Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Pharmacol Sci ; 140(3): 284-290, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31481348

RESUMEN

The human ether-a-go-go-related gene (hERG) encodes the K+ channel that carries the rapid component of the delayed rectifier current in the human heart. Reduction of hERG activity induced by gene mutations or pharmacological inhibition is responsible for the type 2 form of long QT syndrome in patients which can develop into ventricular arrhythmia and sudden cardiac death. Therefore, pharmacological activation of hERG may lead to therapeutic potential for cardiac arrhythmias. In this study we characterized a small and novel compound, N-(2-(tert-butyl)phenyl)-6-(4-chlorophenyl)-4-(trifluoromethyl) nicotinamide, HW-0168, that exhibits potent activation of hERG channel with an EC50 of 0.41 ± 0.2 µM. Using whole-cell patch clamp recording of HEK293 cells stably expressed hERG channels, we found that HW-0168 dramatically increased current amplitude about 2.5 folds and slowed down current inactivation about 4 folds. HW-0168 shifted the voltage-dependent channel activation to hyperpolarizing direction about 3.7 mV and the voltage-dependent channel inactivation to depolarizing direction about 9.4 mV. In addition, recording of guinea-pig ventricular cells confirmed that HW-0168 shortened the action potential duration. In conclusion, we identified a novel hERG channel activator HW-0168 that can be used for studying the physiological role of hERG in cardiac myocytes and may be beneficial for treating long QT syndrome.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/metabolismo , Línea Celular , Cobayas , Células HEK293 , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Humanos , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/metabolismo , Masculino
2.
J Med Chem ; 61(21): 9500-9512, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30346167

RESUMEN

Inhibitors of the voltage-gated sodium channel NaV1.7 are being investigated as pain therapeutics due to compelling human genetics. We previously identified NaV1.7-inhibitory peptides GpTx-1 and JzTx-V from tarantula venom screens. Potency and selectivity were modulated through attribute-based positional scans of native residues via chemical synthesis. Herein, we report JzTx-V lead optimization to identify a pharmacodynamically active peptide variant. Molecular docking of peptide ensembles from NMR into a homology model-derived NaV1.7 structure supported prioritization of key residues clustered on a hydrophobic face of the disulfide-rich folded peptide for derivatization. Replacing Trp24 with 5-Br-Trp24 identified lead peptides with activity in electrophysiology assays in engineered and neuronal cells. 5-Br-Trp24 containing peptide AM-6120 was characterized in X-ray crystallography and pharmacokinetic studies and blocked histamine-induced pruritis in mice after subcutaneous administration, demonstrating systemic NaV1.7-dependent pharmacodynamics. Our data suggests a need for high target coverage based on plasma exposure for impacting in vivo end points with selectivity-optimized peptidic NaV1.7 inhibitors.


Asunto(s)
Descubrimiento de Drogas , Histamina/efectos adversos , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Péptidos/química , Péptidos/farmacología , Prurito/tratamiento farmacológico , Venenos de Araña/química , Animales , Células HEK293 , Humanos , Ratones , Simulación del Acoplamiento Molecular , Canal de Sodio Activado por Voltaje NAV1.7/química , Péptidos/farmacocinética , Péptidos/uso terapéutico , Conformación Proteica , Pliegue de Proteína , Prurito/inducido químicamente , Relación Estructura-Actividad , Distribución Tisular , Bloqueadores del Canal de Sodio Activado por Voltaje/química , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacocinética , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/uso terapéutico
3.
PLoS One ; 13(5): e0196791, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29723257

RESUMEN

Identification of voltage-gated sodium channel NaV1.7 inhibitors for chronic pain therapeutic development is an area of vigorous pursuit. In an effort to identify more potent leads compared to our previously reported GpTx-1 peptide series, electrophysiology screening of fractionated tarantula venom discovered the NaV1.7 inhibitory peptide JzTx-V from the Chinese earth tiger tarantula Chilobrachys jingzhao. The parent peptide displayed nominal selectivity over the skeletal muscle NaV1.4 channel. Attribute-based positional scan analoging identified a key Ile28Glu mutation that improved NaV1.4 selectivity over 100-fold, and further optimization yielded the potent and selective peptide leads AM-8145 and AM-0422. NMR analyses revealed that the Ile28Glu substitution changed peptide conformation, pointing to a structural rationale for the selectivity gains. AM-8145 and AM-0422 as well as GpTx-1 and HwTx-IV competed for ProTx-II binding in HEK293 cells expressing human NaV1.7, suggesting that these NaV1.7 inhibitory peptides interact with a similar binding site. AM-8145 potently blocked native tetrodotoxin-sensitive (TTX-S) channels in mouse dorsal root ganglia (DRG) neurons, exhibited 30- to 120-fold selectivity over other human TTX-S channels and exhibited over 1,000-fold selectivity over other human tetrodotoxin-resistant (TTX-R) channels. Leveraging NaV1.7-NaV1.5 chimeras containing various voltage-sensor and pore regions, AM-8145 mapped to the second voltage-sensor domain of NaV1.7. AM-0422, but not the inactive peptide analog AM-8374, dose-dependently blocked capsaicin-induced DRG neuron action potential firing using a multi-electrode array readout and mechanically-induced C-fiber spiking in a saphenous skin-nerve preparation. Collectively, AM-8145 and AM-0422 represent potent, new engineered NaV1.7 inhibitory peptides derived from the JzTx-V scaffold with improved NaV selectivity and biological activity in blocking action potential firing in both DRG neurons and C-fibers.


Asunto(s)
Analgésicos/aislamiento & purificación , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Péptidos/química , Bloqueadores de los Canales de Sodio/aislamiento & purificación , Venenos de Araña/química , Potenciales de Acción/efectos de los fármacos , Sustitución de Aminoácidos , Analgésicos/farmacología , Animales , Capsaicina/farmacología , Línea Celular , Evaluación Preclínica de Medicamentos , Ganglios Espinales/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Fibras Nerviosas Amielínicas/efectos de los fármacos , Resonancia Magnética Nuclear Biomolecular , Técnicas de Placa-Clamp , Estimulación Física , Ingeniería de Proteínas , Proteínas Recombinantes/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Relación Estructura-Actividad , Tetrodotoxina/farmacología
4.
J Med Chem ; 59(6): 2704-17, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26890998

RESUMEN

There is interest in the identification and optimization of new molecular entities selectively targeting ion channels of therapeutic relevance. Peptide toxins represent a rich source of pharmacology for ion channels, and we recently reported GpTx-1 analogs that inhibit NaV1.7, a voltage-gated sodium ion channel that is a compelling target for improved treatment of pain. Here we utilize multi-attribute positional scan (MAPS) analoging, combining high-throughput synthesis and electrophysiology, to interrogate the interaction of GpTx-1 with NaV1.7 and related NaV subtypes. After one round of MAPS analoging, we found novel substitutions at multiple residue positions not previously identified, specifically glutamic acid at positions 10 or 11 or lysine at position 18, that produce peptides with single digit nanomolar potency on NaV1.7 and 500-fold selectivity against off-target sodium channels. Docking studies with a NaV1.7 homology model and peptide NMR structure generated a model consistent with the key potency and selectivity modifications mapped in this work.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Péptidos/farmacología , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacología , Venenos de Araña/farmacología , Secuencia de Aminoácidos , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Especificidad por Sustrato
5.
Bioorg Med Chem Lett ; 25(21): 4866-4871, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26112439

RESUMEN

Many efforts are underway to develop selective inhibitors of the voltage-gated sodium channel NaV1.7 as new analgesics. Thus far, however, in vitro selectivity has proved difficult for small molecules, and peptides generally lack appropriate pharmacokinetic properties. We previously identified the NaV1.7 inhibitory peptide GpTx-1 from tarantula venom and optimized its potency and selectivity via structure-guided analoging. To further understand GpTx-1 binding to NaV1.7, we have mapped the binding site to transmembrane segments 1-4 of the second pseudosubunit internal repeat (commonly referred to as Site 4) using NaV1.5/NaV1.7 chimeric protein constructs. We also report that select GpTx-1 amino acid residues apparently not contacting NaV1.7 can be derivatized with a hydrophilic polymer without adversely affecting peptide potency. Homodimerization of GpTx-1 with a bifunctional polyethylene glycol (PEG) linker resulted in a compound with increased potency and a significantly reduced off-rate, demonstrating the ability to modulate the function and properties of GpTx-1 by linking to additional molecules.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Péptidos/química , Péptidos/farmacología , Ingeniería de Proteínas , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Dimerización , Relación Dosis-Respuesta a Droga , Humanos , Conformación Molecular , Unión Proteica , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Bloqueadores del Canal de Sodio Activado por Voltaje/química
6.
J Med Chem ; 58(5): 2299-314, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25658507

RESUMEN

NaV1.7 is a voltage-gated sodium ion channel implicated by human genetic evidence as a therapeutic target for the treatment of pain. Screening fractionated venom from the tarantula Grammostola porteri led to the identification of a 34-residue peptide, termed GpTx-1, with potent activity on NaV1.7 (IC50 = 10 nM) and promising selectivity against key NaV subtypes (20× and 1000× over NaV1.4 and NaV1.5, respectively). NMR structural analysis of the chemically synthesized three disulfide peptide was consistent with an inhibitory cystine knot motif. Alanine scanning of GpTx-1 revealed that residues Trp(29), Lys(31), and Phe(34) near the C-terminus are critical for potent NaV1.7 antagonist activity. Substitution of Ala for Phe at position 5 conferred 300-fold selectivity against NaV1.4. A structure-guided campaign afforded additive improvements in potency and NaV subtype selectivity, culminating in the design of [Ala5,Phe6,Leu26,Arg28]GpTx-1 with a NaV1.7 IC50 value of 1.6 nM and >1000× selectivity against NaV1.4 and NaV1.5.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/química , Fragmentos de Péptidos/farmacología , Venenos de Araña/farmacología , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología , Animales , Electrofisiología , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.7/sangre , Fragmentos de Péptidos/química , Conformación Proteica , Ratas , Espectrometría de Masa por Ionización de Electrospray , Venenos de Araña/química , Arañas , Relación Estructura-Actividad , Bloqueadores del Canal de Sodio Activado por Voltaje/química
7.
J Med Chem ; 54(13): 4427-45, 2011 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-21634377

RESUMEN

Clinical human genetic studies have recently identified the tetrodotoxin (TTX) sensitive neuronal voltage gated sodium channel Nav1.7 (SCN9A) as a critical mediator of pain sensitization. Herein, we report structure-activity relationships for a novel series of 2,4-diaminotriazines that inhibit hNav1.7. Optimization efforts culminated in compound 52, which demonstrated pharmacokinetic properties appropriate for in vivo testing in rats. The binding site of compound 52 on Nav1.7 was determined to be distinct from that of local anesthetics. Compound 52 inhibited tetrodotoxin-sensitive sodium channels recorded from rat sensory neurons and exhibited modest selectivity against the hERG potassium channel and against cloned and native tetrodotoxin-resistant sodium channels. Upon oral administration to rats, compound 52 produced dose- and exposure-dependent efficacy in the formalin model of pain.


Asunto(s)
Acetamidas/síntesis química , Analgésicos/síntesis química , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/síntesis química , Triazinas/síntesis química , Acetamidas/farmacocinética , Acetamidas/farmacología , Administración Oral , Analgésicos/farmacocinética , Analgésicos/farmacología , Animales , Sitios de Unión , Línea Celular , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Formaldehído , Ganglios Espinales/citología , Humanos , Técnicas In Vitro , Microsomas Hepáticos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1 , Neuronas/efectos de los fármacos , Neuronas/fisiología , Dimensión del Dolor , Técnicas de Placa-Clamp , Ratas , Bloqueadores de los Canales de Sodio/farmacocinética , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio , Solubilidad , Relación Estructura-Actividad , Tetrodotoxina/farmacología , Triazinas/farmacocinética , Triazinas/farmacología
8.
J Ethnopharmacol ; 131(1): 187-95, 2010 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-20600762

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Aconitum has been widely used to treat various diseases in China for a long time. However, improper use of this drug results in severe intoxication. Aconitine (ACO), a diterpenoid alkaloid from aconitum, mainly contributes to cardio-toxic effects of aconitum and has also been commonly known to induce arrhythmias in animal models. However, its pro-arrhythmic mechanisms are not clear. AIM OF THE STUDY: The effects of ACO on HERG and Kv1.5 channels were investigated. MATERIALS AND METHODS: HERG and Kv1.5 channels were expressed in Xenopus laevis oocytes, and the resulting currents were recorded using a two-microelectrode voltage clamp technique. RESULTS: In HERG channels, ACO exhibited a blockade in a voltage- and time-dependent manner. The blockade was enhanced by further activation of currents, which were consistent with an open-channel blockade. In Kv1.5 channels, ACO produced a voltage-, time-, and frequency-dependent inhibition. The blockade was enhanced by higher rates of stimulation, consistent with preferential binding of the drug to the open state. In addition, ACO blocked Kv1.5 and HERG channels in a concentration-dependent manner with an IC(50) of 0.796+/-0.123 and 1.801+/-0.332 microM, respectively. CONCLUSIONS: ACO blocks HERG and Kv1.5 potassium channels in the open state. Blockade of potassium channels, particular the HERG channel, may be one of the important mechanisms of how ACO induces arrhythmias.


Asunto(s)
Aconitina/farmacología , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canal de Potasio Kv1.5/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/farmacología , Animales , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/fisiología , Femenino , Humanos , Canal de Potasio Kv1.5/fisiología , Xenopus laevis
9.
Chin J Integr Med ; 16(5): 430-4, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20535583

RESUMEN

OBJECTIVE: To observe the effect of matrine on human ether à go-go related gene (HERG) potassium channels expressed in Chinese hamster ovary (CHO) cells and investigate whether HERG channel is a new target of the pharmacological effect of matrine on arrhythmia and tumor METHODS: HERG channel potassium current in CHO cell was recorded using whole-cell patch-clamp technique, and the influence of matrine on the current was explored. RESULTS: Matrine inhibited HERG potassium current in a dose-dependent manner, and the 50% inhibitory concentration (IC IC(50)) was 411±23 µmol/L. Matrine had no significant effect on the activation kinetics, and mainly blocked HERG channels in their closed state. CONCLUSIONS: The blocking effect of matrine on HERG channels might be one of the mechanisms against arrythmias and tumors. Unlike most other blockers exerting blocking effect at the intracellular sites by entering the cell with the opening of HERG channel, matrine blocked HERG channels at the extracellular sites.


Asunto(s)
Alcaloides/farmacología , Canales de Potasio Éter-A-Go-Go/genética , Quinolizinas/farmacología , Animales , Células CHO , Cricetinae , Cricetulus , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Matrinas
10.
Exp Cell Res ; 315(13): 2256-64, 2009 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-19409379

RESUMEN

Stromal cell-derived factor-1 (SDF-1) and its unique receptor, CXCR4, regulate stem/progenitor cell migration and retention in the bone marrow and are required for hematopoiesis. Recent studies found that hERG1 K(+) channels were important regulators of tumor cell migration. In this study, we investigated whether SDF-1 induced acute leukemic cell migration associated with hERG1 K(+) channels. Our results showed that E-4031, a specific hERG1 K(+) channels inhibitor, significantly blocked SDF-1-induced migration of leukemic cell lines, primary acute leukemic cells, leukemic stem cells and HEK293T cells transfected with herg-pEGFP. The migration of phenotypically recognizable subsets gave the indication that lymphoblastic leukemic cells were inhibited more than myeloid cells while in the presence of E-4031 which maybe associated with herg expression. SDF-1 increased hERG1 K(+) current expressed in oocytes and HEK293T cells transfected with herg-pEGFP. There were no significant changes of CXCR4 expression on both HL-60 cells and primary leukemic cells regardless if untreated or treated with E-4031 for 24 h (P>0.05). The hERG1 K(+) current increased by SDF-1 might contribute to the mechanism of SDF-1-induced leukemic cell migration. The data suggested that hERG1 K(+) channels functionally linked to cell migration induced by SDF-1.


Asunto(s)
Movimiento Celular/fisiología , Quimiocina CXCL12/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Adolescente , Adulto , Anciano , Animales , Antiarrítmicos/metabolismo , Niño , Preescolar , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Femenino , Células HL-60 , Humanos , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/fisiología , Oocitos/citología , Oocitos/fisiología , Piperidinas/metabolismo , Piridinas/metabolismo , Receptores CXCR4/metabolismo , Xenopus laevis , Adulto Joven
11.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 25(5): 1068-73, 2008 Oct.
Artículo en Chino | MEDLINE | ID: mdl-19024448

RESUMEN

We have investigated the methods and mechanisms for analysis of the channel kinetics parameters of voltage-gated potassium channels, HERG (Human ether-à-go-go related gene) channels, in the process of electrophysiological recording. The current of HERG K+ channels expressed in Xenopus oocytes was studied using a two-electrode voltage clamp technique, and the channel kinetics parameters were analyzed through compiling different pulse protocol and recording the current. Results showed: (1) The HERG K+ channels, under conditions of being activated with depolarized pulse, expressed an inward-rectified property attributing to rapid inactivation. The activation curve could be obtained through fitting the depolarized potential and the following peak amplitude of tail current, while the parameters of time-dependent activation was obtained through fitting different depolarized duration and the corresponding peak amplitude of tail current. (2) The I-V relationship still exhibit marked inward rectification. Tail current decay traces were fitted with a bi-exponential function to determine the time constants of the fast and slow components of current decay. (3) The inactivation of HERG channels is voltage-dependent. The inactivation process was isolated with two different three-pulse protocols, with which the inactivation curve and nearly linear I-V relationship were obtained, respectively. Thus, altough the kinetics properties of HERG channels were complicated, the channels kinetics could be indirectly analyzed through differently designed pulse protocols, which provided the basis for investigation on Alanine-scanning mutagenesis and agent action.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/análisis , Canales de Potasio Éter-A-Go-Go/genética , Animales , Canal de Potasio ERG1 , Humanos , Cinética , Oocitos/metabolismo , Técnicas de Placa-Clamp , Xenopus laevis/metabolismo
12.
Sheng Li Xue Bao ; 60(4): 525-34, 2008 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-18690396

RESUMEN

In the present study, we investigated the inhibitory action of ketanserin on wild-type (WT) and Y652 mutant human ether-a-go-go-related gene (HERG) potassium channels expressed in Xenopus oocytes and the effects of changing the channel molecular determinants characteristics on the blockade with and without ketanserin intervention using standard two-microelectrode voltage-clamp techniques. Point mutations were introduced into HERG gene (Y652A and Y652R) and subcloned into the pSP64 plasmid expression vector. Complementary RNAs for injection into oocytes were prepared with SP6 Cap-Scribe after linearization of the expression construct with EcoR I. Clampfit 9.2 software was employed for data collection and analysis. Origin 6.0 software was used to fit the data, calculate time constants and plot histograms. The results showed that ketanserin blocked WT HERG currents in voltage- and concentration-dependent manner and showed minimal tonic blockade of HERG current evaluated by the envelope of tails test. The IC50 value was (0.38+/-0.04) micromol/L for WT HERG potassium channel. The peaks of the I-V relationship for HERG channel suggested a negative shift in the voltage-dependence of activation after using ketanserin, whose midpoint of activation values (V1/2) were (-16.59+/-1.01) mV (control) vs (-20.59+/-0.87) mV (ketanserin) at 0.1 micromol/L, (-22.39+/-0.94) mV at 1 micromol/L, (-23.51+/-0.91) mV at 10 micromol/L, respectively (P<0.05, n=6). Characteristics of blockade were consistent with an open-state channel blockade, because the extent and rate of onset of blockade was voltage-dependent, increasing at more potentials even in the condition of leftward shift of activation curve. Meanwhile, in the different depolarization duration, the fractional blockade of end-pulse step current and peak tail current at 100 ms duration was significantly lower than that at 400 ms and 700 ms, which indicated that following the channel activation fractional blockade was enhanced by the activated channels. Ketanserin could also modulate the inactivation of HERG channel, which shifted the voltage-dependence of WT HERG channel inactivation curve from (-51.71+/-2.15) mV to (-80.76+/-14.98) mV (P<0.05, n=4). The S6 mutation, Y652A and Y652R, significantly attenuated the blockade by ketanserin. The IC50 value were (27.13+/-9.40) micromol/L and (20.20+/-2.80) micromol/L, respectively, increased by approximately 72-fold for Y652A and 53-fold for Y652R compared to that of WT HERG channel blockade [(0.38+/-0.04) micromol/L]. However, between the inhibitory effects of Y652A and Y652R, there was no significant difference. In conclusion, ketanserin blocks WT HERG currents in voltage- and concentration-dependent manner and preferentially blocks open-state HERG channels. Tyr-652 is one of the critical residues in the ketanserin-binding sites.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Ketanserina/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Animales , Humanos , Mutación , Oocitos , Técnicas de Placa-Clamp , Xenopus
13.
Acta Pharmacol Sin ; 29(8): 913-22, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18664324

RESUMEN

AIM: The objectives of this study were to investigate the inhibitory action of telmisartan, a selective angiotensin II type 1 receptor antagonist, on hKv1.5 and human ether-a-go-go-related gene (HERG) channels expressed on Xenopus laevis oocytes. METHODS: hKv1.5 and HERG channels were expressed on Xenopus laevis oocytes and studied using the 2-microelectrode voltage clamp technique. RESULTS: In hKv1.5 channels, telmisartan produced a voltage- and concentration-dependent inhibition; the efficacies of blockade were different at peak and 1.5 s end-pulse currents, which were 7.75%+/-2.39% (half-maximal inhibition concentration [IC50]=2.25+/-0.97 micromol/L) and 52.64%+/-3.77% (IC50=0.82+/-0.39 micromol/L) at 1 micromol/L telmisartan, respectively. Meanwhile, telmisartan accelerated the inactivation of the channels. However, telmisartan exhibited a low affinity for HERG channels (IC50=24.35+/-5.06 micromol/L); the blockade was voltage- and concentration-dependent. Telmisartan preferentially blocked open-state HERG channels. The slow time constants of deactivation were accelerated (n=6, P<0.05), which was inconsistent with the "foot-in-the-door"effect. CONCLUSION: Telmisartan blocks hKv1.5 potassium channels involving open and inactivated states at plasma concentration levels of therapeutic doses; whereas the blockade of HERG channels occurs only at supra plasma concentration levels of therapeutic doses and preferentially in open and closed-state channels.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bencimidazoles/farmacología , Benzoatos/farmacología , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canal de Potasio Kv1.5/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio , Animales , Relación Dosis-Respuesta a Droga , Electrofisiología , Femenino , Humanos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Telmisartán , Xenopus laevis
14.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 24(3): 329-33, 2008 Aug.
Artículo en Chino | MEDLINE | ID: mdl-21141595

RESUMEN

AIM: To explore a method of the stable and persistent expression of HERG(human ether-a-go-go-related gene) channels in Xenopus oocytes, and investigate the alteration of rest membrane potential of oocytes and electrophysiological properties of expressed channel in different culture duration. METHODS: HERG mRNA for injection was prepared with in intro transcription using vector plasmid pSP64 containing HERG cDNA fragment. Expressed HERG current was recorded using standard two-microelectrode voltage-clamp technique. RESULTS: (1) Functional channels, with electrophysiological properties consistent with those of HERG channels were persistently expressed in oocytes membrane with this method. Furthermore, channel current could be recorded stably in 10-15 days. (2) The negative value of rest membrane potential increased gradually in the 3, 6, and 9 days of culture, and then decreased in the 12 days. The potential of peak value of inward rectification shifted gradually to the positive direction in 3, 6 and 9 days, and recovered in 12 days. Half-maximal activation potential (V1/2) of heterological expressed current shifted gradually to the negative direction in 3, 6 and 9 days of culture and then recovered in 12 days, the tendency of change was coincident with that of membrane rest potential. CONCLUSION: The investigation provides a method of persistent expression of HERG channel in Xenopus oocytes and offers evidences for the difference of electrophysiological experimental data of studies of molecular site and drugs effect of HERG channel in different experimental conditions.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Potenciales de la Membrana , Oocitos/metabolismo , Animales , Canales de Potasio Éter-A-Go-Go/genética , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Xenopus laevis
15.
Am J Physiol Cell Physiol ; 285(6): C1356-66, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12890647

RESUMEN

The Elk subfamily of the Eag K+ channel gene family is represented in mammals by three genes that are highly conserved between humans and rodents. Here we report the distribution and functional properties of a member of the human Elk K+ channel gene family, KCNH8. Quantitative RT-PCR analysis of mRNA expression patterns showed that KCNH8, along with the other Elk family genes, KCNH3 and KCNH4, are primarily expressed in the human nervous system. KCNH8 was expressed at high levels, and the distribution showed substantial overlap with KCNH3. In Xenopus oocytes, KCNH8 gives rise to slowly activating, voltage-dependent K+ currents that open at hyperpolarized potentials (half-maximal activation at -62 mV). Coexpression of KCNH8 with dominant-negative KCNH8, KCNH3, and KCNH4 subunits led to suppression of the KCNH8 currents, suggesting that Elk channels can form heteromultimers. Similar experiments imply that KCNH8 subunits are not able to form heteromultimers with Eag, Erg, or Kv family K+ channels.


Asunto(s)
Encéfalo/fisiología , Filogenia , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/fisiología , Secuencia de Aminoácidos , Animales , Canales de Potasio Éter-A-Go-Go , Humanos , Potenciales de la Membrana/fisiología , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/fisiología , Técnicas de Placa-Clamp , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA