Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 123
Filtrar
1.
J Biol Chem ; 300(5): 107294, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38636665

RESUMEN

Exenatide, a promising cardioprotective agent, protects against cardiac structural remodeling and diastolic dysfunction. Combined blockade of sodium and potassium channels is valuable for managing atrial fibrillation (AF). Here, we explored whether exenatide displayed anti-AF effects by inhibiting human Kv1.5 and Nav1.5 channels. We used the whole-cell patch-clamp technique to investigate the effects of exenatide on hKv1.5 and hNav1.5 channels expressed in human embryonic kidney 293 cells and studied the effects of exenatide on action potential (AP) and other cardiac ionic currents in rat atrial myocytes. Additionally, an electrical mapping system was used to explore the effects of exenatide on electrical properties and AF activity in isolated rat hearts. Finally, a rat AF model, established using acetylcholine and calcium chloride, was employed to evaluate the anti-AF potential of exenatide in rats. Exenatide reversibly suppressed IKv1.5 with IC50 of 3.08 µM, preferentially blocked the hKv1.5 channel in its closed state, and positively shifted the voltage-dependent activation curve. Exenatide also reversibly inhibited INav1.5 with IC50 of 3.30 µM, negatively shifted the voltage-dependent inactivation curve, and slowed its recovery from inactivation with significant use-dependency at 5 and 10 Hz. Furthermore, exenatide prolonged AP duration and suppressed the sustained K+ current (Iss) and transient outward K+ current (Ito), but without inhibition of L-type Ca2+ current (ICa,L) in rat atrial myocytes. Exenatide prevented AF incidence and duration in rat hearts and rats. These findings demonstrate that exenatide inhibits IKv1.5 and INav1.5in vitro and reduces AF susceptibility in isolated rat hearts and rats.


Asunto(s)
Potenciales de Acción , Fibrilación Atrial , Exenatida , Canal de Potasio Kv1.5 , Miocitos Cardíacos , Canal de Sodio Activado por Voltaje NAV1.5 , Bloqueadores del Canal de Sodio Activado por Voltaje , Animales , Humanos , Masculino , Ratas , Potenciales de Acción/efectos de los fármacos , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/metabolismo , Exenatida/farmacología , Exenatida/uso terapéutico , Células HEK293 , Canal de Potasio Kv1.5/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Ratas Sprague-Dawley , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico
2.
J Enzyme Inhib Med Chem ; 37(1): 462-471, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35012386

RESUMEN

Kv1.5 potassium channel, encoded by KCNA5, is a promising target for the treatment of atrial fibrillation, one of the common arrhythmia. A new series of arylmethylpiperidines derivatives based on DDO-02001 were synthesised and evaluated for their ability to inhibit Kv1.5 channel. Among them, compound DDO-02005 showed good inhibitory activity (IC50 = 0.72 µM), preferable anti-arrhythmic effects and favoured safety. These results indicate that DDO-02005 can be a promising Kv1.5 inhibitor for further studies.


Asunto(s)
Diseño de Fármacos , Canal de Potasio Kv1.5/antagonistas & inhibidores , Piperidinas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Canal de Potasio Kv1.5/metabolismo , Estructura Molecular , Piperidinas/síntesis química , Piperidinas/química , Bloqueadores de los Canales de Potasio/síntesis química , Bloqueadores de los Canales de Potasio/química , Relación Estructura-Actividad
3.
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
4.
Pharmacol Rep ; 73(6): 1724-1733, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34146337

RESUMEN

BACKGROUND: Olanzapine, an FDA-approved atypical antipsychotic, is widely used to treat schizophrenia and bipolar disorder. In this study, the inhibitory effect of olanzapine on voltage-dependent K+ (Kv) channels in rabbit coronary arterial smooth muscle cells was investigated. METHODS: Electrophysiological recordings were performed in freshly isolated coronary arterial smooth muscle cells. RESULTS: Olanzapine inhibited the Kv channels in a concentration-dependent manner with an IC50 value of 7.76 ± 1.80 µM and a Hill coefficient of 0.82 ± 0.09. Although olanzapine did not change the steady-state activation curve, it shifted the inactivation curve to a more negative potential, suggesting that it inhibited Kv currents by affecting the voltage sensor of the Kv channel. Application of 1 or 2 Hz train pulses did not affect the olanzapine-induced inhibition of Kv channels, suggesting that its effect on Kv channels occurs in a use (state)-independent manner. Pretreatment with DPO-1 (Kv1.5 subtype inhibitor) reduced the olanzapine-induced inhibition of Kv currents. In addition, pretreatment with guangxitoxin (Kv2.1 subtype inhibitor) and linopirdine (Kv7 subtype inhibitor) partially decreased the degree of Kv current inhibition. Olanzapine induced membrane depolarization. CONCLUSION: From these results, we suggest that olanzapine inhibits the Kv channels in a concentration-dependent, but state-independent, manner by affecting the gating properties of Kv channels. The primary Kv channel target of olanzapine is the Kv1.5 subtype.


Asunto(s)
Antipsicóticos/farmacología , Canal de Potasio Kv1.5/antagonistas & inhibidores , Olanzapina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Animales , Antipsicóticos/administración & dosificación , Vasos Coronarios/citología , Vasos Coronarios/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Concentración 50 Inhibidora , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Olanzapina/administración & dosificación , Bloqueadores de los Canales de Potasio/administración & dosificación , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Conejos
5.
Eur J Pharmacol ; 904: 174158, 2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-33971179

RESUMEN

Voltage-dependent K+ (Kv) channels play the role of returning the membrane potential to the resting state, thereby maintaining the vascular tone. Here, we used native smooth-muscle cells from rabbit coronary arteries to investigate the inhibitory effect of lorcainide, a class Ic antiarrhythmic agent, on Kv channels. Lorcainide inhibited Kv channels in a concentration-dependent manner with an IC50 of 4.46 ± 0.15 µM and a Hill coefficient of 0.95 ± 0.01. Although application of lorcainide did not change the activation curve, it shifted the inactivation curve toward a more negative potential, implying that lorcainide inhibits Kv channels by changing the channels' voltage sensors. The recovery time constant from channel inactivation increased in the presence of lorcainide. Furthermore, application of train steps (of 1 or 2 Hz) in the presence of lorcainide progressively augmented the inhibition of Kv currents, implying that lorcainide-induced inhibition of Kv channels is use (state)-dependent. Pretreatment with Kv1.5 or Kv2.1/2.2 inhibitors effectively reduced the amplitude of the Kv current but did not affect the inhibitory effect of lorcainide. Based on these results, we conclude that lorcainide inhibits vascular Kv channels in a concentration and use (state)-dependent manner by changing their inactivation gating properties. Considering the clinical efficacy of lorcainide, and the pathophysiological significance of vascular Kv channels, our findings should be considered when prescribing lorcainide to patients with arrhythmia and vascular disease.


Asunto(s)
Antiarrítmicos/farmacología , Bencenoacetamidas/farmacología , Vasos Coronarios/metabolismo , Músculo Liso Vascular/metabolismo , Piperidinas/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Animales , Vasos Coronarios/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Cinética , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/metabolismo , Conejos , Canales de Potasio Shab/antagonistas & inhibidores , Canales de Potasio Shab/metabolismo
6.
J Ethnopharmacol ; 276: 114168, 2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-33932511

RESUMEN

ETHNOPHARMACOLOGY RELEVANCE: In folkloric medicine of many cultures, one of the medical uses of Valeriana officinalis Linn is to treat heart-related disease. Recently, it was shown that the ethanol extracts from V. officinalis could effectively prevent auricular fibrillation, and 8-hydroxypinoresinol-4-O-ß-D-glucoside (HPG) from the extracts is one of the two active compounds showing antiarrhythmia activities. AIM OF THE STUDY: The human Kv1.5 channel (hKv1.5) has potential antiarrhythmia activities, and this study arms at investigating the current blocking effects of HPG on hKv1.5 channel. MATERIAL AND METHODS: HPG was obtained from V. officinalis extracts, and hKv1.5 channels were expressed in HEK 293 cells. HPG was perfused while recording the current through hKv1.5 channels. Patch-clamp recording techniques were used to study the effects of HPG at various concentrations (10 µM, 30 µM, and 50 µM) on hKv1.5 channels. RESULTS: The present study demonstrated that HPG inhibited hKv1.5 channel current in a concentration-dependent manner; the higher the concentration, the greater is the inhibition at each depolarization potential. During washout, the channels did not full recover indicating that the un-coupling between HPG and hKv1.5 channels is a slow process. CONCLUSION: HPG may be an effective and safe active ingredient for AF having translational potential.


Asunto(s)
Antiarrítmicos/farmacología , Canal de Potasio Kv1.5/antagonistas & inhibidores , Extractos Vegetales/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Valeriana/química , Potenciales de Acción/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Canal de Potasio Kv1.5/genética , Técnicas de Placa-Clamp , Factores de Tiempo , Verapamilo/farmacología
7.
Eur J Pharmacol ; 891: 173707, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33137332

RESUMEN

Darifenacin, an anticholinergic agent, has been used to treat overactive bladder syndrome. Despite its extensive clinical use, there is little information about the effect of darifenacin on vascular ion channels, specifically K+ channels. This study aimed to investigate the effect of the anti-muscarinic drug darifenacin on voltage-gated K+ (Kv) channels, vascular contractility, and coronary blood flow in rabbit coronary arteries. We used the whole-cell patch-clamp technique to evaluate the effect of darifenacin on Kv channels. Darifenacin inhibited the Kv current in a concentration-dependent manner. Applying 1 µM darifenacin shifted the activation and inactivation curves toward a more positive and negative potential, respectively. Darifenacin slowed the time constants of recovery from inactivation. Furthermore, blockade of the Kv current with darifenacin was increased gradually by applying a train of pulses, indicating that darifenacin inhibited Kv currents in a use- (state)-dependent manner. The darifenacin-mediated inhibition of Kv currents was associated with the Kv1.5 subtype, not the Kv2.1 or Kv7 subtype. Applying another anti-muscarinic drug atropine or ipratropium did not affect the Kv current or change the inhibitory effect of darifenacin. Isometric organ bath experiments using isolated coronary arteries were applied to evaluate whether darifenacin-induced inhibition of the Kv channel causes vasocontraction. Darifenacin substantially induced vasocontraction. Furthermore, darifenacin caused membrane depolarization and decreased coronary blood flow. From these results, we concluded that darifenacin inhibits the Kv currents in concentration- and use- (state)-dependent manners. Inhibition of the Kv current with darifenacin occurred by shifting the steady-state activation and inactivation curves regardless of its anti-muscarinic effect.


Asunto(s)
Benzofuranos/farmacología , Vasos Coronarios/efectos de los fármacos , Canal de Potasio Kv1.5/antagonistas & inhibidores , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Pirrolidinas/farmacología , Vasoconstricción/efectos de los fármacos , Vasoconstrictores/farmacología , Animales , Vasos Coronarios/metabolismo , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Cinética , Canal de Potasio Kv1.5/metabolismo , Masculino , Potenciales de la Membrana , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Conejos
8.
Toxins (Basel) ; 12(11)2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33238397

RESUMEN

Since 1970s, aplysiatoxins (ATXs), a class of biologically active dermatoxins, were identified from the marine mollusk Stylocheilus longicauda, whilst further research indicated that ATXs were originally metabolized by cyanobacteria. So far, there have been 45 aplysiatoxin derivatives discovered from marine cyanobacteria with various geographies. Recently, we isolated two neo-debromoaplysiatoxins, neo-debromoaplysiatoxin G (1) and neo-debromoaplysiatoxin H (2) from the cyanobacterium Lyngbya sp. collected from the South China Sea. The freeze-dried cyanobacterium was extracted with liquid-liquid extraction of organic solvents, and then was subjected to multiple chromatographies to yield neo-debromoaplysiatoxin G (1) (3.6 mg) and neo-debromoaplysiatoxin H (2) (4.3 mg). They were elucidated with spectroscopic methods. Moreover, the brine shrimp toxicity of the aplysiatoxin derivatives representing differential structural classifications indicated that the debromoaplysiatoxin was the most toxic compound (half inhibitory concentration (IC50) value = 0.34 ± 0.036 µM). While neo-aplysiatoxins (neo-ATXs) did not exhibit apparent brine shrimp toxicity, but showed potent blocking action against potassium channel Kv1.5, likewise, compounds 1 and 2 with IC50 values of 1.79 ± 0.22 µM and 1.46 ± 0.14 µM, respectively. Therefore, much of the current knowledge suggests the ATXs with different structure modifications may modulate multiple cellular signaling processes in animal systems leading to the harmful effects on public health.


Asunto(s)
Toxinas de Lyngbya/química , Toxinas de Lyngbya/toxicidad , Lyngbya , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/toxicidad , Animales , Artemia/efectos de los fármacos , Células CHO , Cricetulus , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/fisiología
9.
Mar Drugs ; 18(8)2020 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-32823677

RESUMEN

Recently, Conorfamide-Sr3 (CNF-Sr3) was isolated from the venom of Conus spurius and was demonstrated to have an inhibitory concentration-dependent effect on the Shaker K+ channel. The voltage-gated potassium channels play critical functions on cellular signaling, from the regeneration of action potentials in neurons to the regulation of insulin secretion in pancreatic cells, among others. In mammals, there are at least 40 genes encoding voltage-gated K+ channels and the process of expression of some of them may include alternative splicing. Given the enormous variety of these channels and the proven use of conotoxins as tools to distinguish different ligand- and voltage-gated ion channels, in this work, we explored the possible effect of CNF-Sr3 on four human voltage-gated K+ channel subtypes homologous to the Shaker channel. CNF-Sr3 showed a 10 times higher affinity for the Kv1.6 subtype with respect to Kv1.3 (IC50 = 2.7 and 24 µM, respectively) and no significant effect on Kv1.4 and Kv1.5 at 10 µM. Thus, CNF-Sr3 might become a novel molecular probe to study diverse aspects of human Kv1.3 and Kv1.6 channels.


Asunto(s)
Venenos de Moluscos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de la Superfamilia Shaker/antagonistas & inhibidores , Animales , Caracol Conus , Activación del Canal Iónico , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/genética , Canal de Potasio Kv1.3/metabolismo , Canal de Potasio Kv1.4/antagonistas & inhibidores , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/metabolismo , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Canal de Potasio Kv1.6/antagonistas & inhibidores , Canal de Potasio Kv1.6/genética , Canal de Potasio Kv1.6/metabolismo , Potenciales de la Membrana , Oocitos , Canales de Potasio de la Superfamilia Shaker/genética , Canales de Potasio de la Superfamilia Shaker/metabolismo , Xenopus laevis
10.
Biochem Biophys Res Commun ; 524(4): 791-797, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32019676

RESUMEN

Increased granulosa cell (GC) proliferation may contribute to abnormal folliculogenesis in patients with polycystic ovary syndrome (PCOS), which affects approximately 10% reproductive aged women. However, the mechanisms underlying increased GC proliferation in PCOS remain incompletely understood. In this study, we identified miR-3940-5p as a hub miRNA in GC from PCOS using weighted gene co-expression network analysis (WGCNA), and real-time polymerase chain reaction (RT-PCR) analysis confirmed that miR-3940-5p was significantly increased in GC from PCOS. Enrichment analysis of predicted target genes of miR-3940-5p indicated potential roles of miR-3940-5p in follicular development and cell proliferation regulation. Consistently, functional study confirmed that miR-3940-5p overexpression promoted granulosa cell proliferation. Integrated analysis of mRNA expression profiling data and predicted target genes of miR-3940-5p identified potassium voltage-gated channel subfamily A member 5 (KCNA5) as a potential target of miR-3940-5p, and was validated by luciferase reporter assay. Finally, functional analysis suggested that miR-3940-5p promoted GC proliferation in a KCNA5 dependent manner. In conclusion, miR-3940-5p was a hub miRNA upregulated in GC from PCOS, and promoted GC proliferation by targeting KCNA5.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Células de la Granulosa/metabolismo , Canal de Potasio Kv1.5/genética , MicroARNs/genética , Proteínas de Neoplasias/genética , Síndrome del Ovario Poliquístico/genética , Adulto , Antagomirs/genética , Antagomirs/metabolismo , Proliferación Celular , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Genes Reporteros , Células de la Granulosa/patología , Humanos , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , MicroARNs/antagonistas & inhibidores , MicroARNs/metabolismo , Anotación de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/patología , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
11.
Biomolecules ; 10(1)2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31861703

RESUMEN

The voltage-gated potassium channel Kv1.5, which mediates the cardiac ultra-rapid delayed-rectifier (IKur) current in human cells, has a crucial role in atrial fibrillation. Therefore, the design of selective Kv1.5 modulators is essential for the treatment of pathophysiological conditions involving Kv1.5 activity. This review summarizes the progress of molecular structures and the functionality of different types of Kv1.5 modulators, with a focus on clinical cardiovascular drugs and a number of active natural products, through a summarization of 96 compounds currently widely used. Furthermore, we also discuss the contributions of Kv1.5 and the regulation of the structure-activity relationship (SAR) of synthetic Kv1.5 inhibitors in human pathophysiology. SAR analysis is regarded as a useful strategy in structural elucidation, as it relates to the characteristics that improve compounds targeting Kv1.5. Herein, we present previous studies regarding the structural, pharmacological, and SAR information of the Kv1.5 modulator, through which we can assist in identifying and designing potent and specific Kv1.5 inhibitors in the treatment of diseases involving Kv1.5 activity.


Asunto(s)
Fibrilación Atrial/metabolismo , Canal de Potasio Kv1.5/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Animales , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/genética , Humanos , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Potasio/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Estructura-Actividad
12.
Mar Drugs ; 17(12)2019 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-31766406

RESUMEN

A pair of stereoisomers possessing novel structures with 6/6/5 fused-ring systems, neo-debromoaplysiatoxin E (1) and neo-debromoaplysiatoxin F (2), were isolated from the marine cyanobacterium Lyngbya sp. Their structures were elucidated using various spectroscopic techniques including high resolution electrospray ionization mass spectroscopy (HRESIMS) and nuclear magnetic resonance (NMR). The absolute stereochemistry was determined by calculated electronic circular dichroism (ECD) and gauge-independent atomic orbital (GIAO) NMR shift calculation followed by DP4+ analysis. Significantly, this is the first report on aplysiatoxin derivatives with different absolute configurations at C9-C12 (1: 9S, 10R, 11S, 12S; 2: 9R, 10S, 11R, 12R). Compounds 1 and 2 exhibited potent blocking activities against Kv1.5 with IC50 values of 1.22 ± 0.22 µM and 2.85 ± 0.29 µM, respectively.


Asunto(s)
Organismos Acuáticos/química , Cianobacterias/química , Canal de Potasio Kv1.5/antagonistas & inhibidores , Toxinas de Lyngbya/farmacología , Animales , Células CHO , Dicroismo Circular , Cricetulus , Canal de Potasio Kv1.5/metabolismo , Toxinas de Lyngbya/química , Toxinas de Lyngbya/aislamiento & purificación , Espectroscopía de Resonancia Magnética , Estructura Molecular , Estereoisomerismo
13.
Cell Physiol Biochem ; 52(2): 302-314, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30816676

RESUMEN

BACKGROUND/AIMS: The phenylalkylamine class of L-type Ca2+ channel antagonist verapamil prolongs the effective refractory period (ERP) of human atrium, which appears to contribute to the efficacy of verapamil in preventing reentrant-based atrial arrhythmias including atrial fibrillation. This study was designed to investigate the molecular and electrophysiological mechanism underlying the action of verapamil on human Kv1.5 (hKv1.5) channel that determines action potential duration and ERP in human atrium. METHODS: Site-directed mutagenesis created 10 single-point mutations within pore region of hKv1.5 channel. Wholecell patch-clamp method investigated the effect of verapamil on wild-type and mutant hKv1.5 channels heterologously expressed in Chinese hamster ovary cells. Docking simulation was conducted using open-state homology model of hKv1.5 channel pore. RESULTS: Verapamil preferentially blocked hKv1.5 channel in its open state with IC50 of 2.4±0.6 µM (n = 6). The blocking effect of verapamil was significantly attenuated in T479A, T480A, I502A, V505A, I508A, L510A, V512A and V516A mutants, compared with wild-type hKv1.5 channel. Computer docking simulation predicted that verapamil is positioned within central cavity of channel pore and has contact with Thr479, Thr480, Val505, Ile508, Ala509, Val512, Pro513 and Val516. CONCLUSION: Verapamil acts as an open-channel blocker of hKv1.5 channel, presumably due to direct binding to specific amino acids within pore region of hKv1.5 channel, such as Thr479, Thr480, Val505, Ile508, Val512 and Val516. This blocking effect of verapamil on hKv1.5 channel appears to contribute at least partly to prolongation of atrial ERP and resultant antiarrhythmic action on atrial fibrillation in humans.


Asunto(s)
Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/química , Simulación del Acoplamiento Molecular , Mutación Puntual , Bloqueadores de los Canales de Potasio/química , Verapamilo/química , Sustitución de Aminoácidos , Animales , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Sitios de Unión , Células CHO , Cricetulus , Humanos , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Verapamilo/farmacología
14.
J Org Chem ; 84(8): 4704-4714, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30395712

RESUMEN

BMS-919373 is a highly functionalized quinazoline under investigation as a selective, potent IKur current blocker. By utilizing the aminomethylpyridine side chain at C-4, a selective C-H functionalization at C-5 was invented, enabling the efficient synthesis of this molecule. The strategy of leveraging this inherent directing group allowed the synthesis of this complex heterocycle in only six steps from commodity chemicals. The scope of the C-H activation was further investigated, and the generality of the transformation across a series of bicyclic aromatic heterocycles was explored.


Asunto(s)
Canal de Potasio Kv1.5/antagonistas & inhibidores , Quinazolinas/farmacología , Canal de Potasio Kv1.5/metabolismo , Estructura Molecular , Quinazolinas/síntesis química , Quinazolinas/química
15.
Exp Mol Med ; 50(4): 1-8, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29700292

RESUMEN

MK801 and ketamine, which are phencyclidine (PCP) derivative N-methyl-d-aspartate receptor (NMDAr) blockers, reportedly enhance the function of 5-hydroxytryptamine (HT)-2A receptors (5-HT2ARs). Both are believed to directly affect the pathogenesis of schizophrenia, as well as hypertension. 5-HT2AR signaling involves the inhibition of Kv conductance. This study investigated the interaction of these drugs with Kv1.5, which plays important roles in 5-HT2AR signaling and in regulating the excitability of the cardiovascular and nervous system, and the potential role of this interaction in the enhancement of the 5-HT2AR-mediated response. Using isometric organ bath experiments with arterial rings and conventional whole-cell patch-clamp recording of Chinese hamster ovary (CHO) cells ectopically overexpressing Kv1.5, we examined the effect of ketamine and MK801 on 5-HT2AR-mediated vasocontraction and Kv1.5 channels. Both ketamine and MK801 potentiated 5-HT2AR-mediated vasocontraction. This potentiation of 5-HT2AR function occurred in a membrane potential-dependent manner, indicating the involvement of ion channel(s). Both ketamine and MK801 rapidly and directly inhibited Kv1.5 channels from the extracellular side independently of NMDArs. The potencies of MK801 in facilitating the 5-HT2AR-mediated response and blocking Kv1.5 were higher than those of ketamine. Our data demonstrated the direct inhibition of Kv1.5 channels by MK801/ketamine and indicated that this inhibition may potentiate the functions of 5-HT2ARs. We suggest that 5-HT2AR-Kv1.5 may serve as a receptor-effector module in response to 5-HT and is a promising target in the pathogenesis of MK801-/ketamine-induced disease states such as hypertension and schizophrenia.


Asunto(s)
Maleato de Dizocilpina/farmacología , Ketamina/farmacología , Canal de Potasio Kv1.5/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/farmacología , Agonistas del Receptor de Serotonina 5-HT2/farmacología , Animales , Células CHO , Cricetulus , Hipertensión/metabolismo , Canal de Potasio Kv1.5/metabolismo , Masculino , Técnicas de Placa-Clamp , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT2A/metabolismo , Esquizofrenia/metabolismo , Vasoconstrictores/farmacología
16.
Biochemistry ; 57(18): 2704-2710, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29652491

RESUMEN

Molecular dynamics simulations are employed to determine the inhibitory mechanisms of three drugs, 5-(4-phenoxybutoxy)psoralen (PAP-1), vernakalant, and flecainide, on the voltage-gated K+ channel Kv1.5, a target for the treatment of cardiac arrhythmia. At neutral pH, PAP-1 is neutral, whereas the other two molecules carry one positive charge. We show that PAP-1 forms stable dimers in water, primarily through hydrophobic interactions between aromatic rings. All three molecules bind to the cavity between the Ile508 and Val512 residues from the four subunits of the channel. Once bound, the drug molecules are flexible, with the average root-mean-square fluctuation being between 2 and 3 Å, which is larger than the radius of gyration of a bulky amino acid. The presence of a monomeric PAP-1 causes the permeating K+ ion to dehydrate, thereby creating a significant energy barrier. In contrast, vernakalant blocks the ion permeation primarily via an electrostatic mechanism and, therefore, must be in the protonated and charged form to be effective.


Asunto(s)
Antiarrítmicos/química , Arritmias Cardíacas/tratamiento farmacológico , Canal de Potasio Kv1.5/química , Secuencia de Aminoácidos/genética , Anisoles/química , Anisoles/farmacología , Antiarrítmicos/uso terapéutico , Arritmias Cardíacas/genética , Sitios de Unión , Cristalografía por Rayos X , Ficusina/química , Ficusina/uso terapéutico , Flecainida/química , Flecainida/uso terapéutico , Humanos , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/genética , Simulación de Dinámica Molecular , Conformación Proteica/efectos de los fármacos , Pirrolidinas/química , Pirrolidinas/farmacología , Homología de Secuencia de Aminoácido
17.
J Cardiovasc Pharmacol ; 71(1): 10-18, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29283926

RESUMEN

Propofol blocks the voltage-gated human Kv1.5 (hKv1.5) channel by preferentially affecting in its open state. A previous mutational study suggested that several amino acids within the pore region of the hKv1.5 channel are involved in mediating the blocking action of propofol. The present investigation was undertaken to elucidate the predicted binding modes of propofol within the pore cavity of the open-state hKv1.5 channel, using computational docking and mutagenesis approaches. The docking simulation using a homology model of the hKv1.5 channel, constructed based on the crystal structure of the Kv1.2 channel, predicted that propofol was positioned at the base of the pore cavity of hKv1.5 channel, adjacent to 4 amino acids Thr479, Thr480, Val505, and Ile508, and formed arene-H interactions with Val505. The patch-clamp experiments on wild-type and mutant hKv1.5 channels constructed by site-directed mutagenesis revealed that the blocking potency of propofol was significantly reduced in T480A, V505A, and I508A but not in T479A mutants compared with wild-type hKv1.5 channel. These computational docking and experimental mutational analyses suggest that propofol is positioned at the base of the pore cavity and forms functional contact with Thr480, Val505, and Ile508 to directly block the hKv1.5 channel.


Asunto(s)
Activación del Canal Iónico/efectos de los fármacos , Canal de Potasio Kv1.5/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Mutagénesis Sitio-Dirigida , Bloqueadores de los Canales de Potasio/farmacología , Propofol/farmacología , Animales , Sitios de Unión , Células CHO , Cricetulus , Humanos , Canal de Potasio Kv1.5/química , Canal de Potasio Kv1.5/genética , Canal de Potasio Kv1.5/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Mutación , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/metabolismo , Propofol/química , Propofol/metabolismo , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
18.
Cell Death Dis ; 8(11): e3163, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29095440

RESUMEN

The voltage-gated K+ channel has key roles in the vasculature and in atrial excitability and contributes to apoptosis in various tissues. In this study, we have explored its regulation by carbon monoxide (CO), a product of the cytoprotective heme oxygenase enzymes, and a recognized toxin. CO inhibited recombinant Kv1.5 expressed in HEK293 cells in a concentration-dependent manner that involved multiple signalling pathways. CO inhibition was partially reversed by superoxide dismutase mimetics and by suppression of mitochondrial reactive oxygen species. CO also elevated intracellular nitric oxide (NO) levels. Prevention of NO formation also partially reversed CO inhibition of Kv1.5, as did inhibition of soluble guanylyl cyclase. CO also elevated intracellular peroxynitrite levels, and a peroxynitrite scavenger markedly attenuated the ability of CO to inhibit Kv1.5. CO caused nitrosylation of Kv1.5, an effect that was also observed in C331A and C346A mutant forms of the channel, which had previously been suggested as nitrosylation sites within Kv1.5. Augmentation of Kv1.5 via exposure to hydrogen peroxide was fully reversed by CO. Native Kv1.5 recorded in HL-1 murine atrial cells was also inhibited by CO. Action potentials recorded in HL-1 cells were increased in amplitude and duration by CO, an effect mimicked and occluded by pharmacological inhibition of Kv1.5. Our data indicate that Kv1.5 is a target for modulation by CO via multiple mechanisms. This regulation has important implications for diverse cellular functions, including excitability, contractility and apoptosis.


Asunto(s)
Monóxido de Carbono/farmacología , Canal de Potasio Kv1.5/metabolismo , Animales , Monóxido de Carbono/química , Monóxido de Carbono/metabolismo , Línea Celular , Células HEK293 , Humanos , Peróxido de Hidrógeno/toxicidad , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/genética , Metaloporfirinas/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutagénesis Sitio-Dirigida , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
19.
Eur J Pharmacol ; 809: 105-110, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28502629

RESUMEN

Levo-tetrahydropalmatine (l-THP) exerts various pharmacological effects on neural and cardiac tissues and K+ channel can be one of its multiple targets. The rapidly activating Kv1.5 channel is expressed in a variety of tissues including atrial cells and hippocampal neurons, and has an essential role in tuning the action potential and excitability in those cells. The aim of current study is to explore whether there are the possible effects of l-THP on Kv1.5 channels expressed in HEK293 cells. Superfusion of l-THP led to a dose-dependent blockage of Kv1.5 currents with an IC50 value of 53.2µM. This blocking effect was substantially attenuated in mutant H452G rather than R476V and R476Y, suggesting a specific binding site in the outer mouth region. In addition, the properties of Kv1.5 channel kinetics were markedly altered by l-THP. Treatment with l-THP resulted in a potential left shift of the inactivation curve, with the half-maximum inactivation potential (V1/2) of 4.5mV in control and -12.8mV in 50µM l-THP. Our data reveal that l-THP can exert an inhibitory effect on the delayed rectifier Kv1.5 channels expressed in HEK293 cells. These lines of evidence provided an insight to understand the possible effects exerted by l-THP on relative tissues.


Asunto(s)
Alcaloides de Berberina/farmacología , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/genética , Bloqueadores de los Canales de Potasio/farmacología , Animales , Fenómenos Electrofisiológicos/efectos de los fármacos , Expresión Génica , Ratas
20.
Toxicol Appl Pharmacol ; 322: 89-96, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28284858

RESUMEN

Excessive consumption of alcohol is a well-established risk factor of atrial fibrillation (AF). However, the effects of moderate alcohol drinking remain to be elucidated. This study was designed to determine the effects of moderate ethanol ingestion on atrial fibrillation and the electrophysiological mechanisms. In acetylcholine-induced canine and mouse AF models, the moderate ethanol prevented the generation and persistence of AF through prolonging the latent period of AF and shortening the duration of AF. The action potential duration (APD) was remarkably prolonged under the concentration range of 12.5-50.0mM ethanol in guinea pig atrial myocytes. Ultra-rapid delayed rectified potassium currents (IKv1.5) were markedly inhibited by 12.5-50.0mM ethanol in a concentration-dependent manner. Ethanol with 50.0mM could inhibit rapid delayed rectifier potassium currents (IhERG). Ethanol under 6.25-50.0mM did not affect on inward rectifier potassium currents (IKir2.1). Collectively, the present study provided an evidence that moderate ethanol intake can prolong the APD of atrial myocytes by inhibition of IKv1.5 and IhERG, which contributed to preventing the development and duration of AF.


Asunto(s)
Antiarrítmicos/administración & dosificación , Fibrilación Atrial/tratamiento farmacológico , Etanol/administración & dosificación , Canal de Potasio Kv1.5/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Bloqueadores de los Canales de Potasio/administración & dosificación , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Fibrilación Atrial/fisiopatología , Perros , Relación Dosis-Respuesta a Droga , Cobayas , Células HEK293 , Humanos , Canal de Potasio Kv1.5/fisiología , Masculino , Ratones , Ratones Endogámicos ICR , Miocitos Cardíacos/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA