Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Bioorg Med Chem ; 18(22): 7816-25, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20965738

RESUMEN

Na(v)1.8 (also known as PN3) is a tetrodotoxin-resistant (TTx-r) voltage-gated sodium channel (VGSC) that is highly expressed on small diameter sensory neurons. It has been implicated in the pathophysiology of inflammatory and neuropathic pain, and we envisioned that selective blockade of Na(v)1.8 would be analgesic, while reducing adverse events typically associated with non-selective VGSC blocking therapeutic agents. Herein, we describe the preparation and characterization of a series of 6-aryl-2-pyrazinecarboxamides, which are potent blockers of the human Na(v)1.8 channel and also block TTx-r sodium currents in rat dorsal root ganglia (DRG) neurons. Selected derivatives display selectivity versus human Na(v)1.2. We further demonstrate that an example from this series is orally bioavailable and produces antinociceptive activity in vivo in a rodent model of neuropathic pain following oral administration.


Asunto(s)
Neuralgia/tratamiento farmacológico , Pirazinas/química , Bloqueadores de los Canales de Sodio/química , Canales de Sodio/química , Administración Oral , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Ganglios Espinales/citología , Humanos , Microsomas/metabolismo , Canal de Sodio Activado por Voltaje NAV1.8 , Neuronas/metabolismo , Pirazinas/farmacocinética , Pirazinas/uso terapéutico , Ratas , Bloqueadores de los Canales de Sodio/farmacocinética , Bloqueadores de los Canales de Sodio/uso terapéutico , Canales de Sodio/metabolismo , Relación Estructura-Actividad
2.
Neuropharmacology ; 59(3): 201-7, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20566409

RESUMEN

Activation of sodium channels is essential to action potential generation and propagation. Recent genetic and pharmacological evidence indicates that activation of Na(v)1.8 channels contributes to chronic pain. Herein, we describe the identification of a novel series of structurally related pyridine derivatives as potent Na(v)1.8 channel blockers. A-887826 exemplifies this series and potently (IC(50)=11nM) blocked recombinant human Na(v)1.8 channels. A-887826 was approximately 3 fold less potent to block Na(v)1.2, approximately 10 fold less potent to block tetrodotoxin-sensitive sodium (TTX-S Na(+)) currents and was >30 fold less potent to block Na(V)1.5 channels. A-887826 potently blocked tetrodotoxin-resistant sodium (TTX-R Na(+)) currents (IC(50)=8nM) from small diameter rat dorsal root ganglion (DRG) neurons in a voltage-dependent fashion. A-887826 effectively suppressed evoked action potential firing when DRG neurons were held at depolarized potentials and reversibly suppressed spontaneous firing in small diameter DRG neurons from complete Freund's adjuvant inflamed rats. Following oral administration, A-887826 significantly attenuated tactile allodynia in a rat neuropathic pain model. Further characterization of TTX-R current block in rat DRG neurons demonstrated that A-887826 (100nM) shifted the mid-point of voltage-dependent inactivation of TTX-R currents by approximately 4mV without affecting voltage-dependent activation and did not exhibit frequency-dependent inhibition. The present data demonstrate that A-887826 is a structurally novel and potent Na(v)1.8 blocker that inhibits rat DRG TTX-R currents in a voltage-, but not frequency-dependent fashion. The ability of this structurally novel Na(v)1.8 blocker to effectively reduce tactile allodynia in neuropathic rats further supports the role of Na(v)1.8 sodium channels in pathological pain states.


Asunto(s)
Hiperalgesia/tratamiento farmacológico , Umbral del Dolor/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Bloqueadores de los Canales de Sodio/uso terapéutico , Canales de Sodio/metabolismo , Animales , Biofisica , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica/métodos , Ganglios Espinales/citología , Humanos , Hiperalgesia/etiología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Morfolinas/química , Morfolinas/farmacología , Morfolinas/uso terapéutico , Canal de Sodio Activado por Voltaje NAV1.8 , Neuralgia/complicaciones , Neuralgia/etiología , Niacinamida/análogos & derivados , Niacinamida/química , Niacinamida/farmacología , Niacinamida/uso terapéutico , Técnicas de Placa-Clamp/métodos , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos , Bloqueadores de los Canales de Sodio/química , Canales de Sodio/efectos de los fármacos , Traumatismos de la Médula Espinal/complicaciones , Tetrodotoxina/farmacología , Transfección/métodos
3.
J Pharmacol Exp Ther ; 319(3): 1376-85, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16982702

