Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Bioorg Med Chem Lett ; 23(12): 3640-5, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23652221

RESUMEN

A series of benzazepinones were synthesized and evaluated for block of Nav1.7 sodium channels. Compound 30 from this series displayed potent channel block, good selectivity versus other targets, and dose-dependent oral efficacy in a rat model of neuropathic pain.


Asunto(s)
Benzazepinas/farmacología , Neuralgia/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/farmacología , Animales , Modelos Animales de Enfermedad , Ratas
2.
ACS Med Chem Lett ; 4(11): 1064-8, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24900606

RESUMEN

We report the investigation of sulfonamide-derived Cav2.2 inhibitors to address drug-metabolism liabilities with this lead class of analgesics. Modification of the benzamide substituent provided improvements in both potency and selectivity. However, we discovered that formation of the persistent 3-(trifluoromethyl)benzenesulfonamide metabolite was an endemic problem in the sulfonamide series and that the replacement of the center aminopiperidine scaffold failed to prevent this metabolic pathway. This issue was eventually addressed by application of a bioisostere strategy. The new gem-dimethyl sulfone series retained Cav2.2 potency without the liability of the circulating sulfonamide metabolite.

3.
J Med Chem ; 55(22): 9847-55, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23098566

RESUMEN

The voltage-gated calcium channel Ca(v)2.2 (N-type calcium channel) is a critical regulator of synaptic transmission and has emerged as an attractive target for the treatment of chronic pain. We report here the discovery of sulfonamide-derived, state-dependent inhibitors of Ca(v)2.2. In particular, 19 is an inhibitor of Ca(v)2.2 that is selective over cardiac ion channels, with a good preclinical PK and biodistribution profile. This compound exhibits dose-dependent efficacy in preclinical models of inflammatory hyperalgesia and neuropathic allodynia and is devoid of ancillary cardiovascular or CNS pharmacology at the doses tested. Importantly, 19 exhibited no efficacy in Ca(v)2.2 gene-deleted mice. The discovery of metabolite 26 confounds further development of members of this aminopiperidine sulfonamide series. This discovery also suggests specific structural liabilities of this class of compounds that must be addressed.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/química , Canales de Calcio Tipo N/fisiología , Dolor Crónico/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Piperidinas/farmacología , Sulfonamidas/farmacología , Animales , Bloqueadores de los Canales de Calcio/síntesis química , Bloqueadores de los Canales de Calcio/farmacocinética , Canales de Calcio Tipo N/metabolismo , Células Cultivadas , Perros , Humanos , Ratones , Ratones Noqueados , Microsomas Hepáticos/efectos de los fármacos , Técnicas de Placa-Clamp , Piperidinas/síntesis química , Piperidinas/farmacocinética , Ratas , Ratas Sprague-Dawley , Sulfonamidas/síntesis química , Sulfonamidas/farmacocinética , Distribución Tisular
4.
Mol Pharmacol ; 81(3): 488-97, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22188924

RESUMEN

Biological, genetic, and clinical evidence provide validation for N-type calcium channels (Ca(V)2.2) as therapeutic targets for chronic pain. A state-dependent Ca(V)2.2 inhibitor may provide an improved therapeutic window over ziconotide, the peptidyl Ca(V)2.2 inhibitor used clinically. Supporting this notion, we recently reported that in preclinical models, the state-dependent Ca(V)2 inhibitor (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1) has an improved therapeutic window compared with ziconotide. Here we characterize TROX-1 inhibition of Cav2.2 channels in more detail. When channels are biased toward open/inactivated states by depolarizing the membrane potential under voltage-clamp electrophysiology, TROX-1 inhibits Ca(V)2.2 channels with an IC(50) of 0.11 µM. The voltage dependence of Ca(V)2.2 inhibition was examined using automated electrophysiology. TROX-1 IC(50) values were 4.2, 0.90, and 0.36 µM at -110, -90, and -70 mV, respectively. TROX-1 displayed use-dependent inhibition of Ca(V)2.2 with a 10-fold IC(50) separation between first (27 µM) and last (2.7 µM) pulses in a train. In a fluorescence-based calcium influx assay, TROX-1 inhibited Ca(V)2.2 channels with an IC(50) of 9.5 µM under hyperpolarized conditions and 0.69 µM under depolarized conditions. Finally, TROX-1 potency was examined across the Ca(V)2 subfamily. Depolarized IC(50) values were 0.29, 0.19, and 0.28 µM by manual electrophysiology using matched conditions and 1.8, 0.69, and 1.1 µM by calcium influx for Ca(V)2.1, Ca(V)2.2, and Ca(V)2.3, respectively. Together, these in vitro data support the idea that a state-dependent, non-subtype-selective Ca(V)2 channel inhibitor can achieve an improved therapeutic window over the relatively state-independent Ca(V)2.2-selective inhibitor ziconotide in preclinical models of chronic pain.


Asunto(s)
Bloqueadores de los Canales de Calcio/química , Canales de Calcio Tipo N/efectos de los fármacos , Indoles/química , Triazoles/química , Bloqueadores de los Canales de Calcio/farmacología , Línea Celular , Humanos , Indoles/farmacología , Concentración 50 Inhibidora , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Triazoles/farmacología
6.
J Pharmacol Exp Ther ; 334(2): 545-55, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20439438

RESUMEN

Voltage-gated calcium channel (Ca(v))2.2 (N-type calcium channels) are key components in nociceptive transmission pathways. Ziconotide, a state-independent peptide inhibitor of Ca(v)2.2 channels, is efficacious in treating refractory pain but exhibits a narrow therapeutic window and must be administered intrathecally. We have discovered an N-triazole oxindole, (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1), as a small-molecule, state-dependent blocker of Ca(v)2 channels, and we investigated the therapeutic advantages of this compound for analgesia. TROX-1 preferentially inhibited potassium-triggered calcium influx through recombinant Ca(v)2.2 channels under depolarized conditions (IC(50) = 0.27 microM) compared with hyperpolarized conditions (IC(50) > 20 microM). In rat dorsal root ganglion (DRG) neurons, TROX-1 inhibited omega-conotoxin GVIA-sensitive calcium currents (Ca(v)2.2 channel currents), with greater potency under depolarized conditions (IC(50) = 0.4 microM) than under hyperpolarized conditions (IC(50) = 2.6 microM), indicating state-dependent Ca(v)2.2 channel block of native as well as recombinant channels. TROX-1 fully blocked calcium influx mediated by a mixture of Ca(v)2 channels in calcium imaging experiments in rat DRG neurons, indicating additional block of all Ca(v)2 family channels. TROX-1 reversed inflammatory-induced hyperalgesia with maximal effects equivalent to nonsteroidal anti-inflammatory drugs, and it reversed nerve injury-induced allodynia to the same extent as pregabalin and duloxetine. In contrast, no significant reversal of hyperalgesia was observed in Ca(v)2.2 gene-deleted mice. Mild impairment of motor function in the Rotarod test and cardiovascular functions were observed at 20- to 40-fold higher plasma concentrations than required for analgesic activities. TROX-1 demonstrates that an orally available state-dependent Ca(v)2 channel blocker may achieve a therapeutic window suitable for the treatment of chronic pain.


Asunto(s)
Analgésicos/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/fisiología , Indoles/farmacología , Triazoles/farmacología , Analgésicos/efectos adversos , Analgésicos/farmacocinética , Animales , Barorreflejo/efectos de los fármacos , Disponibilidad Biológica , Bloqueadores de los Canales de Calcio/efectos adversos , Bloqueadores de los Canales de Calcio/farmacocinética , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo R/fisiología , Proteínas de Transporte de Catión/fisiología , Línea Celular , Perros , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Hiperalgesia/tratamiento farmacológico , Hipotensión Ortostática/inducido químicamente , Indoles/efectos adversos , Indoles/farmacocinética , Masculino , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/fisiología , Dolor/tratamiento farmacológico , Dolor/etiología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Triazoles/efectos adversos , Triazoles/farmacocinética
7.
Assay Drug Dev Technol ; 6(2): 195-212, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18471074

RESUMEN

Cav2.2 channels play a critical role in pain signaling by controlling synaptic transmission between dorsal root ganglion neurons and dorsal horn neurons. The Cav2.2-selective peptide blocker ziconotide (Prialt, Elan Pharmaceuticals, Dublin, Ireland) has proven efficacious in pain relief, but has a poor therapeutic index and requires intrathecal administration. This has provided impetus for finding an orally active, state-dependent Cav2.2 inhibitor with an improved safety profile. Members of the Cav2 subfamily of calcium channels are the main contributors to central and peripheral synaptic transmission, but the pharmacological effects of blocking each subtype is not yet defined. Here we describe a high-throughput fluorescent assay using a fluorometric imaging plate reader (FLIPR [Molecular Devices, Sunnyvale, CA]) designed to quickly evaluate the state dependence and selectivity of inhibitors across the Cav2 subfamily. Stable cell lines expressing functional Cav2 channels (Ca(V)alpha, beta(3), and alpha(2)delta subunits) were co-transfected with an inward rectifier (Kir2.3) so that membrane potential, and therefore channel state, could be controlled by external potassium concentration. Following cell incubation in drug with varying concentrations of potassium, a high potassium trigger was added to elicit calcium influx through available, unblocked channels. State-dependent inhibitors that preferentially bind to channels in the open or inactivated state can be identified by their increased potency at higher potassium concentrations, where cells are depolarized and channels are biased towards these states. Although the Cav2 channel subtypes differ in their voltage dependence of inactivation, by adjusting pre-trigger potassium concentrations, the degree of steady-state inactivation can be more closely matched across Cav2 subtypes to assess molecular selectivity.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Caveolina 2/efectos de los fármacos , Caveolina 2/fisiología , Evaluación Preclínica de Medicamentos/métodos , Western Blotting , Calcio/metabolismo , Línea Celular , Electrofisiología , Humanos , Inmunohistoquímica , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Potasio/farmacología , Canales de Potasio de Rectificación Interna/efectos de los fármacos , Canales de Potasio de Rectificación Interna/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Bioorg Med Chem Lett ; 18(5): 1696-701, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18243692

RESUMEN

A series of imidazopyridines were evaluated as potential sodium channel blockers for the treatment of neuropathic pain. Several members were identified with good hNa(v)1.7 potency and excellent rat pharmacokinetic profiles. Compound 4 had good efficacy (52% and 41% reversal of allodynia at 2 and 4h post-dose, respectively) in the Chung rat spinal nerve ligation (SNL) model of neuropathic pain when dosed orally at 10mg/kg.


Asunto(s)
Piridinas/química , Piridinas/farmacología , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Analgésicos/química , Analgésicos/farmacología , Animales , Inflamación/tratamiento farmacológico , Estructura Molecular , Canal de Sodio Activado por Voltaje NAV1.7 , Dolor/tratamiento farmacológico , Ratas , Bloqueadores de los Canales de Sodio/farmacocinética , Relación Estructura-Actividad
9.
Bioorg Med Chem Lett ; 18(6): 1963-6, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18289851
11.
Bioorg Med Chem Lett ; 17(16): 4630-4, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17588748
12.
Toxicon ; 49(2): 194-201, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17087985

RESUMEN

The tarantula venom peptides ProTx-I and ProTx-II inhibit voltage-gated sodium channels by shifting their voltage dependence of activation to a more positive potential, thus acting by a mechanism similar to that of potassium channel gating modifiers such as hanatoxin and VSTX1. ProTx-I and ProTx-II inhibit all sodium channel (Nav1) subtypes tested with similar potency and represent the first potent peptidyl inhibitors of TTX-resistant sodium channels. Like gating modifiers of potassium channels, ProTx-I and ProTx-II conform to the inhibitory cystine knot motif, and ProTx-II was demonstrated to bind to sodium channels in the closed state. Both toxins have been synthesized chemically, and ProTx-II, produced by recombinant means, has been used to map the interaction surface of the peptide with the Nav1.5 channel. In comparison, beta-scorpion toxins activate sodium channels by shifting the voltage dependence of activation to more negative potentials, and together these peptides represent valuable tools for exploring the gating mechanism of sodium channels.


Asunto(s)
Activación del Canal Iónico , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/efectos de los fármacos , Venenos de Araña/farmacología , Animales , Humanos , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología
13.
Diabetes ; 55(4): 1034-42, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16567526

RESUMEN

Delayed-rectifier K+ currents (I(DR)) in pancreatic beta-cells are thought to contribute to action potential repolarization and thereby modulate insulin secretion. The voltage-gated K+ channel, K(V)2.1, is expressed in beta-cells, and the biophysical characteristics of heterologously expressed channels are similar to those of I(DR) in rodent beta-cells. A novel peptidyl inhibitor of K(V)2.1/K(V)2.2 channels, guangxitoxin (GxTX)-1 (half-maximal concentration approximately 1 nmol/l), has been purified, characterized, and used to probe the contribution of these channels to beta-cell physiology. In mouse beta-cells, GxTX-1 inhibits 90% of I(DR) and, as for K(V)2.1, shifts the voltage dependence of channel activation to more depolarized potentials, a characteristic of gating-modifier peptides. GxTX-1 broadens the beta-cell action potential, enhances glucose-stimulated intracellular calcium oscillations, and enhances insulin secretion from mouse pancreatic islets in a glucose-dependent manner. These data point to a mechanism for specific enhancement of glucose-dependent insulin secretion by applying blockers of the beta-cell I(DR), which may provide advantages over currently used therapies for the treatment of type 2 diabetes.


Asunto(s)
Canales de Potasio de Tipo Rectificador Tardío/fisiología , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/fisiología , Bloqueadores de los Canales de Potasio/farmacología , Secuencia de Aminoácidos , Animales , Canales de Potasio de Tipo Rectificador Tardío/efectos de los fármacos , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Cinética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Datos de Secuencia Molecular , Péptidos/química , Péptidos/farmacología , Bloqueadores de los Canales de Potasio/química , Venenos de Araña/química , Venenos de Araña/farmacología
14.
Assay Drug Dev Technol ; 2(3): 260-8, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15285907

RESUMEN

The discovery of novel therapeutic agents that act on voltage-gated sodium channels requires the establishment of high-capacity screening assays that can reliably measure the activity of these proteins. Fluorescence resonance energy transfer (FRET) technology using membrane potential-sensitive dyes has been shown to provide a readout of voltage-gated sodium channel activity in stably transfected cell lines. Due to the inherent rapid inactivation of sodium channels, these assays require the presence of a channel activator to prolong channel opening. Because sodium channel activators and test compounds may share related binding sites on the protein, the assay protocol is critical for the proper identification of channel inhibitors. In this study, high throughput, functional assays for the voltage-gated sodium channels, hNa(V)1.5 and hNa(V)1.7, are described. In these assays, channels stably expressed in HEK cells are preincubated with test compound in physiological medium and then exposed to a sodium channel activator that slows channel inactivation. Sodium ion movement through open channels causes membrane depolarization that can be measured with a FRET dye membrane potential-sensing system, providing a large and reproducible signal. Unlike previous assays, the signal obtained in the agonist initiation assay is sensitive to all sodium channel modulators that were tested and can be used in high throughput mode, as well as in support of Medicinal Chemistry efforts for lead optimization.


Asunto(s)
Colorantes/análisis , Transferencia Resonante de Energía de Fluorescencia/métodos , Canales de Sodio/análisis , Canales de Sodio/fisiología , Línea Celular , Colorantes/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Proteínas Musculares/análisis , Proteínas Musculares/fisiología , Canal de Sodio Activado por Voltaje NAV1.5 , Canal de Sodio Activado por Voltaje NAV1.7 , Bloqueadores de los Canales de Sodio/farmacología , Veratridina/farmacología
15.
Biochemistry ; 43(30): 9866-76, 2004 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-15274641

RESUMEN

Sodium channel blockers are used clinically to treat a number of neuropathic pain conditions, but more potent and selective agents should improve on the therapeutic index of currently used drugs. In a high-throughput functional assay, a novel sodium channel (Na(V)) blocker, N-[[2'-(aminosulfonyl)biphenyl-4-yl]methyl]-N'-(2,2'-bithien-5-ylmethyl)succinamide (BPBTS), was discovered. BPBTS is 2 orders of magnitude more potent than anticonvulsant and antiarrhythmic sodium channel blockers currently used to treat neuropathic pain. Resembling block by these agents, block of Na(V)1.2, Na(V)1.5, and Na(V)1.7 by BPBTS was found to be voltage- and use-dependent. BPBTS appeared to bind preferentially to open and inactivated states and caused a dose-dependent hyperpolarizing shift in the steady-state availability curves for all sodium channel subtypes tested. The affinity of BPBTS for the resting and inactivated states of Na(V)1.2 was 1.2 and 0.14 microM, respectively. BPBTS blocked Na(V)1.7 and Na(V)1.2 with similar potency, whereas block of Na(V)1.5 was slightly more potent. The slow tetrodotoxin-resistant Na(+) current in small-diameter DRG neurons was also potently blocked by BPBTS. [(3)H]BPBTS bound with high affinity to a single class of sites present in rat brain synaptosomal membranes (K(d) = 6.1 nM), and in membranes derived from HEK cells stably expressing Na(V)1.5 (K(d) = 0.9 nM). BPBTS dose-dependently attenuated nociceptive behavior in the formalin test, a rat model of tonic pain. On the basis of these findings, BPBTS represents a structurally novel and potent sodium channel blocker that may be used as a template for the development of analgesic agents.


Asunto(s)
Amidas/uso terapéutico , Compuestos de Bifenilo/uso terapéutico , Proteínas Musculares/metabolismo , Dimensión del Dolor/efectos de los fármacos , Bloqueadores de los Canales de Sodio/uso terapéutico , Canales de Sodio/metabolismo , Amidas/síntesis química , Amidas/metabolismo , Analgésicos/síntesis química , Analgésicos/metabolismo , Analgésicos/uso terapéutico , Animales , Sitios de Unión , Compuestos de Bifenilo/síntesis química , Compuestos de Bifenilo/metabolismo , Encéfalo/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Formaldehído/administración & dosificación , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Humanos , Ratones , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Canal de Sodio Activado por Voltaje NAV1.2 , Canal de Sodio Activado por Voltaje NAV1.5 , Canal de Sodio Activado por Voltaje NAV1.7 , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Técnicas de Placa-Clamp , Ratas , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/biosíntesis , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/metabolismo , Canales de Sodio/biosíntesis , Canales de Sodio/genética , Succinatos , Sinaptosomas/metabolismo , Tetrodotoxina/antagonistas & inhibidores , Tetrodotoxina/química
16.
Biochemistry ; 41(50): 14734-47, 2002 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-12475222

RESUMEN

Two peptides, ProTx-I and ProTx-II, from the venom of the tarantula Thrixopelma pruriens, have been isolated and characterized. These peptides were purified on the basis of their ability to reversibly inhibit the tetrodotoxin-resistant Na channel, Na(V) 1.8, and are shown to belong to the inhibitory cystine knot (ICK) family of peptide toxins interacting with voltage-gated ion channels. The family has several hallmarks: cystine bridge connectivity, mechanism of channel inhibition, and promiscuity across channels within and across channel families. The cystine bridge connectivity of ProTx-II is very similar to that of other members of this family, i.e., C(2) to C(16), C(9) to C(21), and C(15) to C(25). These peptides are the first high-affinity ligands for tetrodotoxin-resistant peripheral nerve Na(V) channels, but also inhibit other Na(V) channels (IC(50)'s < 100 nM). ProTx-I and ProTx-II shift the voltage dependence of activation of Na(V) 1.5 to more positive voltages, similar to other gating-modifier ICK family members. ProTx-I also shifts the voltage dependence of activation of Ca(V) 3.1 (alpha(1G), T-type, IC(50) = 50 nM) without affecting the voltage dependence of inactivation. To enable further structural and functional studies, synthetic ProTx-II was made; it adopts the same structure and has the same functional properties as the native peptide. Synthetic ProTx-I was also made and exhibits the same potency as the native peptide. Synthetic ProTx-I, but not ProTx-II, also inhibits K(V) 2.1 channels with 10-fold less potency than its potency on Na(V) channels. These peptides represent novel tools for exploring the gating mechanisms of several Na(V) and Ca(V) channels.


Asunto(s)
Péptidos/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Venenos de Araña/farmacología , Secuencia de Aminoácidos , Animales , Bloqueadores de los Canales de Calcio/química , Bloqueadores de los Canales de Calcio/aislamiento & purificación , Bloqueadores de los Canales de Calcio/farmacología , Línea Celular , Disulfuros/química , Electrofisiología , Humanos , Activación del Canal Iónico/efectos de los fármacos , Datos de Secuencia Molecular , Péptidos/química , Péptidos/aislamiento & purificación , Bloqueadores de los Canales de Potasio/química , Bloqueadores de los Canales de Potasio/aislamiento & purificación , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Wistar , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/aislamiento & purificación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Venenos de Araña/química , Venenos de Araña/aislamiento & purificación
17.
Biochemistry ; 41(20): 6548-60, 2002 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-12009920

RESUMEN

35S-labeled derivatives of the insecticides nodulisporic acid and ivermectin were synthesized and demonstrated to bind with high affinity to a population of receptors in Drosophila head membranes that were previously shown to be associated with a glutamate-gated chloride channel. Nodulisporic acid binding was modeled as binding to a single population of receptors. Ivermectin binding was composed of at least two kinetically distinct receptor populations, only one of which was associated with nodulisporic acid binding. The binding of these two ligands was modulated by glutamate, ivermectin, and antagonists of invertebrate gamma-aminobutyric acid (GABA)ergic receptors. Because solubilized nodulisporic acid and ivermectin receptors comigrated as 230-kDa complexes by gel filtration, antisera specific for both the Drosophila glutamate-gated chloride channel subunit GluCl alpha (DmGluCl alpha) and the GABA-gated chloride channel subunit Rdl (DmRdl) proteins were generated and used to examine the possible coassembly of these two subunits within a single receptor complex. DmGluCl alpha antibodies immunoprecipitated all of the ivermectin and nodulisporic acid receptors solubilized by detergent from Drosophila head membranes. DmRdl antibodies also immunoprecipitated all solubilized nodulisporic receptors, but only approximately 70% of the ivermectin receptors. These data suggest that both DmGluCl alpha and DmRdl are components of nodulisporic acid and ivermectin receptors, and that there also exists a distinct class of ivermectin receptors that contains the DmGluCl alpha subunit but not the DmRdl subunit. This co-association of DmGluCl alpha and DmRdl represents the first biochemical and immunological evidence of coassembly of subunits from two different subclasses of ligand-gated ion channel subunits.


Asunto(s)
Canales de Cloruro/metabolismo , Proteínas de Drosophila/fisiología , Ácido Glutámico/fisiología , Indoles/metabolismo , Ivermectina/metabolismo , Receptores de Droga/metabolismo , Receptores de GABA-A/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Sitios de Unión , Membrana Celular/metabolismo , Proteínas de Drosophila/química , Drosophila melanogaster , Sueros Inmunes/metabolismo , Activación del Canal Iónico , Pruebas de Precipitina , Ensayo de Unión Radioligante , Receptores de Droga/inmunología , Solubilidad , Radioisótopos de Azufre/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...