Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Arch Biochem Biophys ; 667: 36-48, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-31047869

RESUMEN

Piperine is an attractive therapeutic alkaloid from black pepper that exhibits a broad spectrum of pharmacological properties over various pathological disorders including cancer. Voltage gated K+ (KV) channels play an important role in regulating cancer cell proliferation and are considered as potential target for cancer treatment. However, the implication of piperine in KV associated anticancer activities on human prostate cancer cells LNCaP and PC-3 remains unrevealed. The electrophysiological and pharmacological data identifies that both androgen sensitive (LNCaP) and insensitive (PC-3) prostate cancer cells typically expressed voltage gated K+ current (IK). This current was significantly blocked by piperine in a concentration-dependent manner with an IC50 value 39.91 µM in LNCaP and 49.45 µM in PC-3 cells. Analysis of voltage-dependence of activation kinetics showed that piperine induces a positive shift in the relative activation curve in both the cells. Piperine also depolarized the resting membrane potential by an average of 10.2 mV and 8.3 mV in LNCaP and PC-3 cells, respectively. The anticancer studies showed that, treatment with piperine concentration dependently induced G1 phase cell cycle arrest and apoptosis in LNCaP and PC-3 cells. These results unravel that the IK inhibition might be responsible for the anticancer effect of piperine on androgen sensitive and insensitive human prostate cancer cells.


Asunto(s)
Alcaloides/farmacología , Antineoplásicos Fitogénicos/farmacología , Benzodioxoles/farmacología , Piperidinas/farmacología , Alcamidas Poliinsaturadas/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Células PC-3 , Técnicas de Placa-Clamp , Fitoterapia , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología
2.
Microvasc Res ; 117: 22-27, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29247720

RESUMEN

Local skin heating to 42°C causes rapid increases in cutaneous perfusion (initial peak), followed by a brief nadir and subsequent sustained elevation (plateau). Several studies have demonstrated that nitric oxide synthase (NOS) largely contributes to the plateau response during local heating. In this study, we tested the hypothesis that voltage-gated potassium (Kv) channels contribute to the plateau of the cutaneous vasodilation during local heating through NOS-dependent mechanisms. Eleven young males (25±4years) participated in this study wherein cutaneous vascular conductance (CVC) was measured at four intradermal microdialysis sites that were continuously perfused with either 1) lactated Ringer (Control), 2) 10mM 4-aminopyridine (Kv channel blocker), 3) 10mM Nω-Nitro-L-arginine (NOS inhibitor), or 4) a combination of 4-aminopyridine and Nω-Nitro-L-arginine. In comparison to the Control site, the inhibition of Kv channels alone attenuated the increase in CVC observed at the initial peak, nadir, and plateau phases measured during local heating; in contrast, the inhibition of NOS alone attenuated the increase in CVC at the nadir and plateau phases only (e.g., plateau response: Control site: 59±5%max, Kv channel blockade site: 49±8%max, NOS inhibition site: 35±11%max, combined inhibition site: 40±12%max). Further, no effect of Kv channel blockade on CVC was measured at any phase of the local heating response when the modulating influence of NOS was simultaneously removed. We show that Kv channels and NOS contribute to the local heating mediated sustained increase (i.e., plateau) in cutaneous vasodilation in an interactive manner. (243/250 words).


Asunto(s)
Arterias/enzimología , Calefacción , Hipertermia Inducida , Óxido Nítrico Sintasa/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Piel/irrigación sanguínea , Vasodilatación , Administración Cutánea , Adulto , Arterias/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Humanos , Masculino , Microdiálisis , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/administración & dosificación , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Transducción de Señal , Vasodilatación/efectos de los fármacos , Adulto Joven
3.
Am J Physiol Heart Circ Physiol ; 313(3): H548-H557, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28626074

RESUMEN

Recent studies have supported an important contribution of prorenin (PR) and its receptor (PRR) to the regulation of hypothalamic, sympathetic, and neurosecretory outflows to the cardiovascular system, including systemic release of vasopressin (VP), both under physiological and cardiovascular disease conditions. Still, the identification of precise cellular mechanisms and neuronal/molecular targets remain unknown. We have recently shown that PRR is expressed in VP neurons and that their activation increases neuronal activity. However, the underlying ionic channel mechanisms are undefined. Here, we performed patch-clamp electrophysiology from identified VP neurons in acute hypothalamic slices obtained from enhanced green fluorescent protein-VP transgenic rats. Voltage-clamp recordings showed that PR inhibited the magnitude of A-type K+ current (IA; ~50% at -25 mV), a subthreshold voltage-dependent current that restrains VP firing activity. PR also increased the inactivation rate of IA and shifted the steady-state voltage-dependent inactivation function toward more hyperpolarized membrane potential (~7 mV shift), thus resulting in less channel availability to be activated at any given membrane potential. PR also inhibited a sustained component of IA ("window" current). PR-mediated changes in action potential waveform and increased firing activity were occluded when IA was blocked by 4-aminopyridine. Finally, PR failed to increase superoxide production within the supraoptic nucleus/paraventricular nucleus, and PR excitatory effects persisted in slices treated with the SOD mimetic tempol. Taken together, these experiments indicated that PR excitatory effects on vasopressin neurons involve inhibition of IA, due, in part, to increases in its voltage-dependent inactivation properties. Moreover, our results indicate that PR effects did not involve an increase in oxidative stress.NEW & NOTEWORTHY Here, we demonstrate that prorenin/the prorenin receptor is an important signaling unit for the regulation of vasopressin firing activity and, thus, systemic hormonal release. We identified A-type K+ channels as key molecular targets mediating prorenin stimulation of vasopressin neuronal activity, thus standing as a potential therapeutic target for neurohumoral activation in cardiovascular disease.


Asunto(s)
Precursores Enzimáticos/farmacología , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Renina/farmacología , Vasopresinas/metabolismo , Potenciales de Acción , Animales , Genotipo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipotálamo/citología , Hipotálamo/metabolismo , Técnicas In Vitro , Activación del Canal Iónico/efectos de los fármacos , Masculino , Neuronas/metabolismo , Neurosecreción , Técnicas de Placa-Clamp , Fenotipo , Canales de Potasio con Entrada de Voltaje/metabolismo , Ratas Transgénicas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Vasopresinas/genética
4.
Sci Rep ; 6: 33314, 2016 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-27650866

RESUMEN

Ion channels play a central role in a host of physiological and pathological processes and are the second largest target for existing drugs. There is an increasing need for reliable tools to detect and visualize particular ion channels, but existing solutions suffer from a number of limitations such as high price, poor specificity, and complicated protocols. As an alternative, we produced recombinant chimeric constructs (FP-Tx) consisting of fluorescent proteins (FP) fused with potassium channel toxins from scorpion venom (Tx). In particular, we used two FP, eGFP and TagRFP, and two Tx, OSK1 and AgTx2, to create eGFP-OSK1 and RFP-AgTx2. We show that these chimeras largely retain the high affinity of natural toxins and display selectivity to particular ion channel subtypes. FP-Tx are displaced by other potassium channel blockers and can be used as an imaging tool in ion channel ligand screening setups. We believe FP-Tx chimeras represent a new efficient molecular tool for neurobiology.


Asunto(s)
Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Venenos de Escorpión/farmacología , Animales , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Escherichia coli , Proteínas Fluorescentes Verdes/farmacología , Concentración 50 Inhibidora , Potenciales de la Membrana/efectos de los fármacos , Oocitos , Canales de Potasio con Entrada de Voltaje/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Xenopus laevis
5.
Hum Exp Toxicol ; 35(5): 501-10, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26187900

RESUMEN

The second-generation selective 5-HT2 receptor antagonists and reuptake inhibitors (SARIs) class antidepressants are known to have fewer cardiovascular side effects than the older ones. However, several case reports showed that trazodone, one of the second-generation SARIs, induces QT prolongation, cardiac arrhythmia, and ventricular tachycardia. Although these clinical cases suggested trazodone-induced cardiotoxicity, the toxicological actions of trazodone on cardiac action potentials (APs) beyond the human ether-a-go-go related gene (hERG) remain unclear. To elucidate the cellular mechanism for the adverse cardiac effects of trazodone, we investigated its effects on cardiac APs and ion channels using whole-cell patch clamp techniques in human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and transiently transfected human embryonic kidney cells (HEK293) with cardiac ion channel complementary DNA. Trazodone dose-dependently decreased the maximum upstroke velocity (Vmax) and prolonged the AP duration, inducing early after depolarizations at 3 and 10 µM that triggered ventricular arrhythmias in hiPSC-CMs. Trazodone also inhibited all of the major ion channels (IKr, IKs, INa, and ICa), with an especially high inhibitory potency on hERG. These data indicate that the prolonged AP duration and decreased Vmax due to trazodone are mainly the result of hERG and sodium ion inhibition, and its inhibitory effects on cardiac ion channels can be exhibited in hiPSC-CMs.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Antidepresivos de Segunda Generación/toxicidad , Canal de Potasio ERG1/antagonistas & inhibidores , Miocitos Cardíacos/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/toxicidad , Trazodona/toxicidad , Cardiotoxicidad , Técnicas de Cultivo de Célula , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1/genética , Células HEK293 , Humanos , Canal de Potasio KCNQ1/antagonistas & inhibidores , Canal de Potasio KCNQ1/genética , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/genética , Transfección
6.
Epilepsy Behav ; 52(Pt B): 308-18, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26074183

RESUMEN

Epilepsy is the most common chronic neurological disease, affecting about 1% of the world's population during their lifetime. Most people with epilepsy can attain a seizure-free life upon treatment with antiepileptic drugs (AEDs). Unfortunately, seizures in up to 30% do not respond to treatment. It is estimated that 90% of people with epilepsy live in developing countries, and most of them receive no drug treatment for the disease. This treatment gap has motivated investigations into the effects of plants that have been used by traditional healers all over the world to treat seizures. Extracts of hundreds of plants have been shown to exhibit anticonvulsant activity in phenotypic screens performed in experimental animals. Some of those extracts appear to exhibit anticonvulsant efficacy similar to that of synthetic AEDs. Dozens of plant-derived chemical compounds have similarly been shown to act as anticonvulsants in various in vivo and in vitro assays. To a significant degree, anticonvulsant effects of plant extracts can be attributed to widely distributed flavonoids, (furano)coumarins, phenylpropanoids, and terpenoids. Flavonoids and coumarins have been shown to interact with the benzodiazepine site of the GABAA receptor and various voltage-gated ion channels, which are targets of synthetic AEDs. Modulation of the activity of ligand-gated and voltage-gated ion channels provides an explanatory basis of the anticonvulsant effects of plant secondary metabolites. Many complex extracts and single plant-derived compounds exhibit antiinflammatory, neuroprotective, and cognition-enhancing activities that may be beneficial in the treatment of epilepsy. Thus, botanicals provide a base for target-oriented antiepileptic drug discovery and development. In the future, preclinical work should focus on the characterization of the effects of plant extracts and plant-derived compounds on well-defined targets rather than on phenotypic screening using in vivo animal models of acute seizures. At the same time, available data provide ample justification for clinical studies with selected standardized botanical extracts and plant-derived compounds. This article is part of a Special Issue entitled "Botanicals for Epilepsy".


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/tratamiento farmacológico , Medicina de Hierbas/métodos , Extractos Vegetales/uso terapéutico , Animales , Anticonvulsivantes/farmacología , Epilepsia/diagnóstico , Epilepsia/epidemiología , Flavonoides/farmacología , Flavonoides/uso terapéutico , Humanos , Fitoterapia/métodos , Extractos Vegetales/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/fisiología , Receptores de GABA-A/fisiología , Convulsiones/diagnóstico , Convulsiones/tratamiento farmacológico , Convulsiones/epidemiología
7.
Cent Nerv Syst Agents Med Chem ; 15(2): 74-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25751086

RESUMEN

Cnidarians are numbered among the most venomous organisms. Their venoms are contained in intracellular capsules, nematocysts, which inject the content into preys/attackers through an eversion system resembling a syringe needle. Several cnidarian venoms have activity against the nervous system, being neurotoxic, or affect other systems whose functioning is under nerve control. Besides direct damage to nerve cells, the activity on ionic conductance, blockade of neuromuscular junctions, and influence on action potentials and on voltage-gated channels have been described. Therefore, cnidarians can be a useful source of nervous system-targeted compounds which could have, in perspective, a role in the therapy of some nervous system diseases. Following this idea, this article aims to review the existing data about the neuroactive properties of cnidarian venoms and their possible usefulness in tackling some neurological diseases as well as neurodegenerative age-related diseases whose incidence is expected to raise in the next decades owing to the increase of life expectancy.


Asunto(s)
Analgésicos/aislamiento & purificación , Venenos de Cnidarios/farmacología , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Fármacos Neuroprotectores/aislamiento & purificación , Neurotoxinas/aislamiento & purificación , Analgésicos/uso terapéutico , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/uso terapéutico , Cnidarios/química , Venenos de Cnidarios/aislamiento & purificación , Venenos de Cnidarios/toxicidad , Evaluación Preclínica de Medicamentos , Humanos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Unión Neuromuscular/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas/toxicidad , Bloqueadores de los Canales de Potasio/aislamiento & purificación , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Bloqueadores del Canal de Sodio Activado por Voltaje/aislamiento & purificación , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología
8.
J Pharmacol Sci ; 123(2): 147-58, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24096832

RESUMEN

To develop a simple screening system for blockers of voltage-gated Kv1.3 and Kv1.5 channels, new cell lines co-expressing mutated Nav1.5 (IFM/Q3), Kir2.1 (Kir), and Kv1.3 or Kv1.5 were introduced as IFM/Q3+Kir+Kv1.3 and IFM/Q3+Kir+Kv1.5, respectively. Electrical stimulation (ES) of a cell line, IFM/Q3+Kir, induced prolonged action potentials due to the slow inactivation of IFM/Q3 and subsequent cell death. Additional co-expression of Kv1.3 or Kv1.5 to IFM/Q3+Kir shortened the evoked action potentials and prevented cell death. In the presence of margatoxin, a selective Kv1.3-blocker, ES induced cell death in IFM/Q3+Kir+Kv1.3, but not in IFM/Q3+Kir+Kv1.5. In the presence of 4-aminopyridine, a non-selective Kv-channel blocker, ES application elicited cell death in both cell lines. The IC50s of acacetin, a Kv1.5-blocker, was 10.2 µM in IFM/Q3+Kir+Kv1.3 and almost identical to that in IFM/Q3+Kir+Kv1.5 (7.6 µM). The IC50s of citalopram, a 5-HT uptake-inhibitor, were 1.8 µM in IFM/Q3+Kir+Kv1.3 and 1.5 µM in IFM/Q3+Kir+Kv1.5, respectively. These IC50s were comparable to those determined electrophysiologically. In conclusion, acacetin and citalopram block both Kv1.3 and Kv1.5 without selectivity. The Kv1.3 or Kv1.5 channel inhibition assay using these new cell lines may be applicable to high-throughput screening because of its simplicity, accuracy, and high cost-performance.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , 4-Aminopiridina/farmacología , Supervivencia Celular/efectos de los fármacos , Citalopram/farmacología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Potenciales Evocados/efectos de los fármacos , Flavonas/farmacología , Células HEK293 , Humanos , Mutación , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Venenos de Escorpión/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
9.
Pharm Biol ; 50(6): 760-5, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22471999

RESUMEN

CONTEXT: Wogonin is a flavone derivative isolated from Scutellaria baicalensis Georgi (Labiatae) root, which is a traditional Chinese drug used as an anti-inflammatory and for management of dysmenorrhea. OBJECTIVE: The effect of wogonin on the uterus has not yet been examined. We investigated the relaxant effects of wogonin on contractile activity of isolated uterine strips of rats. MATERIALS AND METHODS: The effect of wogonin on spontaneous uterine contraction, and uterine contraction induced by agonists, K⁺-depolarization and oxytocin in Ca²âº-free solution was observed. To clarify the type of potassium channel, we tested the effects of 4-aminopyridine, tetraethylammonium and glibenclamide. RESULTS: Wogonin reduced the contractile amplitude of uterine strip smooth muscle of rats in a dose-dependent manner. The concentration of wogonin for reducing the contraction amplitude by 50% (IC50) on spontaneous contractions was 60.5 µM. Wogonin also inhibited the contraction induced by three agonists (oxytocin, prostaglandin F(2α) and acetylcholine). For the uterine strips pretreated with oxytocin in Ca²âº-free solution or K⁺-depolarization, wogonin showed relaxant effect on the induced uterine contractions. In addition, whereas the inhibitive effect of wogonin on the contraction of uterine smooth muscle in rats could be partly blocked by 4-aminopyridine and tetraethylammonium, it was not influenced by glibenclamide. DISCUSSION AND CONCLUSION: Wogonin significantly inhibited the contraction of rat uterine smooth muscle probably through the inhibition of the inflow of extracellular calcium into cells via cell membrane, and intracellular release of calcium ions. In addition, the relaxant effect induced by wogonin might be due in part to the opening of voltage-dependent and large conductance Ca²âº-activated K⁺ channels.


Asunto(s)
Flavanonas/farmacología , Moduladores del Transporte de Membrana/farmacología , Relajación Muscular/efectos de los fármacos , Miometrio/efectos de los fármacos , Fármacos Neuromusculares/farmacología , Raíces de Plantas/química , Scutellaria baicalensis/química , Animales , Antiinflamatorios no Esteroideos , Señalización del Calcio/efectos de los fármacos , China , Estro , Etnofarmacología , Femenino , Flavanonas/aislamiento & purificación , Técnicas In Vitro , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Moduladores del Transporte de Membrana/aislamiento & purificación , Fármacos Neuromusculares/aislamiento & purificación , Oxitócicos/antagonistas & inhibidores , Oxitócicos/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/agonistas , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Ratas , Ratas Wistar
10.
Mol Membr Biol ; 28(2): 79-89, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21231794

RESUMEN

The heterotetrameric K(+)-channel KCNQ1/KCNE1 is expressed in heart, skeletal muscle, liver and several epithelia including the renal proximal tubule. In the heart, it contributes to the repolarization of cardiomyocytes. The repolarization is impaired in ischemia. Ischemia stimulates the AMP-activated protein kinase (AMPK), a serine/threonine kinase, sensing energy depletion and stimulating several cellular mechanisms to enhance energy production and to limit energy utilization. AMPK has previously been shown to downregulate the epithelial Na(+) channel ENaC, an effect mediated by the ubiquitin ligase Nedd4-2. The present study explored whether AMPK regulates KCNQ1/KCNE1. To this end, cRNA encoding KCNQ1/KCNE1 was injected into Xenopus oocytes with and without additional injection of wild type AMPK (AMPKα1 + AMPKß1 + AMPKγ1), of the constitutively active (γR70Q)AMPK (α1ß1γ1(R70Q)), of the kinase dead mutant (αK45R)AMPK (α1(K45R)ß1γ1), or of the ubiquitin ligase Nedd4-2. KCNQ1/KCNE1 activity was determined in two electrode voltage clamp experiments. Moreover, KCNQ1 abundance in the cell membrane was determined by immunostaining and subsequent confocal imaging. As a result, wild type and constitutively active AMPK significantly reduced KCNQ1/KCNE1-mediated currents and reduced KCNQ1 abundance in the cell membrane. Similarly, Nedd4-2 decreased KCNQ1/KCNE1-mediated currents and KCNQ1 protein abundance in the cell membrane. Activation of AMPK in isolated perfused proximal renal tubules by AICAR (10 mM) was followed by significant depolarization. In conclusion, AMPK is a potent regulator of KCNQ1/KCNE1.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Canal de Potasio KCNQ1/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Western Blotting , Membrana Celular/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Activación del Canal Iónico , Canal de Potasio KCNQ1/genética , Canal de Potasio KCNQ1/metabolismo , Túbulos Renales Proximales/metabolismo , Microscopía Confocal , Mutagénesis Sitio-Dirigida , Ubiquitina-Proteína Ligasas Nedd4 , Técnicas de Placa-Clamp , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , ARN Complementario , Ribonucleótidos/farmacología , Xenopus , Proteínas de Xenopus
11.
Biol Pharm Bull ; 33(8): 1360-3, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20686232

RESUMEN

Ligusticum wallichii is an herb widely used to treat vascular disorders in Asian countries, and tetramethylpyrazine (TMP) has been identified as one of its vasorelaxant active components. This study was performed to examine the endothelium-independent relaxation produced by the butanol-soluble fraction of L. wallichii extract (LwBt) and its possible mechanisms of action in isolated rat aortic rings. The effects were compared with those of TMP. LwBt produced vasorelaxation that increased gradually after 2-3 min of LwBt administration and reached a maximum within 30 min. LwBt-induced relaxation was significantly attenuated by pretreatment with 4-aminopyridine and apamin. Additionally, LwBt attenuated CaCl(2)-induced vasoconstriction in high-potassium depolarized medium. Thus, LwBt-induced vasorelaxation apparently involved inhibition of calcium influx, mediated by the opening of voltage-dependent and/or Ca(2+)-activated potassium channels. On the other hand, the effect of TMP was significantly attenuated by pretreatment with glibenclamide, and 4-aminopyridine had no effect. In conclusion, LwBt-induced endothelium-independent vasorelaxation was mediated by the opening of voltage-dependent potassium channels, while TMP-induced relaxation was mediated by the opening of ATP-dependent potassium channels. These effects of LwBt may be due to a substance other than TMP.


Asunto(s)
Aorta Torácica/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Endotelio Vascular/efectos de los fármacos , Ligusticum/química , Pirazinas/farmacología , Vasodilatación/efectos de los fármacos , Animales , Aorta Torácica/metabolismo , Butanoles/química , Medicamentos Herbarios Chinos/aislamiento & purificación , Endotelio Vascular/metabolismo , Técnicas In Vitro , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Pirazinas/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Solubilidad , Factores de Tiempo , Vasoconstrictores/farmacología
12.
Phytomedicine ; 18(1): 46-51, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-20684875

RESUMEN

Lignans are natural phytochemicals which exhibit multiple pharmacological effects such as anti-inflammation, antivirus and anti-tumor activities. Whether they have effects on neural tissues and ion channels is still unknown. The effects of several arylnaphathalene lignans purified from Taiwania cryptomerioides on voltage-gated K(+) (Kv) channels in mouse neuroblastoma N2A cells were examined. These lignans included Taiwanin E, helioxanthin (HXT) and diphyllin. All lignans showed inhibitory effects on Kv channels and HXT was the most potent compound (IC(50)=1.7 µM). The mechanism of HXT block was further investigated. Its action was found to be extracellular but not intracellular. HXT accelerated current decay, caused a left-shift in steady-state inactivation curve but had no effect on voltage-dependence of activation. HXT block was unaffected by intracellular K(+) concentrations. Further, it did not affect ATP-sensitive K(+) channels. Our data therefore suggest that HXT is a potent and specific blocker of Kv channels, possibly with an inhibitory mechanism involving acceleration of slow inactivation.


Asunto(s)
Cupressaceae/química , Lignanos/farmacología , Extractos Vegetales/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Animales , Benzodioxoles , Línea Celular Tumoral , Dioxolanos/aislamiento & purificación , Dioxolanos/farmacología , Lignanos/aislamiento & purificación , Ratones , Neuroblastoma/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo
14.
Bioorg Med Chem Lett ; 19(5): 1477-80, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19185489

RESUMEN

The combination of I(Kr) and I(Ks) blockade could lead to synergistic and safe class III anti-arrhythmic effect with the enhanced efficacy and reduced risk. On the rationale of structural hybridization of azimilide and HMR-1556, a novel series of I(Kr) and I(Ks) dual blockers were designed, synthesized and evaluated in vitro. One compound, 3r (CPUY11018), deserves further evaluation for its potent anti-arrhythmic activity and favorable cardiovascular profile.


Asunto(s)
Antiarrítmicos/síntesis química , Cromanos/síntesis química , Bloqueadores de los Canales de Potasio/síntesis química , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Animales , Antiarrítmicos/farmacología , Cromanos/química , Cromanos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Cobayas , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Hibridación de Ácido Nucleico , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/fisiología , Ratas , Ratas Sprague-Dawley , Sulfonamidas/química , Sulfonamidas/farmacología
15.
Pain ; 139(3): 594-602, 2008 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18694623

RESUMEN

The glial cytokine, interleukin-1beta (IL-1beta), potentiates the excitability of nociceptive trigeminal ganglion (TRG) neurons via membrane depolarization following peripheral inflammation. Perforated patch-clamp technique was used this study to show that the mechanism underlying the excitability of small-diameter TRG neurons following inflammation is due to IL-1beta. Inflammation was induced by injection of complete Freund's adjuvant (CFA) into the whisker pad. The TRG neurons innervating the site of inflammation were identified by fluorogold (FG) labeling. The threshold for escape from mechanical stimulation applied to the orofacial area in inflamed rats was significantly lower than observed for control rats. IL-1beta at 1nM suppressed total voltage-gated K(+) currents in most TRG neurons (70%) under voltage-clamp conditions in control and inflamed rats. IL-1beta significantly decreased the total, transient (I(A)) and sustained (I(K)) currents in FG-labeled small-diameter TRG neurons in both groups. The IL-1beta-induced suppression of TRG neuron excitability was abolished by co-administration of ILra, an IL-1beta receptor blocker. The magnitude of inhibition of I(A) and I(K) currents by IL-1beta was significantly greater in inflamed rats than in controls. IL-1beta inhibited I(A) to a significantly greater extent than I(K). These results suggest that the inhibitory effect of I(A) and I(K) currents by IL-1beta in small-diameter TRG neurons potentiates neuronal excitability thereby contributing to trigeminal inflammatory hyperalgesia. These findings provide evidence for the development of voltage-gated K(+) channel openers and IL-1beta antagonists as therapeutic agents for the treatment of trigeminal inflammatory hyperalgesia.


Asunto(s)
Interleucina-1beta/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Receptores de Interleucina-1/fisiología , Ganglio del Trigémino/efectos de los fármacos , Neuralgia del Trigémino/fisiopatología , Potenciales de Acción/efectos de los fármacos , Animales , Reacción de Fuga/fisiología , Adyuvante de Freund/toxicidad , Inflamación/inducido químicamente , Inflamación/fisiopatología , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Masculino , Umbral del Dolor , Técnicas de Placa-Clamp , Estimulación Física/efectos adversos , Canales de Potasio con Entrada de Voltaje/fisiología , Ratas , Ratas Wistar , Receptores de Interleucina-1/efectos de los fármacos , Proteínas Recombinantes/farmacología , Tacto , Ganglio del Trigémino/fisiopatología , Neuralgia del Trigémino/prevención & control , Vibrisas/inervación
16.
J Clin Invest ; 118(1): 272-80, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18097472

RESUMEN

Leptin is a hormone that reduces excitability in some hypothalamic neurons via leptin receptor activation of the JAK2 and PI3K intracellular signaling pathways. We hypothesized that leptin receptor activation in other neuronal subtypes would have anticonvulsant activity and that intranasal leptin delivery would be an effective route of administration. We tested leptin's anticonvulsant action in 2 rodent seizure models by directly injecting it into the cortex or by administering it intranasally. Focal seizures in rats were induced by neocortical injections of 4-aminopyridine, an inhibitor of voltage-gated K+ channels. These seizures were briefer and less frequent upon coinjection of 4-aminopyridine and leptin. In mice, intranasal administration of leptin produced elevated brain and serum leptin levels and delayed the onset of chemical convulsant pentylenetetrazole-induced generalized convulsive seizures. Leptin also reduced neuronal spiking in an in vitro seizure model. Leptin inhibited alpha-amino-3-hydroxy-5-methyl-4-isoxazole proprionic acid (AMPA) receptor-mediated synaptic transmission in mouse hippocampal slices but failed to inhibit synaptic responses in slices from leptin receptor-deficient db/db mice. JAK2 and PI3K antagonists prevented leptin inhibition of AMPAergic synaptic transmission. We conclude that leptin receptor activation and JAK2/PI3K signaling may be novel targets for anticonvulsant treatments. Intranasal leptin administration may have potential as an acute abortive treatment for convulsive seizures in emergency situations.


Asunto(s)
Hipotálamo/metabolismo , Leptina/farmacología , Receptores AMPA/metabolismo , Convulsiones/tratamiento farmacológico , Transmisión Sináptica/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo , 4-Aminopiridina/toxicidad , Administración Intranasal , Animales , Convulsivantes/toxicidad , Hipotálamo/patología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Leptina/farmacocinética , Leptina/uso terapéutico , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Pentilenotetrazol/toxicidad , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Bloqueadores de los Canales de Potasio/toxicidad , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores AMPA/genética , Receptores de Leptina/agonistas , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Convulsiones/inducido químicamente , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/patología , Transmisión Sináptica/genética
17.
J Clin Invest ; 118(1): 26-8, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18097479

RESUMEN

Secreted from adipose tissue at levels proportional to fat stores, the hormone leptin is a critical regulator of the hypothalamic machinery that controls feeding and energy metabolism. Despite the critical role of leptin in the maintenance of energy homeostasis, no leptin-based therapeutic approaches have emerged to combat metabolic disorders such as obesity or diabetes. In this issue of the JCI, Xu et al. report a robust influence of leptin, beyond its role in metabolism, on hippocampal neuronal processes implicated in the etiology of epileptic seizures, learning, and memory (see the related article beginning on page 272). They show, in two rodent seizure models, that leptin administered directly to the brain or nasal epithelium suppresses seizures via direct effects on glutamate neurotransmission in the hippocampus. These observations suggest that leptin may have therapeutic potential in the treatment of epilepsy and strengthen the notion that peripheral metabolic hormones such as leptin play important roles in the regulation of higher brain functions.


Asunto(s)
Hipotálamo/metabolismo , Leptina/farmacología , Receptores AMPA/metabolismo , Convulsiones/tratamiento farmacológico , Transmisión Sináptica/efectos de los fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo , 4-Aminopiridina/toxicidad , Administración Intranasal , Animales , Convulsivantes/toxicidad , Hipotálamo/patología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Leptina/farmacocinética , Leptina/uso terapéutico , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Pentilenotetrazol/toxicidad , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Bloqueadores de los Canales de Potasio/toxicidad , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores AMPA/genética , Receptores de Leptina/agonistas , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Convulsiones/inducido químicamente , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/patología , Transmisión Sináptica/genética
18.
Biochemistry ; 45(51): 15591-600, 2006 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-17176080

RESUMEN

JZTX-XI is a peptide toxin isolated from the venom of the Chinese spider Chilobrachys jingzhao. It contains 34 residues including six cysteine residues with disulfide bridges linked in the pattern of I-IV, II-V, and III-VI. Using 3'- and 5'-RACE methods, the full-length cDNA was identified as encoding an 86-residue precursor of JZTX-XI. In the electrophysiological assay, JZTX-XI shows activity toward the Kv2.1 channel in a way similar to hanatoxin1 and SGTx1 that both the activation and the deactivation processes are affected, which is in accordance with the high sequence homology among them (over 60% identity). On the other hand, JZTX-XI also exhibits specific interaction against the Nav channels of rat cardiac myocytes with a significant reduction in the peak current and slowing of channel inactivation. The solution structure of native JZTX-XI was determined by 1H NMR methods to identify the structural basis of these specific activities. Structural comparison of JZTX-XI with other gating modifier toxins shows that they all adopt a similar surface profile, a hydrophobic patch surrounded by charged residues such as Arg or Lys, which might be a common structural factor responsible for toxin-channel interaction. JZTX-XI might be an ideal tool to further investigate how spider toxins recognize various ion channels as their targets.


Asunto(s)
Neuropéptidos/química , Neurotoxinas/química , Péptidos/química , Péptidos/fisiología , Canales de Potasio con Entrada de Voltaje/metabolismo , Canales de Sodio/metabolismo , Venenos de Araña/química , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cristalografía por Rayos X , ADN Complementario/aislamiento & purificación , Femenino , Activación del Canal Iónico/genética , Activación del Canal Iónico/fisiología , Masculino , Ratones , Datos de Secuencia Molecular , Neuropéptidos/genética , Neuropéptidos/aislamiento & purificación , Neuropéptidos/fisiología , Neurotoxinas/genética , Neurotoxinas/aislamiento & purificación , Neurotoxinas/toxicidad , Oocitos/química , Oocitos/metabolismo , Péptidos/genética , Péptidos/aislamiento & purificación , Periplaneta , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/metabolismo , Bloqueadores de los Canales de Sodio/toxicidad , Venenos de Araña/genética , Venenos de Araña/aislamiento & purificación , Venenos de Araña/toxicidad , Xenopus laevis
19.
Assay Drug Dev Technol ; 4(4): 443-50, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16945016

RESUMEN

The slow delayed rectifier K+ current, Iks, encoded by KCNQ1 (KvLQT1)/KCNE1 (mink) genes, contributes to cardiac action potential repolarization and determines the heartbeat rate. Mutations in either KCNQ1 or KCNE1 that reduce Iks cause long-QT syndrome (LQTS), a disorder of ventricular repolarization that results in cardiac arrhythmia and sudden death. A well-recognized potential treatment for LQTS caused by reduction of Iks is to enhance functional activation of cardiac KCNQ1/KCNE1 channels. In the present study, we generated a stable Chinese hamster ovary cell line that expresses KCNQ1/KCNE1 channels confirmed by electrophysiology. Using a pharmacological tool compound R-L3 (L-364,373 [(3-R)-1,3-dihydro-5-(2-fluorophenyl)-3-(1H-indol- 3-ylmethyl)-1-methyl-2H-1,4-benzodiazepin-2-one]), which activates KCNQ1/mink channels, we then developed and validated a non-radioactive rubidium (Rb+) efflux assay that directly measures the functional activity of KCNQ1/KCNE1 channels by atomic absorption spectroscopy. Our results show that the validated Rb+ efflux assay can be used for screening of KCNQ1/KCNE1 openers that potentially treat LQTS in both inherited and acquired forms. In addition, the assay also can be used for evaluation of possible long-QT liability during cardiac selectivity of new chemical entities.


Asunto(s)
Benzodiazepinas/farmacología , Canal de Potasio KCNQ1/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Rubidio/metabolismo , Animales , Antracenos/farmacología , Células CHO , Cromanos/farmacología , Cricetinae , Canales Catiónicos Regulados por Nucleótidos Cíclicos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/genética , Canales Iónicos/fisiología , Canal de Potasio KCNQ1/antagonistas & inhibidores , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Miocardio/metabolismo , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/genética , Cloruro de Potasio/farmacología , Ratas , Reproducibilidad de los Resultados , Factores de Riesgo , Espectrofotometría Atómica/métodos , Tetraetilamonio/farmacología
20.
J Cardiovasc Pharmacol ; 47(2): 211-20, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16495758

RESUMEN

Flunarizine has been widely used for the management of a variety of disorders such as peripheral vascular diseases, migraine, and epilepsy. The majority of its beneficial effects have been attributed to its ability to inhibit voltage-gated Ca2+ channels in the low micromolar range, albeit non-selectively, as flunarizine has been shown to inhibit a variety of ion channels. We examined the effects of flunarizine on potassium currents through cardiac channels encoded by the human ether-a-go-go related gene (hERG) stably expressed in CHO cells. In this study, we have characterized the effect of flunarizine on biophysical properties of hERG potassium currents with standard whole-cell voltage-clamp techniques. Notably, flunarizine is a highly potent inhibitor of hERG current with an IC50 value of 5.7 nM. The effect of flunarizine on hERG potassium current is concentration and time dependent, and displays voltage dependence over the voltage range between -40 and 0 mV. At concentrations near or above the IC50, flunarizine causes a negative shift in the voltage dependence of hERG current activation and accelerates tail current deactivation. Flunarizine preferentially blocks the activated state of the channel and displays weak frequency dependence of inhibition. Flunarizine also inhibits KCNQ1/KCNE1 channel current with an IC50 of 0.76 microM.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Potasio Éter-A-Go-Go/efectos de los fármacos , Flunarizina/farmacología , Corazón/fisiología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Animales , Células CHO , Bloqueadores de los Canales de Calcio/química , Técnicas de Cultivo de Célula , Línea Celular , Cricetinae , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Técnicas Electrofisiológicas Cardíacas , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/fisiología , Flunarizina/química , Corazón/efectos de los fármacos , Humanos , Estructura Molecular , Técnicas de Placa-Clamp , Potasio/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA