Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Bioorg Med Chem Lett ; 20(22): 6812-5, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20855211

RESUMEN

A series of aryl-substituted nicotinamide derivatives with selective inhibitory activity against the Na(v)1.8 sodium channel is reported. Replacement of the furan nucleus and homologation of the anilide linker in subtype-selective blocker A-803467 (1) provided potent, selective derivatives with improved aqueous solubility and oral bioavailability. Representative compounds from this series displayed efficacy in rat models of inflammatory and neuropathic pain.


Asunto(s)
Niacinamida/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Administración Oral , Animales , Disponibilidad Biológica , Niacinamida/química , Niacinamida/farmacocinética , Ratas , Bloqueadores de los Canales de Sodio/administración & dosificación , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacocinética , Relación Estructura-Actividad
2.
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
3.
J Pharmacol Exp Ther ; 324(2): 409-15, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18042830

RESUMEN

Multiple P2 receptor-mediated mechanisms exist by which ATP can alter nociceptive sensitivity following tissue injury. Evidence from a variety of experimental strategies, including genetic disruption studies and the development of selective antagonists, has indicated that the activation of P2X receptor subtypes, including P2X(3), P2X(2/3), P2X(4) and P2X(7), and P2Y (e.g., P2Y(2)) receptors, can modulate pain. For example, administration of a selective P2X(3) antagonist, A-317491, has been shown to effectively block both hyperalgesia and allodynia in different animal models of pathological pain. Intrathecally delivered antisense oligonucleotides targeting P2X(4) receptors decrease tactile allodynia following nerve injury. Selective antagonists for the P2X(7) receptor also reduce sensitization in animal models of inflammatory and neuropathic pain, providing evidence that purinergic glial-neural interactions are important modulators of noxious sensory neurotransmission. Furthermore, activation of P2Y(2) receptors leads to sensitization of polymodal transient receptor potential-1 receptors. Thus, ATP acting at multiple purinergic receptors, either directly on neurons (e.g., P2X(3), P2X(2/3), and P2Y receptors) or indirectly through neural-glial cell interactions (P2X(4) and P2X(7) receptors), alters nociceptive sensitivity. The development of selective antagonists for some of these P2 receptors has greatly aided investigations into the nociceptive role of ATP. This perspective highlights some of the recent advances to identify selective P2 receptor ligands, which has enhanced the investigation of ATP-related modulation of pain sensitivity.


Asunto(s)
Dolor/fisiopatología , Receptores Purinérgicos/fisiología , Adenosina Trifosfato/química , Adenosina Trifosfato/farmacología , Adenosina Trifosfato/uso terapéutico , Animales , Humanos , Dolor/prevención & control , Dimensión del Dolor/métodos , Agonistas Purinérgicos , Antagonistas Purinérgicos , Uridina Trifosfato/química , Uridina Trifosfato/farmacología , Uridina Trifosfato/uso terapéutico
4.
J Med Chem ; 51(3): 407-16, 2008 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-18176998

RESUMEN

Nav1.8 (also known as PN3) is a tetrodotoxin-resistant (TTx-r) voltage-gated sodium channel (VGSC) that is highly expressed on small diameter sensory neurons and has been implicated in the pathophysiology of inflammatory and neuropathic pain. Recent studies using an Nav1.8 antisense oligonucleotide in an animal model of chronic pain indicated that selective blockade of Nav1.8 was analgesic and could provide effective analgesia with a reduction in the adverse events associated with nonselective VGSC blocking therapeutic agents. Herein, we describe the preparation and characterization of a series of 5-substituted 2-furfuramides, which are potent, voltage-dependent blockers (IC50 < 10 nM) of the human Nav1.8 channel. Selected derivatives, such as 7 and 27, also blocked TTx-r sodium currents in rat dorsal root ganglia (DRG) neurons with comparable potency and displayed >100-fold selectivity versus human sodium (Nav1.2, Nav1.5, Nav1.7) and human ether-a-go-go (hERG) channels. Following systemic administration, compounds 7 and 27 dose-dependently reduced neuropathic and inflammatory pain in experimental rodent models.


Asunto(s)
Amidas/síntesis química , Analgésicos/síntesis química , Antiinflamatorios no Esteroideos/síntesis química , Furanos/síntesis química , Bloqueadores de los Canales de Sodio/síntesis química , Canales de Sodio/fisiología , Amidas/química , Amidas/farmacología , Analgésicos/farmacocinética , Analgésicos/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacocinética , Antiinflamatorios no Esteroideos/farmacología , Línea Celular , Cricetinae , Cricetulus , Furanos/química , Furanos/farmacocinética , Furanos/farmacología , Ganglios Espinales/citología , Humanos , Técnicas In Vitro , Masculino , Ratones , Canal de Sodio Activado por Voltaje NAV1.8 , Proteínas del Tejido Nervioso/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Dolor/tratamiento farmacológico , Dolor/etiología , Técnicas de Placa-Clamp , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/antagonistas & inhibidores , Bloqueadores de los Canales de Sodio/farmacocinética , Bloqueadores de los Canales de Sodio/farmacología , Relación Estructura-Actividad
5.
Bioorg Med Chem ; 16(12): 6379-86, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18501613

RESUMEN

The synthesis and pharmacological characterization of a novel furan-based class of voltage-gated sodium channel blockers is reported. Compounds were evaluated for their ability to block the tetrodotoxin-resistant sodium channel Na(v)1.8 (PN3) as well as the Na(v)1.2 and Na(v)1.5 subtypes. Benchmark compounds from this series possessed enhanced potency, oral bioavailability, and robust efficacy in a rodent model of neuropathic pain, together with improved CNS and cardiovascular safety profiles compared to the clinically used sodium channel blockers mexiletine and lamotrigine.


Asunto(s)
Analgésicos no Narcóticos/química , Analgésicos no Narcóticos/farmacología , Furanos/química , Furanos/farmacología , Neuralgia/tratamiento farmacológico , Piperazinas/química , Piperazinas/farmacología , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/efectos de los fármacos , Analgésicos no Narcóticos/síntesis química , Animales , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Furanos/síntesis química , Humanos , Masculino , Ratones , Piperazinas/síntesis química , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/síntesis química , Relación Estructura-Actividad
6.
Brain Res ; 1052(1): 63-70, 2005 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-16005856

RESUMEN

The P2X7 receptor is an ATP-sensitive ligand-gated cation channel, expressed predominantly in cells with immune origin. Recent studies have demonstrated that P2X7 may play an important role in pain signaling. In the present study, the expression of P2X7 receptors in non-neuronal cells and neurons isolated from dorsal root ganglia was characterized using patch clamp, pharmacological and confocal microscopy approaches. In small diameter DRG neurons, 100 microM 2', 3'-O-(4-benzoylbenzoyl)-ATP (BzATP) evoked an inward current, which was inhibited completely by 1 microM A-317491, a potent and selective P2X3 receptor antagonist. In contrast, BzATP evoked concentration-dependent increases in inward currents in non-neuronal DRG cells with an EC50 value of 26 +/- 0.14 microM, which were resistant to the blockade by A-317491. The activity to evoke cationic currents by P2X receptor agonists in non-neuronal cells showed a rank order of BzATP > ATP > alpha,beta-meATP. Pyridoxal-phosphate-6-azophenyl-,2',4'-disulphonic acid (PPADS) and Mg2+ produced concentration-dependent inhibition of BzATP-evoked currents in non-neuronal cells. Confocal microscopy revealed positive immunoreactivity of anti-P2X7 receptor antibodies on non-neuronal cells. No anti-P2X7 immunoreactivity was observed on DRG neurons. Further electrophysiological studies showed that prolonged agonist activation of P2X7 receptors in non-neuronal cells did not lead to cytolytic pore formation. Taken together, the present study demonstrated functional expression of P2X7 receptors in non-neuronal but not in small diameter neurons from rat DRG. Modulation of P2X7 receptors in non-neuronal cells might have impact on peripheral sensory transduction under normal and pathological states.


Asunto(s)
Ganglios Espinales/citología , Potenciales de la Membrana/fisiología , Neuroglía/metabolismo , Receptores Purinérgicos P2/fisiología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Estimulación Eléctrica/métodos , Inmunohistoquímica/métodos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/efectos de la radiación , Microscopía Confocal/métodos , Técnicas de Placa-Clamp/métodos , Fenoles/farmacología , Fosfopiruvato Hidratasa/metabolismo , Compuestos Policíclicos/farmacología , Agonistas del Receptor Purinérgico P2 , Antagonistas del Receptor Purinérgico P2 , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Ratas , Receptores Purinérgicos P2X7 , Sodio/farmacología
7.
Br J Pharmacol ; 143(1): 81-90, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15302680

RESUMEN

1. Openers of ATP-sensitive K(+) channels are of interest in several therapeutic indications including overactive bladder and other lower urinary tract disorders. This study reports on the in vitro and in vivo characterization of a structurally novel naphthylamide N-[2-(2,2,2-trifluoro-1-hydroxy-1-trifluoromethyl-ethyl)-naphthalen-1-yl]-acetamide (A-151892), as an opener of the ATP-sensitive potassium channels. 2. A-151892 was found to be a potent and efficacious potassium channel opener (KCO) as assessed by glibenclamide-sensitive whole-cell current and fluorescence-based membrane potential responses (-log EC(50)=7.63) in guinea-pig bladder smooth muscle cells. 3. Evidence for direct interaction with KCO binding sites was derived from displacement of binding of the 1,4-dihydropyridine opener [(125)I]A-312110. A-151892 displaced [(125)I]A-312110 binding to bladder membranes with a -log Ki value of 7.45, but lacked affinity against over 70 neurotransmitter receptor and ion channel binding sites. 4. In pig bladder strips, A-151892 suppressed phasic, carbachol-evoked and electrical field stimulus-evoked contractility in a glibenclamide-reversible manner with -log IC(50) values of 8.07, 7.33 and 7.02 respectively, comparable to that of the potencies of the prototypical cyanoguanidine KCO, P1075. The potencies to suppress contractions in thoracic aorta (-log IC(50)=7.81) and portal vein (-log IC(50)=7.98) were not substantially different from those observed for suppression of phasic contractility of the bladder smooth muscle. 5. In vivo, A-151892 was found to potently suppress unstable bladder contractions in obstructed models of unstable contractions in both pigs and rats with pED(35%) values of 8.05 and 7.43, respectively. 6. These results demonstrate that naphthylamide analogs exemplified by A-151892 are novel K(ATP) channel openers and may serve as chemotypes to exploit additional analogs with potential for the treatment of overactive bladder and lower urinary tract symptoms.


Asunto(s)
Acetamidas/farmacología , Adenosina Trifosfato/fisiología , Naftalenos/farmacología , Canales de Potasio/agonistas , Animales , Barbitúricos/metabolismo , Unión Competitiva/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Vasos Sanguíneos/efectos de los fármacos , Femenino , Guanidinas/farmacología , Cobayas , Técnicas In Vitro , Radioisótopos de Yodo , Isoxazoles/metabolismo , Potenciales de la Membrana/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Técnicas de Placa-Clamp , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Porcinos , Vejiga Urinaria/efectos de los fármacos
8.
Assay Drug Dev Technol ; 1(5): 647-54, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15090237

RESUMEN

Although techniques such as (86)Rb(+) flux provide a sensitive measure of K(+) channel activity, the relatively short half-life and high-energy emission, together with the quantities of radioactive material generated, hinder the usefulness of flux-based formats in high throughput screening efforts. This study elaborates on the utilization of flame atomic absorption spectrometry (AAS) techniques for a nonradioactive rubidium efflux assay for large conductance Ca(2+)-activated K(+) channels (BK(Ca)) channels. Utilizing human embryonic kidney (HEK293) cells expressing the BK(Ca) alpha subunit, a 96-well cell-based nonradioactive rubidium efflux screen for channel openers and inhibitors was established. Known BK(Ca) channel openers, including NS1608, NS1619, and NS-8, activated rubidium efflux with EC(50) values ranging from 1 to 4 microM in both radioactive and nonradioactive efflux formats. Compounds such as iberiotoxin, paxilline, and charybdotoxin inhibited rubidium efflux responses evoked by the BK(Ca) channel opener NS1608 in both radioactive and nonradioactive efflux formats. The IC(50) values of the inhibitors in AAS format were comparable to those derived from (86)Rb(+) efflux assays. The present studies show that the pharmacological profiles of BK(Ca) channels assessed by AAS compare well with those obtained using the (86)Rb(+) efflux assay, and support the utility of nonradioactive efflux format for higher throughput screening campaigns for novel K(+) channel modulators.


Asunto(s)
Activación del Canal Iónico/fisiología , Riñón/metabolismo , Canales de Potasio Calcio-Activados/fisiología , Rubidio/metabolismo , Espectrofotometría Atómica/métodos , Bencimidazoles/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Activación del Canal Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Riñón/efectos de los fármacos , Riñón/embriología , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Calcio-Activados/efectos de los fármacos , Técnica de Dilución de Radioisótopos , Reproducibilidad de los Resultados , Radioisótopos de Rubidio/metabolismo , Sensibilidad y Especificidad
9.
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
10.
Brain Res ; 1009(1-2): 147-58, 2004 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-15120592

RESUMEN

The profile of tetrodotoxin sensitive (TTX-S) and resistant (TTX-R) Na(+) channels and their contribution to action potentials and firing patterns were studied in isolated small dorsal root ganglion (DRG) neurons after L5/L6 spinal nerve ligation (SNL). Total TTX-R Na(+) currents and Na(v) 1.8 mRNA were reduced in injured L5 DRG neurons 14 days after SNL. In contrast, TTX-R Na(+)currents and Na(v) 1.8 mRNA were upregulated in uninjured L4 DRG neurons after SNL. Voltage-dependent inactivation of TTX-R Na(+) channels in these neurons was shifted to hyperpolarized potentials by 4 mV. Two types of neurons were identified in injured L5 DRG neurons after SNL. Type I neurons (57%) had significantly lower threshold but exhibited normal resting membrane potential (RMP) and action potential amplitude. Type II neurons (43%) had significantly smaller action potential amplitude but retained similar RMP and threshold to those from sham rats. None of the injured neurons could generate repetitive firing. In the presence of TTX, only 26% of injured neurons could generate action potentials that had smaller amplitude, higher threshold, and higher rheobase compared with sham rats. In contrast, action potentials and firing patterns in uninjured L4 DRG neurons after SNL, in the presence or absence of TTX, were not affected. These results suggest that TTX-R Na(+) channels play important roles in regulating action potentials and firing patterns in small DRG neurons and that downregulation in injured neurons and upregulation in uninjured neurons confer differential roles in shaping electrogenesis, and perhaps pain transmission, in these neurons.


Asunto(s)
Potenciales de Acción/fisiología , Ganglios Espinales/citología , Neuronas Aferentes/fisiología , Nervios Espinales/lesiones , Nervios Espinales/fisiopatología , Amidinas , Anestésicos Locales/farmacología , Animales , Células Cultivadas , Estimulación Eléctrica , Ganglios Espinales/lesiones , Regulación de la Expresión Génica , Activación del Canal Iónico , Ligadura/métodos , Masculino , Potenciales de la Membrana/fisiología , Canal de Sodio Activado por Voltaje NAV1.8 , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas Aferentes/clasificación , Neuronas Aferentes/efectos de los fármacos , Dimensión del Dolor , Técnicas de Placa-Clamp/métodos , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Nervio Ciático/fisiología , Canales de Sodio/genética , Canales de Sodio/metabolismo , Tetrodotoxina/farmacología
11.
Eur J Pharmacol ; 471(3): 157-64, 2003 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-12826234

RESUMEN

The effects of 1-ethyl-2-benzimidazolinone (1-EBIO) and riluzole on human prostate cancer cells, LNCaP and PC-3, were evaluated using rubidium (86Rb(+)) efflux and proliferation assays. 1-EBIO and riluzole evoked concentration-dependent increases in 86Rb(+) efflux from LNCaP and PC-3 cells that were sensitive to inhibition by intermediate-conductance Ca(2+)-activated K(+) channel (IK(Ca)) blockers clotrimazole and charybdotoxin. Blockers of large-conductance Ca(2+)-activated K(+) (BK(Ca)) channel, iberiotoxin, or small-conductance Ca(2+)-activated K(+) (SK(Ca)) channel, apamin or scyllatoxin, had no effect. Concurrently, both 1-EBIO and riluzole evoked concentration-dependent increases in proliferation from human prostate cancer cell lines (LNCaP and PC-3 cells). Clotrimazole and charybdotoxin, but not iberiotoxin, apamin or scyllatoxin, inhibited 1-EBIO- and riluzole-evoked increases in proliferation from LNCaP and PC-3 cells. N-(3-(trifluoromethyl)phenyl)-N'-(2-hydroxy-5-chlorophenyl)urea (NS-1608) and 2-amino-5-(2-fluorophenyl)-4-methyl-1H-pyrrole-3-carbonitrile (NS-8), BK(Ca) channel openers had no effect on LNCaP and PC-3 proliferation. These results demonstrate that IK(Ca) channels play an important role in the regulation of human prostate cancer cell proliferation.


Asunto(s)
Agonistas de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Potasio Calcio-Activados/agonistas , Canales de Potasio Calcio-Activados/antagonistas & inhibidores , Neoplasias de la Próstata/tratamiento farmacológico , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Agonistas de los Canales de Calcio/uso terapéutico , Bloqueadores de los Canales de Calcio/uso terapéutico , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular Tumoral/citología , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Canales de Potasio Calcio-Activados/fisiología , Neoplasias de la Próstata/patología , Riluzol/farmacología , Riluzol/uso terapéutico
12.
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
14.
Proc Natl Acad Sci U S A ; 104(20): 8520-5, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17483457

RESUMEN

Activation of tetrodotoxin-resistant sodium channels contributes to action potential electrogenesis in neurons. Antisense oligonucleotide studies directed against Na(v)1.8 have shown that this channel contributes to experimental inflammatory and neuropathic pain. We report here the discovery of A-803467, a sodium channel blocker that potently blocks tetrodotoxin-resistant currents (IC(50) = 140 nM) and the generation of spontaneous and electrically evoked action potentials in vitro in rat dorsal root ganglion neurons. In recombinant cell lines, A-803467 potently blocked human Na(v)1.8 (IC(50) = 8 nM) and was >100-fold selective vs. human Na(v)1.2, Na(v)1.3, Na(v)1.5, and Na(v)1.7 (IC(50) values >or=1 microM). A-803467 (20 mg/kg, i.v.) blocked mechanically evoked firing of wide dynamic range neurons in the rat spinal dorsal horn. A-803467 also dose-dependently reduced mechanical allodynia in a variety of rat pain models including: spinal nerve ligation (ED(50) = 47 mg/kg, i.p.), sciatic nerve injury (ED(50) = 85 mg/kg, i.p.), capsaicin-induced secondary mechanical allodynia (ED(50) approximately 100 mg/kg, i.p.), and thermal hyperalgesia after intraplantar complete Freund's adjuvant injection (ED(50) = 41 mg/kg, i.p.). A-803467 was inactive against formalin-induced nociception and acute thermal and postoperative pain. These data demonstrate that acute and selective pharmacological blockade of Na(v)1.8 sodium channels in vivo produces significant antinociception in animal models of neuropathic and inflammatory pain.


Asunto(s)
Compuestos de Anilina/farmacología , Compuestos de Anilina/farmacocinética , Furanos/farmacología , Furanos/farmacocinética , Mononeuropatías/terapia , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Manejo del Dolor , Dolor/patología , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Potenciales de Acción/efectos de los fármacos , Analgésicos/farmacología , Compuestos de Anilina/administración & dosificación , Compuestos de Anilina/química , Animales , Capsaicina/farmacología , Potenciales Evocados/efectos de los fármacos , Furanos/administración & dosificación , Furanos/química , Ganglios Espinales/citología , Ganglios Espinales/efectos de los fármacos , Humanos , Inflamación , Cinética , Masculino , Canal de Sodio Activado por Voltaje NAV1.8 , Neuronas/citología , Neuronas/efectos de los fármacos , Dolor/inducido químicamente , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Bloqueadores de los Canales de Sodio/administración & dosificación , Bloqueadores de los Canales de Sodio/química , Bloqueadores de los Canales de Sodio/farmacocinética
15.
Expert Opin Ther Pat ; 16(8): 1113-27, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20144056

RESUMEN

P2X receptors belong to a superfamily of ligand-gated ion channels that conduct the influx of Ca(2+), Na(+) and K(+) cations following activation by extracellular nucleotides such as ATP. Molecular cloning studies have identified seven subunits, namely P2X(1-7), that share approximately 40 - 50% identity in amino acid sequences within the subfamily. Using gene-silencing, pharmacological and electrophysiological approaches, recent studies have revealed roles for P2X(2), P2X(3), P2X(4) and P2X(7) receptors in nociceptive signalling. Homomeric P2X(3) and heteromeric P2X(2/3) receptors are highly localised in the peripheral sensory afferent neurons that conduct nociceptive sensory information to the spinal chord and brain. The discovery of A-317491, a selective and potent non-nucleotide P2X(3) antagonist, provided a pharmacological tool to determine the site and mode of action of P2X(3)-containing receptors in different pain behaviours, including neuropathic, inflammatory and visceral pain. Other P2X receptors (P2X(4) and P2X(7)) that are predominantly expressed in microglia, macrophages and cells of immune origin can trigger the release of cytokines, such as IL-1-beta and TNF-alpha. Genetic disruption of P2X(4) and P2X(7) signalling has been demonstrated to reduce inflammatory and neuropathic pain, suggesting that these two receptors might serve as integrators of neuroinflammation and pain. This article provides an overview of recent scientific literature and patents focusing on P2X(3), P2X(4) and P2X(7) receptors, and the identification of small molecule ligands for the potential treatment of neuropathic and inflammatory pain.

16.
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
17.
J Cardiovasc Pharmacol ; 46(2): 232-40, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16044036

RESUMEN

The pharmacological profile of the novel dihydropyridine K channel opener (KCO), (9R)-9-(3-iodo-4-methylphenyl)-5,9-dihydro-3H-furo[3,4-b]pyrano[4,3-e]pyridine-1,8(4H,7H)-dione (A-325100), is described in numerous in vitro assays. Furthermore, the cardiovascular effects of A-325100 are characterized in both the anesthetized and conscious dog. In vitro, A-325100 selectively activated KATP currents and potently relaxed vascular smooth muscle (IC50 between 7.69x10 M and 7.78x10 M), an effect that was abolished by glyburide. Moreover, A-325100 did not interact with L-type Ca2+ channels at concentrations up to 30 microM. In anesthetized dogs A-325100 produced a dose-dependent reduction in systemic vascular resistance and mean arterial pressure concomitant with dose-dependent increases in dP/dtmax and heart rate. In conscious telemetry-instrumented dogs oral administration of A-325100 produced a similar response profile, including dose-dependent reductions in MAP and increases in heart rate and dP/dtmax. When concentration-dependent changes in MAP, heart rate, and dP/dtmax were compared relative to circulating plasma concentrations, A-325100 produced similar effects in both the anesthetized and conscious dog. In conclusion, the present study provides the first pharmacological description of the novel and selective tricyclic dihydropyridine KCO, A-325100. When studied in vivo, A-325100 produced similar concentration-dependent cardiovascular effects in both models consistent with its mode of action and independent of route of administration. Thus, these data demonstrate that the hemodynamic effects of vasoactive compounds, such as KCOs, can be effectively profiled in both the conscious and anesthetized dog.


Asunto(s)
Dihidropiridinas/farmacología , Hemodinámica/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Canales de Potasio/metabolismo , Pironas/farmacología , Adenosina Trifosfato/metabolismo , Anestesia , Animales , Aorta Torácica/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Dihidropiridinas/sangre , Perros , Relación Dosis-Respuesta a Droga , Cobayas , Frecuencia Cardíaca/efectos de los fármacos , Técnicas In Vitro , Ligandos , Masculino , Relajación Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Técnicas de Placa-Clamp , Vena Porta/efectos de los fármacos , Pironas/sangre , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/fisiología
19.
Expert Opin Ther Targets ; 8(5): 437-58, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15469394

RESUMEN

Potassium channels have re-emerged as attractive targets for overactive bladder and other urological diseases in recent years, in part due to an enhanced understanding of their molecular heterogeneity, tissue distribution, functional roles and regulation in physiological and pathological states. Cloning and heterologous expression analysis, coupled with the advancement of improved high-throughput screening techniques, have enabled expeditious identification of selective small-molecule openers and blockers for ATP-sensitive K+ channels, Ca2+-activated K+ channels and voltage-dependent K+ channel-KQT-like subfamily (KCNQ) members, and has paved the way in the assessment of efficacy and adverse effects in preclinical models. This review focuses on the rationale for molecular targeting of K+ channels, the current status of target validation, including preclinical proof-of-concept studies, and provides perspectives on the limitations and hurdles to be overcome in realising the potential of these targets for diverse urological indications such as overactive bladder, erectile dysfunction and prostate diseases.


Asunto(s)
Canales de Potasio/efectos de los fármacos , Enfermedades de la Vejiga Urinaria/tratamiento farmacológico , Anciano , Animales , Calcio/metabolismo , Método Doble Ciego , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Disfunción Eréctil/tratamiento farmacológico , Disfunción Eréctil/fisiopatología , Femenino , Haplorrinos , Humanos , Transporte Iónico/efectos de los fármacos , Masculino , Persona de Mediana Edad , Estructura Molecular , Erección Peniana/fisiología , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio/clasificación , Canales de Potasio/fisiología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/fisiopatología , Ensayos Clínicos Controlados Aleatorios como Asunto , Ratas , Estimulación Química , Porcinos , Enfermedades de la Vejiga Urinaria/fisiopatología , Vejiga Urinaria Neurogénica/tratamiento farmacológico , Vejiga Urinaria Neurogénica/fisiopatología , Micción/fisiología , Trastornos Urinarios/tratamiento farmacológico , Trastornos Urinarios/fisiopatología
20.
Curr Protoc Pharmacol ; Chapter 11: Unit11.6, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21956803

RESUMEN

This unit describes general methodologies for the characterization of ATP-sensitive K+ channels and the study of ligand-channel interactions in native tissues and clonal cell lines by electrophysiological techniques. Detailed protocols on how to establish patch-clamp single-channel and whole-cell current recording are presented. Two-electrode voltage clamp techniques for studying ATP-sensitive K+ channels expressed in Xenopus oocytes are also included.


Asunto(s)
Electrofisiología/métodos , Canales KATP/fisiología , Animales , Femenino , Humanos , Canales KATP/efectos de los fármacos , Oocitos , Técnicas de Placa-Clamp , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA