5 currents. The extracellular acidification significantly shifted the voltage dependence of current activation to more depolarized potentials and accelerated deactivation kinetics of the current. The ancillary ß subunits Kvß1.3 and Kvß1.2, known to modify the pharmacological sensitivities of hKv1.5, enhanced the extracellular proton-induced inhibitory effect on hKv1.5 current. In addition, several mutants (T462C, T479A, T480A, and I508A) exhibited significantly higher sensitivity to acidic pH-induced inhibition compared with WT channel, whereas the inhibitory effect of acidic pH was markedly reduced in H463G mutant. These observations indicate that (1) extracellular acidification modifies hKv1.5 gating and activity, (2) ß subunits and several residues (T462, T479, T480, and I508) play critical roles in determining the sensitivity of the channel to acidic exposure, and (3) H463 may be a critical sensor for the channel inhibition by extracellular protons.


Asunto(s)
Canal de Potasio Kv1.5/metabolismo , Protones , Potenciales de Acción , Sustitución de Aminoácidos , Animales , Células CHO , Cricetinae , Cricetulus , Espacio Extracelular/metabolismo , Humanos , Activación del Canal Iónico , Canal de Potasio Kv1.5/efectos de los fármacos , Canal de Potasio Kv1.5/genética
5.
Eur J Pharmacol ; 764: 622-632, 2015 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-26256861

RESUMEN

The intravenous anesthetic propofol affects the function of a diversity of ligand-gated and voltage-gated ion channels. However, there is little information as to whether propofol directly interacts with voltage-gated ion channel proteins to modulate their functions. The Kv1.5 channel is activated by membrane depolarization during action potentials and contributes to atrial repolarization in the human heart. This study was undertaken to examine the effect of propofol on voltage-gated human Kv1.5 (hKv1.5) channel and to elucidate the underlying molecular determinants. Site-directed mutagenesis was carried out through six amino acids that reside within the pore domain of hKv1.5 channel. Whole-cell patch-clamp technique was used to record membrane currents through the wild type and mutant hKv1.5 channels heterologously expressed in Chinese hamster ovary cells. Propofol (≥5 µM) reversibly and concentration-dependently (IC50 of 49.3±9.4 µM; n=6) blocked hKv1.5 current. Propofol-induced block of hKv1.5 current gradually progressed during depolarizing voltage-clamp steps and was enhanced by higher frequency of activation, consistent with a preferential block of the channels in their open state. The degree of current block by propofol was significantly attenuated in T480A, I502A, I508A and V516A, but not in H463C and L510A mutants of hKv1.5 channel. Thus, several amino acids near the selectivity filter (Thr480) or within S6 (Ile502, Ile508 and Val516) are found to be critically involved in the blocking action of propofol. This study provides the first evidence suggesting that direct interaction with specific amino acids underlies the blocking action of propofol on voltage-gated hKv1.5 channel.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Canal de Potasio Kv1.5/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Propofol/farmacología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Humanos , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Potenciales de la Membrana , Mutagénesis Sitio-Dirigida , Mutación , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/metabolismo , Propofol/metabolismo , Unión Proteica , Factores de Tiempo , Transfección
6.
Circ Arrhythm Electrophysiol ; 5(6): 1193-201, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23060423

RESUMEN

BACKGROUND: We evaluated the viability of I(Kur) as a target for maintenance of sinus rhythm in patients with a history of atrial fibrillation through the testing of MK-0448, a novel I(Kur) inhibitor. METHODS AND RESULTS: In vitro MK-0448 studies demonstrated strong inhibition of I(Kur) with minimal off-target activity. In vivo MK-0448 studies in normal anesthetized dogs demonstrated significant prolongation of the atrial refractory period compared with vehicle controls without affecting the ventricular refractory period. In studies of a conscious dog heart failure model, sustained atrial fibrillation was terminated with bolus intravenous MK-0448 doses of 0.03 and 0.1 mg/kg. These data led to a 2-part first-in-human study: Part I evaluated safety and pharmacokinetics, and part II was an invasive electrophysiological study in healthy subjects. MK-0448 was well-tolerated with mild adverse experiences, most commonly irritation at the injection site. During the electrophysiological study, ascending doses of MK-0448 were administered, but no increases in atrial or ventricular refractoriness were detected, despite achieving plasma concentrations in excess of 2 µmol/L. Follow-up studies in normal anesthetized dogs designed to assess the influence of autonomic tone demonstrated that prolongation of atrial refractoriness with MK-0448 was markedly attenuated in the presence of vagal nerve simulation, suggesting that the effects of I(Kur) blockade on atrial repolarization may be negated by enhanced parasympathetic neural tone. CONCLUSIONS: The contribution of I(Kur) to human atrial electrophysiology is less prominent than in preclinical models and therefore is likely to be of limited therapeutic value for the prevention of atrial fibrillation.


Asunto(s)
Fibrilación Atrial/prevención & control , Fenómenos Electrofisiológicos/fisiología , Canal de Potasio Kv1.5/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/efectos adversos , Bloqueadores de los Canales de Potasio/farmacocinética , Adulto , Animales , Fibrilación Atrial/fisiopatología , Canales de Potasio de Tipo Rectificador Tardío/antagonistas & inhibidores , Canales de Potasio de Tipo Rectificador Tardío/efectos de los fármacos , Canales de Potasio de Tipo Rectificador Tardío/fisiología , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Fenómenos Electrofisiológicos/efectos de los fármacos , Femenino , Sistema de Conducción Cardíaco/fisiología , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/prevención & control , Humanos , Técnicas In Vitro , Canal de Potasio Kv1.5/efectos de los fármacos , Canal de Potasio Kv1.5/fisiología , Masculino , Bloqueadores de los Canales de Potasio/farmacología , Piridinas/efectos adversos , Piridinas/farmacocinética , Piridinas/farmacología , Nodo Sinoatrial/fisiología , Sulfonamidas/efectos adversos , Sulfonamidas/farmacocinética , Sulfonamidas/farmacología , Nervio Vago/fisiología
7.
Clin Exp Nephrol ; 16(1): 73-80, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22038256

RESUMEN

Serum- and glucocorticoid inducible-kinase 1 (SGK1) is an early gene transcriptionally upregulated by cell stress such as cell shrinkage and hypoxia and several hormones including gluco- and mineralocorticoids. It is activated by insulin and growth factors. SGK1 is a powerful regulator of a wide variety of channels and transporters. The present review describes the role of SGK1 in the regulation of potassium (K(+)) channels, K(+) transporters and K(+) homeostasis. SGK1-regulated K(+) channels include renal outer medullary K+ channel, Kv1.3, Kv1.5, KCNE1/KCNQ1, KCNQ4 and, via regulation of calcium (Ca(2+)) entry, Ca(2+)-sensitive K(+) channels. SGK1-sensitive transporters include sodium-potassium-chloride cotransporter 2 and sodium/potassium-adenosine triphosphatase. SGK1-dependent regulation of K(+) channels and K(+) transport contributes to the stimulation of renal K(+) excretion following high K(+) intake, to insulin-induced cellular K(+) uptake and hypokalemia, to inhibition of insulin release by glucocorticoids, to stimulation of mast cell degranulation and gastric acid secretion, and to cardiac repolarization. Thus, SGK1 has a profound effect on K(+) homeostasis and on a multitude of K(+)-sensitive cellular functions.


Asunto(s)
Proteínas Inmediatas-Precoces/fisiología , Riñón/metabolismo , Potasio/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Tránsito Gastrointestinal , Corazón/efectos de los fármacos , Homeostasis , Humanos , Hipopotasemia/inducido químicamente , Insulina/metabolismo , Secreción de Insulina , Transporte Iónico , Canal de Potasio Kv1.5/efectos de los fármacos , Neuronas/efectos de los fármacos , Canales de Potasio/efectos de los fármacos , Canales de Potasio/metabolismo , Canales de Potasio de Rectificación Interna/efectos de los fármacos
8.
Sheng Li Xue Bao ; 63(3): 219-24, 2011 Jun 25.
Artículo en Chino | MEDLINE | ID: mdl-21681339

RESUMEN

The purpose of the present study was to investigate the effects of different concentrations of ethanol on action potential (AP) in the isolated rat myocardium and the possible mechanism of electric-physiological changes. Standard microelectrode technique was used to record AP in isolated rat myocardium, and whole cell patch clamp technique was used to record the human Kv1.5 (hKv1.5) channel currents in HEK293 cells. The effects of different concentrations of ethanol (6.25, 12.5, 25.0, 50.0, 100.0 and 200.0 mmol/L) on AP parameters in rat atrium and papillary and Kv1.5 channel currents in HEK293 cells were analyzed. The results showed that in isolated atrium, action potential amplitude (APA), action potential duration (APD), action potential duration of 50% repolarization (APD(50)) and action potential duration of 90% repolarization (APD(90)) were not affected by 6.25 and 12.5 mmol/L ethanol, while APD, APD(50) and APD(90) were prolonged significantly by 25.0-200.0 mmol/L ethanol (P < 0.05 or P < 0.01), and APA was reduced with 100.0 and 200.0 mmol/L ethanol (P < 0.05 or P < 0.01). In isolated papillary, APA, APD, APD(50) and APD(90) were not affected by 6.25-25.0 mmol/L ethanol, while APD, APD(50) and APD(90) were prolonged significantly with 50.0-200.0 mmol/L ethanol (P < 0.05 or P < 0.01), and APA was reduced with 200.0 mmol/L ethanol (P < 0.05). The Kv1.5 channel currents were inhibited by ethanol in a concentration dependent manner in HEK293 cells. These findings suggest that 6.25 and 12.5 mmol/L ethanol produce no effects on AP parameters, and 50.0-200.0 mmol/L ethanol prolong APD significantly in isolated rat atrium and papillary. The prolonged effect on APD in isolated myocardium may be due to the inhibition of the Kv1.5 channel currents.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Etanol/farmacología , Canal de Potasio Kv1.5/efectos de los fármacos , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , Animales , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Canal de Potasio Kv1.5/antagonistas & inhibidores , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
9.
Pflugers Arch ; 460(5): 851-62, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20734202

RESUMEN

The goal of the present study was to examine the effects of lobeline, an agonist at nicotinic receptors, on cloned Kv channels, Kv1.5, Kv3.1, Kv4.3, and human ether-a-gogo-related gene (HERG), which are stably expressed in Chinese hamster ovary (CHO) or human embryonic kidney 293 (HEK293) cells. Whole-cell patch-clamp experiments revealed that lobeline accelerated the decay rate of Kv1.5 inactivation, decreasing the current amplitude at the end of the pulse in a concentration-dependent manner with a half-maximal inhibitory concentration (IC(50)) value of 15.1 µM. Using a time constant for the time course of drug-channel interaction, the apparent association (k(+1)), and dissociation rate (k(-1)) constants were 2.4 µΜ(-1) s(-1) and 40.9 s(-1), respectively. The calculated K(D) was 17.0 µΜ. Lobeline slowed the decay rate of the tail current, resulting in a tail crossover phenomenon. The inhibition of Kv1.5 by lobeline steeply increased at potentials between -10 and +10 mV, which corresponds to the voltage range of channel activation. At more depolarized potentials a weaker voltage dependence was observed (δ=0.26). The voltage dependence of the steady-state activation curve was not affected by lobeline, but lobeline shifted the steady-state inactivation curve of Kv1.5 in the hyperpolarizing direction. Lobeline produced use-dependent inhibition of Kv1.5 at frequencies of 1 and 2 Hz, and slowed the recovery from inactivation. Lobeline also inhibited Kv3.1, Kv4.3, and HERG in a concentration-dependent manner, with IC(50) values of 21.7, 28.2, and 0.34 µM, respectively. These results indicate that lobeline produces a concentration-, time-, voltage-, and use-dependent inhibition of Kv1.5, which can be interpreted as an open-channel block mechanism.


Asunto(s)
Canal de Potasio Kv1.5/efectos de los fármacos , Lobelina/farmacología , Animales , Células CHO , Clonación Molecular , Cricetinae , Cricetulus , Células HEK293 , Humanos , Canal de Potasio Kv1.5/antagonistas & inhibidores , Técnicas de Placa-Clamp , Canales de Potasio Shal/antagonistas & inhibidores , Canales de Potasio Shaw/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Regulador Transcripcional ERG
10.
Zhonghua Xin Xue Guan Bing Za Zhi ; 37(2): 165-8, 2009 Feb.
Artículo en Chino | MEDLINE | ID: mdl-19719998

RESUMEN

OBJECTIVE: To observe the effects of telmisartan on Kv1.3 and Kv1.5 potassium channels expressed in Xenopus oocytes. METHODS: Kv1.3 and Kv1.5 potassium channel currents expressed in Xenopus oocytes were recorded and observed in the absence and presence of telmisartan using standard two-microelectrode voltage clamp techniques. RESULTS: Telmisartan resulted in a concentration- and voltage-dependent inhibition effect on Kv1.3 channel current (IC(50) 2.05 micromol/L)and on Kv1.5 channel current (IC(50) 2.37 micromol/L). CONCLUSIONS: Telmisartan blocks open-state Kv1.3 channel which could be one of the mechanisms related to its immunomodulatory and anti-atherosclerosis effect. Telmisartan also blocks open-state Kv1.5 channel which might partly account for its effect on reducing the incidence of atrial fibrillation.


Asunto(s)
Bencimidazoles/farmacología , Benzoatos/farmacología , Canal de Potasio Kv1.3/efectos de los fármacos , Canal de Potasio Kv1.5/efectos de los fármacos , Oocitos/efectos de los fármacos , Animales , Técnicas In Vitro , Oocitos/metabolismo , Técnicas de Placa-Clamp , Telmisartán , Xenopus
11.
Curr Top Med Chem ; 9(4): 339-47, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19442205

RESUMEN

Attributed to the extensive researches towards the functions of Kv1.5 gene (also known as KCN5A gene), the therapeutic significances of Kv1.5 channel have been paid special attentions to cardiovascular diseases as well as tumor, the top two public-health problems for the aging population. During recent years, the hindrance for developing cardiovascular drugs is how to discriminate their therapeutic effect from cardiotoxicities. Therefore, targeting Kv1.5 gene, which encodes the atrial selective I(Kur) potassium channel, has been confirmed as an appealing strategy for the treatment of atrial diseases. Moreover, the functions of Kv1.5 gene in carcinoma progression have also provoked a flurry of research in the hope of developing adjunctive or combined treatment of Kv1.5 modulators to enhance curative effects of classic chemotherapeutic agents.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Canal de Potasio Kv1.5/efectos de los fármacos , Humanos , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/farmacología , Relación Estructura-Actividad
12.
Biochem Pharmacol ; 77(2): 177-85, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18955031

RESUMEN

The voltage-gated potassium channel Kv1.5 is one of the key regulators of membrane potential repolarization in human atrial myocytes and is considered a potential drug target to treat atrial fibrillation. In this study we sought to determine molecular mechanism of action of DPO-1, a diphenylphosphine oxide derivative recently shown to terminate experimental atrial arrhythmia without affecting ventricular refractory period. In addition, we provided similar analysis for additional two small molecule blockers, representing different structural classes: cyclohexanones (PAC) and nor-triterpenoids (correolide). To rapidly identify the residues within the Kv1.5 channel critical for blocking activity of these molecules, two functional high-throughput ion channel assays were employed together with site-directed mutagenesis. Our study revealed that the residues critical for blocking activity of for DPO-1 include T480, localized at the outer mouth of the pore, and two residues along S6 helix: V505 and I508. The overlapping site was identified for PAC and included residues T480 and V505. In contrast to DPO-1, the I508A mutation resulted in only a modest reduction in the block of Kv1.5 by PAC (9-fold). Correolide, the largest molecule examined, made widespread interactions along the entire length of the pore (from T480 to V516). In summary, we have identified multiple residues involved in forming high affinity binding site for Kv1.5 blockers. Similar approaches of high-throughput ion channel technologies, combined with site-directed mutagenesis, may allow for parallel, rapid and accurate analysis of ion channel interactions with multiple compounds and could facilitate the design of more potent and selective ion channel blockers.


Asunto(s)
Ciclohexanonas/farmacología , Canal de Potasio Kv1.5/fisiología , Fosfinas/farmacología , Animales , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/fisiopatología , Células CHO , Cricetinae , Cricetulus , Venenos Elapídicos/farmacología , Electrofisiología/métodos , Humanos , Cinética , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/efectos de los fármacos , Canal de Potasio Kv1.5/genética , Mutagénesis Sitio-Dirigida , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Fosfinas/uso terapéutico , Reacción en Cadena de la Polimerasa , Rubidio/metabolismo , Espectrofotometría Atómica , Triterpenos/farmacología
13.
J Pharmacol Sci ; 108(1): 49-55, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18818480

RESUMEN

Kv1.5 is considered to be a potential molecular target for treatment of atrial fibrillation or flutter. Disopyramide is widely used in the treatment of atrial flutter and/or atrial fibrillation. The present study was undertaken to characterize the effects of disopyramide on currents mediated by Kv1.5 channels and to determine the putative binding site involved in the inhibitory effects of disopyramide. Experiments were carried out on wild-type and site directed mutated hKv1.5 channels expressed on HEK 293 cells using the patch-clamp technique. Disopyramide acting from the cytoplasmic side of the membrane produced blocking effects on Kv1.5 that exhibited several features typical of an open channel blocker. Ala-scanning mutagenesis of the Kv1.5 pore domain combined with macroscopic current analysis suggested that disopyramide interacted only with the Val512 residue that faces to the central cavity of the channel. Mutation of this key residue to Ala caused marked change in the IC(50) of disopyramide (22-fold). The single interaction between disopyramide and Val512 in the PVP region is able to change the mechanism of channel closure, reproducing the "foot-in-the-door" phenomenon.


Asunto(s)
Antiarrítmicos/farmacología , Disopiramida/farmacología , Canal de Potasio Kv1.5/efectos de los fármacos , Bloqueadores de los Canales de Potasio , Alanina/genética , Línea Celular , Interpretación Estadística de Datos , Humanos , Canal de Potasio Kv1.5/química , Canal de Potasio Kv1.5/genética , Mutagénesis/efectos de los fármacos , Técnicas de Placa-Clamp
14.
Eur J Pharmacol ; 590(1-3): 317-21, 2008 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-18573250

RESUMEN

Tanshinone IIA, one of the main active components from Chinese herb Danshen, is widely used to treat cardiovascular diseases including arrhythmia in Asian countries especially in China. However, the mechanisms underlying its anti-arrythmia effects are not clear. In this study we investigate the effects of tanshinone IIA on human KCNQ1/KCNE1 potassium channels (I(Ks)), human ether-a-go-go-related gene potassium channels (hERG), Kv1.5 potassium channels, inward rectifier potassium channels (I(K1)) expressed in HEK 293 cells using patch clamp technique. Tanshinone IIA potently and reversibly enhanced the amplitude of I(Ks) in a concentration dependent manner with an EC(50) of 64.5 microM, accelerated the activation rate of I(Ks) channels, decelerated their deactivation and shifted the voltage dependence of I(Ks) activation to negative direction. Isoproteronol, a stimulator of beta-adrenergic receptor, at 1 microM and sodium nitroprusside (SNP), a NO donor, at 1 mM, had no significant effects on the enhancement of I(Ks) by 30 microM tanshinone IIA. N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide (H89), a selective protein kinase A inhibitor, at 0.1 microM and 1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one (ODQ), a selective nitric oxide-sensitive guanylyl cyclase inhibitor, at 10 microM, also had no significant effects on the enhancement of I(Ks) by 30 microM tanshinone IIA. Tanshinone IIA did not affect expressed hERG channels, Kv1.5 channels and I(K1) channels. These results indicate that tanshinone IIA directly and specifically activate human cardiac KCNQ1/KCNE1 potassium channels (I(Ks)) in HEK 293 cell through affecting the channels' kinetics.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Corazón/efectos de los fármacos , Canal de Potasio KCNQ1/efectos de los fármacos , Fenantrenos/farmacología , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Abietanos , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Guanilato Ciclasa/antagonistas & inhibidores , Humanos , Canal de Potasio Kv1.5/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Oxadiazoles/farmacología , Quinoxalinas/farmacología
15.
Anesthesiology ; 107(4): 641-51, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17893461

RESUMEN

BACKGROUND: Kvbeta1.3 subunit modifies the gating and the pharmacology of Kv1.5 channels, decreasing their sensitivity to block induced by drugs, suggesting that Kvbeta1.3 competes with them for a binding site at Kv1.5 channels. METHODS: Currents generated by the activation of Kv1.5 and Kv1.5 + Kvbeta1.3 channels expressed in HEK293 cells and Xenopus oocytes were recorded by using whole cell patch clamp and voltage clamp techniques. RESULTS: Block of Kv1.5, but not that produced on Kv1.5 + Kvbeta1.3 channels, was voltage dependent. In both channels, bupivacaine block was time dependent. R(+)- and S(-)-bupivacaine blocked Kv1.5 with IC50 4.4 +/- 0.5 microM (n = 15) and 39.8 +/- 8.2 microM (n = 16; P < 0.05), respectively. These values increased fourfold for R(+)-bupivacaine (17.2 +/- 2.2 microM) and twofold for S(-)-bupivacaine (71.9 +/- 11.5 microM) in Kv1.5 + Kvbeta1.3 channels. Therefore, the degree of stereoselectivity (theta) decreased from 9 to 4 in the presence of Kvbeta1.3. The decrease in potency to block Kv1.5 + Kvbeta1.3 channels was the result of a less stable interaction between bupivacaine enantiomers and channels. Differences in stereoselectivity in each situation were due to a more favorable interaction between the channel and R(+)-bupivacaine. In the presence of Kvbeta1.3, stereoselectivity was abolished for V514A mutant channels (involved in bupivacaine binding but not in Kvbeta1.3 binding) but not for L510A (part of Kvbeta1.3 binding site). CONCLUSIONS: The degree of stereoselective block of Kv1.5 decreases from 9 to 4 when Kvbeta1.3 is present. L510 is determinant for the modulation of bupivacaine block, because it is the only residue of the S6 segment that binds to both bupivacaine and Kvbeta1.3. These findings support an overlapping binding site for drugs and Kvbeta1.3.


Asunto(s)
Anestésicos Locales/química , Anestésicos Locales/farmacología , Bupivacaína/química , Bupivacaína/farmacología , Canal de Potasio Kv1.3/fisiología , Canal de Potasio Kv1.5/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Algoritmos , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Línea Celular , Electrofisiología , Humanos , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.5/genética , Mutación/genética , Mutación/fisiología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Estereoisomerismo , Transfección , Xenopus laevis
16.
Am J Physiol Heart Circ Physiol ; 293(4): H2231-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17675568

RESUMEN

Small coronary arteries (SCA) from diabetic rats exhibit enhanced peroxynitrite (ONOO(-)) formation and concurrent impairment of voltage-dependent potassium (K(v)) channel function. However, it is unclear whether ONOO(-) plays a causative role in this impairment. We hypothesized that functional loss of K(v) channels in coronary smooth muscle cells (SMC) in diabetes is due to ONOO(-) with subsequent tyrosine nitration of K(v) channel proteins. Diabetic rats and nondiabetic controls were treated with or without ebselen (Eb) for 4 wk. SCA were prepared for immunohistochemistry (IHC), immunoprecipitation (IP) followed by Western blot (WB), videomicroscopy, and patch-clamp analysis. IHC revealed excess ONOO(-) in SCA from diabetic rats. IP and WB revealed elevated nitration of the K(v)1.2 alpha-subunit and reduced K(v)1.2 protein expression in diabetic rats. Each of these changes was improved in Eb-treated rats. Protein nitration and K(v)1.5 expression were unchanged in SCA from diabetic rats. Forskolin, a direct cAMP activator that induces K(v)1 channel activity, dilated SCA from nondiabetic rats in a correolide (Cor; a selective K(v)1 channel blocker)-sensitive fashion. Cor did not alter the reduced dilation to forskolin in diabetic rats; however, Eb partially restored the Cor-sensitive component of dilation. Basal K(v) current density and response to forskolin were improved in smooth muscle cells from Eb-treated DM rats. We conclude that enhanced nitrosative stress in diabetes mellitus contributes to K(v)1 channel dysfunction in the coronary microcirculation. Eb may be beneficial for the therapeutic treatment of vascular complications in diabetes mellitus.


Asunto(s)
Antioxidantes/farmacología , Azoles/farmacología , Vasos Coronarios/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Canal de Potasio Kv.1.2/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Ácido Peroxinitroso/metabolismo , Tirosina/análogos & derivados , Vasodilatación/efectos de los fármacos , Adenilil Ciclasas/metabolismo , Animales , Antioxidantes/uso terapéutico , Azoles/uso terapéutico , Colforsina/farmacología , Vasos Coronarios/enzimología , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiopatología , AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Relación Dosis-Respuesta a Droga , Activadores de Enzimas/farmacología , Isoindoles , Canal de Potasio Kv.1.2/metabolismo , Canal de Potasio Kv1.5/efectos de los fármacos , Canal de Potasio Kv1.5/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Compuestos de Organoselenio/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Subunidades de Proteína , Ratas , Ratas Sprague-Dawley , Proyectos de Investigación , Triterpenos/farmacología , Tirosina/metabolismo
18.
Am J Physiol Heart Circ Physiol ; 292(6): H2898-905, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17293496

RESUMEN

Atrial fibrillation (AF) is the most frequent arrhythmia found in clinical practice. In recent studies, a decrease in the development or recurrence of AF was found in hypertensive patients treated with angiotensin-converting enzyme inhibitors or angiotensin receptor-blocking agents. Hypertension is related to an increased wall tension in the atria, resulting in increased stretch of the individual myocyte, which is one of the major stimuli for the remodeling process. In the present study, we used a model of cultured atrial neonatal rat cardiomyocytes under conditions of stretch to provide insight into the mechanisms of the preventive effect of the angiotensin receptor-blocking agent losartan against AF on a molecular level. Stretch significantly increased protein-to-DNA ratio and atrial natriuretic factor mRNA expression, indicating hypertrophy. Expression of genes encoding for the inward rectifier K(+) current (I(K1)), Kir 2.1, and Kir 2.3, as well as the gene encoding for the ultrarapid delayed rectifier K(+) current (I(Kur)), Kv 1.5, was significantly increased. In contrast, mRNA expression of Kv 4.2 was significantly reduced in stretched myocytes. Alterations of gene expression correlated with the corresponding current densities: I(K1) and I(Kur) densities were significantly increased in stretched myocytes, whereas transient outward K(+) current (I(to)) density was reduced. These alterations resulted in a significant abbreviation of the action potential duration. Losartan (1 microM) prevented stretch-induced increases in the protein-to-DNA ratio and atrial natriuretic peptide mRNA expression in stretched myocytes. Concomitantly, losartan attenuated stretch-induced alterations in I(K1), I(Kur), and I(to) density and gene expression. This prevented the stretch-induced abbreviation of action potential duration. Prevention of stretch-induced electrical remodeling might contribute to the clinical effects of losartan against AF.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Antihipertensivos/farmacología , Expresión Génica/efectos de los fármacos , Losartán/farmacología , Mecanotransducción Celular/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Potasio/metabolismo , Potenciales de Acción/efectos de los fármacos , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Animales Recién Nacidos , Antihipertensivos/uso terapéutico , Fibrilación Atrial/etiología , Fibrilación Atrial/prevención & control , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Aumento de la Célula/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Atrios Cardíacos/citología , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Hipertensión/complicaciones , Hipertensión/tratamiento farmacológico , Cinética , Canal de Potasio Kv1.5/efectos de los fármacos , Canal de Potasio Kv1.5/metabolismo , Losartán/uso terapéutico , Miocitos Cardíacos/metabolismo , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio de Rectificación Interna/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , ARN Mensajero/metabolismo , Ratas , Canales de Potasio Shal/efectos de los fármacos , Canales de Potasio Shal/metabolismo
19.
J Pharmacol Exp Ther ; 320(2): 525-34, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17095614

RESUMEN

Direct block of the cardiac potassium channel human ether-a-go-go-related gene (hERG) by a large, structurally diverse group of therapeutic compounds causes drug-induced QT prolongation and torsades de pointes arrhythmias. In addition, several therapeutic compounds have been identified more recently that prolong the QT interval by inhibition of hERG trafficking to the cell surface. We used a surface expression assay to identify novel compounds that interfere with hERG trafficking and found that cardiac glycosides are potent inhibitors of hERG expression at the cell surface. Further investigation of digitoxin, ouabain, and digoxin revealed that all three cardiac glycosides reduced expression of the fully glycosylated cell surface form of hERG on Western blots, indicating that channel exit from the endoplasmic reticulum is blocked. Likewise, hERG currents were reduced with nanomolar affinity on long-term exposure. hERG trafficking inhibition was initiated by cardiac glycosides through direct block of Na(+)/K(+) pumps and not via off-target interactions with hERG or another closely associated protein in its processing or export pathway. In isolated guinea pig myocytes, long-term exposure to 30 nM of the clinically used drugs digoxin or digitoxin reduced hERG/rapidly activating delayed rectifier K(+) current (I(Kr)) currents by approximately 50%, whereas three other cardiac membrane currents--inward rectifier current, slowly activating delayed rectifier K(+) current, and calcium current--were not affected. Importantly, 100 nM digitoxin prolonged action potential duration on long-term exposure consistent with a reduction in hERG/I(Kr) channel number. Thus, cardiac glycosides are able to delay cardiac repolarization at nanomolar concentrations via hERG trafficking inhibition, and this may contribute to the complex electrocardiographic changes seen with compounds such as digitoxin.


Asunto(s)
Glicósidos Cardíacos/farmacología , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Potenciales de Acción/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , Retículo Endoplásmico/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Cobayas , Humanos , Canal de Potasio KCNQ1/efectos de los fármacos , Canal de Potasio KCNQ1/metabolismo , Canal de Potasio Kv1.5/efectos de los fármacos , Canal de Potasio Kv1.5/metabolismo , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/metabolismo , Transporte de Proteínas/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/antagonistas & inhibidores
20.
Am J Physiol Heart Circ Physiol ; 292(2): H1001-8, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17040965

RESUMEN

Little is known about the molecular characteristics of the voltage-activated K(+) (K(v)) channels that underlie the A-type K(+) current in vascular smooth muscle cells of the systemic circulation. We investigated the molecular identity of the A-type K(+) current in retinal arteriolar myocytes using patch-clamp techniques, RT-PCR, immunohistochemistry, and neutralizing antibody studies. The A-type K(+) current was resistant to the actions of specific inhibitors for K(v)3 and K(v)4 channels but was blocked by the K(v)1 antagonist correolide. No effects were observed with pharmacological agents against K(v)1.1/2/3/6 and 7 channels, but the current was partially blocked by riluzole, a K(v)1.4 and K(v)1.5 inhibitor. The current was not altered by the removal of extracellular K(+) but was abolished by flecainide, indicative of K(v)1.5 rather than K(v)1.4 channels. Transcripts encoding K(v)1.5 and not K(v)1.4 were identified in freshly isolated retinal arterioles. Immunofluorescence labeling confirmed a lack of K(v)1.4 expression and revealed K(v)1.5 to be localized to the plasma membrane of the arteriolar smooth muscle cells. Anti-K(v)1.5 antibody applied intracellularly inhibited the A-type K(+) current, whereas anti-K(v)1.4 antibody had no effect. Co-expression of K(v)1.5 with K(v)beta1 or K(v)beta3 accessory subunits is known to transform K(v)1.5 currents from delayed rectifers into A-type currents. K(v)beta1 mRNA expression was detected in retinal arterioles, but K(v)beta3 was not observed. K(v)beta1 immunofluorescence was detected on the plasma membrane of retinal arteriolar myocytes. The findings of this study suggest that K(v)1.5, most likely co-assembled with K(v)beta1 subunits, comprises a major component underlying the A-type K(+) current in retinal arteriolar smooth muscle cells.


Asunto(s)
Canal de Potasio Kv1.5/metabolismo , Músculo Liso Vascular/metabolismo , Vasos Retinianos/metabolismo , Animales , Arteriolas/metabolismo , Inmunohistoquímica , Cinética , Canal de Potasio Kv1.5/análisis , Canal de Potasio Kv1.5/efectos de los fármacos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Músculo Liso Vascular/química , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Técnicas de Placa-Clamp , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Subunidades de Proteína/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Vasos Retinianos/química , Vasos Retinianos/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...