RESUMEN

ATP-sensitive P2X(7) receptors are localized on cells of immunological origin including glial cells in the central nervous system. Activation of P2X(7) receptors leads to rapid changes in intracellular calcium concentrations, release of the proinflammatory cytokine interleukin-1beta (IL-1beta), and following prolonged agonist exposure, cytolytic plasma membrane pore formation. P2X(7) knockout mice show reduced inflammation as well as decreased nociceptive sensitivity following peripheral nerve injury. A-740003 (N-(1-{[(cyanoimino)(5-quinolinylamino) methyl] amino}-2,2-dimethylpropyl)-2-(3,4-dimethoxyphenyl)acetamide) is a novel competitive antagonist of P2X(7) receptors (IC(50) values = 40 nM for human and 18 nM for rat) as measured by agonist-stimulated changes in intracellular calcium concentrations. A-740003 showed weak or no activity (IC(50) > 10 muM) at other P2 receptors and an array of other neurotransmitter and peptide receptors, ion channels, reuptake sites, and enzymes. A-740003 potently blocked agonist-evoked IL-1beta release (IC(50) = 156 nM) and pore formation (IC(50) = 92 nM) in differentiated human THP-1 cells. Systemic administration of A-740003 produced dose-dependent antinociception in a spinal nerve ligation model (ED(50) = 19 mg/kg i.p.) in the rat. A-740003 also attenuated tactile allodynia in two other models of neuropathic pain, chronic constriction injury of the sciatic nerve and vincristine-induced neuropathy. In addition, A-740003 effectively reduced thermal hyperalgesia observed following intraplantar administration of carrageenan or complete Freund's adjuvant (ED(50) = 38-54 mg/kg i.p.). A-740003 was ineffective in attenuating acute thermal nociception in normal rats and did not alter motor performance at analgesic doses. These data demonstrate that selective blockade of P2X(7) receptors in vivo produces significant antinociception in animal models of neuropathic and inflammatory pain.


Asunto(s)
Acetamidas/farmacología , Analgésicos , Dolor/tratamiento farmacológico , Dolor/etiología , Enfermedades del Sistema Nervioso Periférico/complicaciones , Antagonistas del Receptor Purinérgico P2 , Quinolinas/farmacología , Animales , Antineoplásicos Fitogénicos/toxicidad , Calcio/metabolismo , Línea Celular , Colorantes , Relación Dosis-Respuesta a Droga , Edema/inducido químicamente , Edema/tratamiento farmacológico , Adyuvante de Freund/farmacología , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Inflamación/inducido químicamente , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Interleucina-1beta/metabolismo , Masculino , Actividad Motora/efectos de los fármacos , Nociceptores/efectos de los fármacos , Equilibrio Postural/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2X7 , Neuropatía Ciática/prevención & control , Nervios Espinales/lesiones , Vincristina/toxicidad
4.
Pain ; 123(1-2): 75-82, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16545521

RESUMEN

Antisense (AS) oligodeoxynucleotides (ODNs) targeting the Nav 1.8 sodium channel have been reported to decrease inflammatory hyperalgesia and L5/L6 spinal nerve ligation-induced mechanical allodynia in rats. The present studies were conducted to further characterize Nav 1.8 AS antinociceptive profile in rats to better understand the role of Nav 1.8 in different pain states. Consistent with earlier reports, chronic intrathecal Nav 1.8 AS, but not mismatch (MM), ODN decreased TTX-resistant sodium current density (by 60.5+/-10.2% relative to MM; p<0.05) in neurons from L4 to L5 dorsal root ganglia and significantly attenuated mechanical allodynia following intraplantar complete Freund's adjuvant. In addition, 10 days following chronic constriction injury of the sciatic nerve, Nav 1.8 AS, but not MM, ODN also attenuated mechanical allodynia (54.3+/-8.2% effect, p<0.05 vs. MM) 2 days after initiation of ODN treatment. The anti-allodynic effects remained for the duration of the AS treatment, and CCI rats returned to an allodynic state 4 days after discontinuing AS. In contrast, Nav 1.8 AS ODN failed to reduce mechanical allodynia in the vincristine chemotherapy-induced neuropathic pain model or a skin-incision model of post-operative pain. Finally, Nav 1.8 AS, but not MM, ODN treatment produced a small but significant attenuation of acute noxious mechanical sensitivity in naïve animals (17.6+/-6.2% effect, p<0.05 vs. MM). These data demonstrate a greater involvement of Nav 1.8 in frank nerve injury and inflammatory pain as compared to acute, post-operative or chemotherapy-induced neuropathic pain states.


Asunto(s)
Hiperalgesia/fisiopatología , Inflamación/fisiopatología , Proteínas del Tejido Nervioso/fisiología , Neuralgia/fisiopatología , Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Dolor Postoperatorio/fisiopatología , Canales de Sodio/fisiología , Vincristina/toxicidad , Animales , Conducta Animal , Evaluación Preclínica de Medicamentos , Adyuvante de Freund/toxicidad , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Inyecciones Espinales , Transporte Iónico , Ligadura , Masculino , Canal de Sodio Activado por Voltaje NAV1.8 , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Neuralgia/inducido químicamente , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Oligodesoxirribonucleótidos Antisentido/farmacología , Técnicas de Placa-Clamp , Presión/efectos adversos , Ratas , Ratas Sprague-Dawley , Nervio Ciático/lesiones , Sodio/fisiología , Canales de Sodio/efectos de los fármacos , Canales de Sodio/genética , Nervios Espinales/lesiones , Estrés Mecánico , Tetrodotoxina/farmacología
5.
J Pharmacol Exp Ther ; 314(1): 400-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15837819

RESUMEN

The vanilloid receptor transient receptor potential type V1 (TRPV1) integrates responses to multiple stimuli, such as capsaicin, acid, heat, and endovanilloids and plays an important role in the transmission of inflammatory pain. Here, we report the identification and in vitro characterization of A-425619 [1-isoquinolin-5-yl-3-(4-trifluoromethyl-benzyl)-urea], a novel, potent, and selective TRPV1 antagonist. A-425619 was found to potently block capsaicin-evoked increases in intracellular calcium concentrations in HEK293 cells expressing recombinant human TRPV1 receptors (IC50 = 5 nM). A-425619 showed similar potency (IC50 = 3-4 nM) to block TRPV1 receptor activation by anandamide and N-arachidonoyl-dopamine. Electrophysiological experiments showed that A-425619 also potently blocked the activation of native TRPV1 channels in rat dorsal root ganglion neurons (IC50 = 9 nM). When compared with other known TRPV1 antagonists, A-425619 exhibited superior potency in blocking both naive and phorbol ester-sensitized TRPV1 receptors. Like capsazepine, A-425619 demonstrated competitive antagonism (pA2 = 2.5 nM) of capsaicin-evoked calcium flux. Moreover, A-425619 was 25- to 50-fold more potent than capsazepine in blocking TRPV1 activation. A-425619 showed no significant interaction with a wide range of receptors, enzymes, and ion channels, indicating a high degree of selectivity for TRPV1 receptors. These data show that A-425619 is a structurally novel, potent, and selective TRPV1 antagonist.


Asunto(s)
Calor , Canales Iónicos/antagonistas & inhibidores , Isoquinolinas/farmacología , Urea/análogos & derivados , Ácidos , Animales , Calcio/metabolismo , Células Cultivadas , Evaluación Preclínica de Medicamentos , Electrofisiología , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/efectos de los fármacos , Canales Catiónicos TRPV , Urea/farmacología
6.
Assay Drug Dev Technol ; 1(5): 655-63, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15090238

RESUMEN

Ligand-gated ion channels (LGICs) play important roles in the regulation of cellular function and signaling and serve as excellent drug targets. However, fast desensitization of most LGICs limits the choice of reliable methods to identify agonists, antagonists, and/or modulators in a high throughput manner. In this study, automated Parallel Oocyte Electrophysiology Test station (POETs) was used to screen a directed compound library against a rapidly desensitizing LGIC and to characterize further the pharmacological properties of the hits. POETs allows up to six two-electrode voltage-clamp experiments to be performed in parallel by automatically loading of the oocytes into flowcells, assessing individual oocyte behavior prior to initiating experiments. Oocytes injected with cRNA were transferred from a chilled 96-well plate into flowcells by the instrument, where they were impaled under software control by two independent electrodes. Expression was tested by measuring current responses to rapid application of agonists. Compounds, prepared in a 96-well format, were tested for effects by coapplication with agonist at a single concentration of 30 microM over 2 s. After compound application, oocytes were washed for a minimum of 30 s, and used repeatedly if the test compounds had no significant effect on the control response. Typical throughput could reach approximately 14 plates/day depending on the protocol. Pilot library screening revealed a hit rate of 0.06%, with active compounds having IC(50) values of 4-40 microM. Hits were also confirmed in native neurons using patch-clamp techniques. We conclude that automated POETs serves as a suitable platform for screening and expedient identification of LGIC modulators.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Evaluación Preclínica de Medicamentos/instrumentación , Canales Iónicos/fisiología , Potenciales de la Membrana/fisiología , Oocitos/efectos de los fármacos , Oocitos/fisiología , Técnicas de Placa-Clamp/instrumentación , Robótica/instrumentación , Algoritmos , Animales , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Evaluación Preclínica de Medicamentos/métodos , Electrofisiología/instrumentación , Electrofisiología/métodos , Diseño de Equipo , Análisis de Falla de Equipo , Estudios de Factibilidad , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Canales Iónicos/efectos de los fármacos , Ligandos , Potenciales de la Membrana/efectos de los fármacos , Microelectrodos , Técnicas de Placa-Clamp/métodos , Reproducibilidad de los Resultados , Robótica/métodos , Sensibilidad y Especificidad , Interfaz Usuario-Computador , Xenopus laevis
7.
Proc Natl Acad Sci U S A ; 99(26): 17179-84, 2002 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-12482951

RESUMEN

P2X3 and P2X2/3 receptors are highly localized on peripheral and central processes of sensory afferent nerves, and activation of these channels contributes to the pronociceptive effects of ATP. A-317491 is a novel non-nucleotide antagonist of P2X3 and P2X2/3 receptor activation. A-317491 potently blocked recombinant human and rat P2X3 and P2X2/3 receptor-mediated calcium flux (Ki = 22-92 nM) and was highly selective (IC50 >10 microM) over other P2 receptors and other neurotransmitter receptors, ion channels, and enzymes. A-317491 also blocked native P2X3 and P2X2/3 receptors in rat dorsal root ganglion neurons. Blockade of P2X3 containing channels was stereospecific because the R-enantiomer (A-317344) of A-317491 was significantly less active at P2X3 and P2X2/3 receptors. A-317491 dose-dependently (ED50 = 30 micromolkg s.c.) reduced complete Freund's adjuvant-induced thermal hyperalgesia in the rat. A-317491 was most potent (ED50 = 10-15 micromolkg s.c.) in attenuating both thermal hyperalgesia and mechanical allodynia after chronic nerve constriction injury. The R-enantiomer, A-317344, was inactive in these chronic pain models. Although active in chronic pain models, A-317491 was ineffective (ED50 >100 micromolkg s.c.) in reducing nociception in animal models of acute pain, postoperative pain, and visceral pain. The present data indicate that a potent and selective antagonist of P2X3 and P2X2/3 receptors effectively reduces both nerve injury and chronic inflammatory nociception, but P2X3 and P2X2/3 receptor activation may not be a major mediator of acute, acute inflammatory, or visceral pain.


Asunto(s)
Analgésicos no Narcóticos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Fenoles/farmacología , Compuestos Policíclicos/farmacología , Antagonistas del Receptor Purinérgico P2 , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Relación Dosis-Respuesta a Droga , Hemodinámica/efectos de los fármacos , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2X2 , Receptores Purinérgicos P2X3
8.
Pain ; 99(1-2): 11-9, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12237180

RESUMEN

Extracellular adenosine triphosphate (ATP), acting at P2X ionotropic receptors, is implicated in numerous sensory processes. Exogenous ATP has been shown to be algogenic in both animals and humans. Research focus has been directed towards the P2X(3) receptor, as it is preferentially expressed on nociceptive C-fibers and its implication in pain processing is supported by an altered nociceptive phenotype in P2X(3) knock-out mice. In order to further characterize the role of P2X(3) receptor activation in nociception, we evaluated the effects of continuous intrathecal administration of P2X(3) antisense oligonucleotides for 7 days in the rat. P2X(3) receptor antisense oligonucleotide treatment significantly decreased nociceptive behaviors observed after injection of complete Freund's adjuvant (CFA), formalin or alphabeta-methylene ATP into the rat's hind paw. The anti-hyperalgesic effects of the antisense treatment in the CFA model of inflammatory pain were dose related. Similar effects were observed with two distinct P2X(3) antisense oligonucleotides. These behavioral effects were significantly correlated with a decrease in P2X(3) receptor protein expression in the dorsal root ganglia (DRG). In contrast, a decrease in P2X(3) receptor protein expression in the DRG did not affect nociceptive behavior in the carrageenan model of acute thermal hyperalgesia. P2X(3) receptor antisense oligonucleotide treatment also significantly reduced mechanical allodynia observed after spinal nerve ligation. Overall, the present data demonstrate that activation of P2X(3) receptors contribute to the expression of chronic inflammatory and neuropathic pain states and that relief form these forms of chronic pain might be achieved by selective blockade of P2X(3 )receptor expression or activation.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Neuralgia/terapia , Inflamación Neurogénica/terapia , Oligonucleótidos Antisentido/farmacología , Receptores Purinérgicos P2/genética , Enfermedad Aguda , Adenosina Trifosfato/farmacología , Animales , Antineoplásicos/farmacología , Enfermedad Crónica , Hiperalgesia/terapia , Inyecciones Espinales , Ligadura , Masculino , Nociceptores/efectos de los fármacos , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2X3 , Nervios Espinales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA