Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 372(1): 107-118, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31732698

RESUMO

Cystic fibrosis (CF) is the most common monogenic autosomal recessive disease in Caucasians caused by pathogenic mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene (CFTR). Significant small molecule therapeutic advances over the past two decades have been made to target the defective CFTR protein and enhance its function. To address the most prevalent defect of the defective CFTR protein (i.e., F508del mutation) in CF, two biomolecular activities are required, namely, correctors to increase the amount of properly folded F508delCFTR levels at the cell surface and potentiators to allow the effective opening, i.e., function of the F508delCFTR channel. Combined, these activities enhance chloride ion transport yielding improved hydration of the lung surface and subsequent restoration of mucociliary clearance. To enhance clinical benefits to CF patients, a complementary triple combination therapy consisting of two corrector molecules, type 1 (C1) and type 2, with additive mechanisms along with a potentiator are being investigated in the clinic for maximum restoration of mutated CFTR function. We report the identification and in vitro biologic characterization of ABBV-2222/GLPG2222 (4-[(2R,4R)-4-({[1-(2,2-difluoro-1,3-benzodioxol-5-yl)cyclopropyl]carbonyl}amino)-7-(difluoromethoxy)-3,4-dihydro-2H-chromen-2-yl]benzoic acid),-a novel, potent, and orally bioavailable C1 corrector developed by AbbVie-Galapagos and currently in clinical trials-which exhibits substantial improvements over the existing C1 correctors. This includes improvements in potency and drug-drug interaction (DDI) compared with 3-(6-(1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl)cyclopropanecarboxamido)-3-methylpyridin-2-yl)benzoic acid (VX-809, Lumacaftor) and improvements in potency and efficacy compared with 1-(2,2-difluoro-1,3-benzodioxol-5-yl)-N-[1-[(2R)-2,3-dihydroxypropyl]-6-fluoro-2-(1-hydroxy-2-methylpropan-2-yl)indol-5-yl]cyclopropane-1-carboxamide (VX-661, Tezacaftor). ABBV-2222/GLPG2222 exhibits potent in vitro functional activity in primary patient cells harboring F508del/F508del CFTR with an EC50 value <10 nM. SIGNIFICANCE STATEMENT: To address the most prevalent defect of the defective CFTR protein (i.e., F508del mutation) in cystic fibrosis, AbbVie-Galapagos has developed ABBV-2222/GLPG2222, a novel, potent, and orally bioavailable C1 corrector of this protein. ABBV-2222/GLPG2222, which is currently in clinical trials, exhibits potent in vitro functional activity in primary patient cells harboring F508del/F508del CFTR and substantial improvements over the existing C1 correctors.


Assuntos
Benzoatos/farmacologia , Benzopiranos/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Dobramento de Proteína/efeitos dos fármacos , Animais , Sítios de Ligação , Membrana Celular/metabolismo , Células Cultivadas , Cloretos/metabolismo , Cricetinae , Regulador de Condutância Transmembrana em Fibrose Cística/química , Células HEK293 , Humanos , Moduladores de Transporte de Membrana/farmacologia , Ligação Proteica , Transporte Proteico/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo
2.
J Med Chem ; 59(10): 4926-47, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-27077528

RESUMO

Transient receptor potential vanilloid 3 (TRPV3) is a Ca(2+)- and Na(+)-permeable channel with a unique expression pattern. TRPV3 is found in both neuronal and non-neuronal tissues, including dorsal root ganglia, spinal cord, and keratinocytes. Recent studies suggest that TRPV3 may play a role in inflammation, pain sensation, and skin disorders. TRPV3 studies have been challenging, in part due to a lack of research tools such as selective antagonists. Herein, we provide the first detailed report on the development of potent and selective TRPV3 antagonists featuring a pyridinyl methanol moiety. Systematic optimization of pharmacological, physicochemical, and ADME properties of original lead 5a resulted in identification of a novel and selective TRPV3 antagonist 74a, which demonstrated a favorable preclinical profile in two different models of neuropathic pain as well as in a reserpine model of central pain.


Assuntos
Ciclobutanos/síntese química , Ciclobutanos/farmacologia , Piridinas/síntese química , Piridinas/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Cálcio/metabolismo , Ciclobutanos/química , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Conformação Molecular , Piridinas/química , Relação Estrutura-Atividade , Canais de Cátion TRPV/metabolismo
3.
J Med Chem ; 57(17): 7412-24, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25100568

RESUMO

The synthesis and characterization of a series of selective, orally bioavailable 1-(chroman-4-yl)urea TRPV1 antagonists is described. Whereas first-generation antagonists that inhibit all modes of TRPV1 activation can elicit hyperthermia, the compounds disclosed herein do not elevate core body temperature in preclinical models and only partially block acid activation of TRPV1. Advancing the SAR of this series led to the eventual identification of (R)-1-(7-chloro-2,2-bis(fluoromethyl)chroman-4-yl)-3-(3-methylisoquinolin-5-yl)urea (A-1165442, 52), an analogue that possesses excellent pharmacological selectivity, has a favorable pharmacokinetic profile, and demonstrates good efficacy against osteoarthritis pain in rodents.


Assuntos
Analgésicos/química , Temperatura Corporal/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Ureia/química , Analgésicos/farmacocinética , Analgésicos/farmacologia , Animais , Área Sob a Curva , Temperatura Corporal/fisiologia , Cães , Relação Dose-Resposta a Droga , Descoberta de Drogas , Células HEK293 , Humanos , Isoquinolinas/química , Isoquinolinas/farmacocinética , Isoquinolinas/farmacologia , Taxa de Depuração Metabólica , Modelos Químicos , Estrutura Molecular , Ratos , Relação Estrutura-Atividade , Canais de Cátion TRPV/química , Canais de Cátion TRPV/metabolismo , Ureia/análogos & derivados , Ureia/farmacocinética , Ureia/farmacologia
4.
Assay Drug Dev Technol ; 11(1): 17-24, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23002961

RESUMO

The KCNQ2/3 channel has emerged as a drug target for a number of neurological disorders including pain and epilepsy. Known KCNQ2/3 openers have effects on two distinct biophysical properties of the channel: (1) a hyperpolarizing shift in the voltage dependence of channel activation (V(1/2)), and (2) an increase in channel open probability or peak whole-cell current. The current high-throughput screening assays for KCNQ2/3 openers measure changes of channel activity at sub-peak conductances and the output measure is a combination of effects on V(1/2) shift and peak current. Here, we describe a medium-throughput electrophysiological assay for screening KCNQ2/3 openers using the QPatch HT platform. We employed a double-pulse protocol that measures the shift in V(1/2) and the change in current amplitude at peak conductance voltage. Retigabine along with novel KCNQ2/3 openers were evaluated in this assay. Three classes of KCNQ2/3 openers were identified based on the hyperpolarizing shift in V(1/2) and the change in peak current. All three classes of compounds caused a hyperpolarizing shift in V(1/2), but they were differentiated by their respective effects on peak current amplitude (increase, decrease, or only modestly affecting peak current amplitude). KCNQ2/3 blockers were also identified with this assay. These compounds blocked currents without affecting voltage-dependent activation. In summary, we have developed a medium-throughput assay that can reliably detect changes in the biophysical properties of the KCNQ2/3 channel, V(1/2), and peak current amplitude, and therefore may serve as a reliable assay to evaluate KCNQ2/3 openers and blockers.


Assuntos
Ensaios de Triagem em Larga Escala/normas , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Moduladores de Transporte de Membrana/metabolismo , Técnicas de Patch-Clamp/normas , Carbamatos/química , Carbamatos/metabolismo , Fenômenos Eletrofisiológicos/fisiologia , Células HEK293 , Ensaios de Triagem em Larga Escala/métodos , Humanos , Canal de Potássio KCNQ2/classificação , Canal de Potássio KCNQ3/classificação , Moduladores de Transporte de Membrana/química , Técnicas de Patch-Clamp/métodos , Fenilenodiaminas/química , Fenilenodiaminas/metabolismo
5.
J Pharmacol Exp Ther ; 342(2): 416-28, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22570364

RESUMO

The transient receptor potential vanilloid-1 (TRPV1) channel is involved in the development and maintenance of pain and participates in the regulation of temperature. The channel is activated by diverse agents, including capsaicin, noxious heat (≥ 43°C), acidic pH (< 6), and endogenous lipids including N-arachidonoyl dopamine (NADA). Antagonists that block all modes of TRPV1 activation elicit hyperthermia. To identify efficacious TRPV1 antagonists that do not affect temperature antagonists representing multiple TRPV1 pharmacophores were evaluated at recombinant rat and human TRPV1 channels with Ca(2+) flux assays, and two classes of antagonists were identified based on their differential ability to inhibit acid activation. Although both classes of antagonists completely blocked capsaicin- and NADA-induced activation of TRPV1, select compounds only partially inhibited activation of the channel by protons. Electrophysiology and calcitonin gene-related peptide release studies confirmed the differential pharmacology of these antagonists at native TRPV1 channels in the rat. Comparison of the in vitro pharmacological properties of these TRPV1 antagonists with their in vivo effects on core body temperature confirms and expands earlier observations that acid-sparing TRPV1 antagonists do not significantly increase core body temperature. Although both classes of compounds elicit equivalent analgesia in a rat model of knee joint pain, the acid-sparing antagonist tested is not effective in a mouse model of bone cancer pain.


Assuntos
Temperatura Corporal/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Analgésicos/farmacologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Cálcio/metabolismo , Capsaicina/farmacologia , Linhagem Celular Transformada , Febre/tratamento farmacológico , Febre/fisiopatologia , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Dor/tratamento farmacológico , Dor/metabolismo , Dor/fisiopatologia , Prótons , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Canais de Cátion TRPV/metabolismo
6.
J Gen Physiol ; 138(3): 341-52, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21844219

RESUMO

Transient receptor potential vanilloid 1 (TRPV1) is a ligand-gated nonselective cation channel expressed predominantly in peripheral nociceptors. By detecting and integrating diverse noxious thermal and chemical stimuli, and as a result of its sensitization by inflammatory mediators, the TRPV1 receptor plays a key role in inflammation-induced pain. Activation of TRPV1 leads to a cascade of pro-nociceptive mechanisms, many of which still remain to be identified. Here, we report a novel effect of TRPV1 on the activity of the potassium channel KCNQ2/3, a negative regulator of neuronal excitability. Using ion influx assays, we revealed that TRPV1 activation can abolish KCNQ2/3 activity, but not vice versa, in human embryonic kidney (HEK)293 cells. Electrophysiological studies showed that coexpression of TRPV1 caused a 7.5-mV depolarizing shift in the voltage dependence of KCNQ2/3 activation compared with control expressing KCNQ2/3 alone. Furthermore, activation of TRPV1 by capsaicin led to a 54% reduction of KCNQ2/3-mediated current amplitude and attenuation of KCNQ2/3 activation. The inhibitory effect of TRPV1 appears to depend on Ca(2+) influx through the activated channel followed by Ca(2+)-sensitive depletion of phosphatidylinositol 4,5-bisphosphate and activation of protein phosphatase calcineurin. We also identified physical interactions between TRPV1 and KCNQ2/3 coexpressed in HEK293 cells and in rat dorsal root ganglia neurons. Mutation studies established that this interaction is mediated predominantly by the membrane-spanning regions of the respective proteins and correlates with the shift of KCNQ2/3 activation. Collectively, these data reveal that TRPV1 activation may deprive neurons from inhibitory control mediated by KCNQ2/3. Such neurons may thus have a lower threshold for activation, which may indirectly facilitate TRPV1 in integrating multiple noxious signals and/or in the establishment or maintenance of chronic pain.


Assuntos
Canal de Potássio KCNQ2/antagonistas & inibidores , Canal de Potássio KCNQ3/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Animais , Células Cultivadas , Células HEK293 , Humanos , Canal de Potássio KCNQ2/metabolismo , Canal de Potássio KCNQ3/metabolismo , Masculino , Neurônios/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Ratos , Ratos Sprague-Dawley , Transfecção
7.
Inflamm Res ; 60(7): 683-93, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21394563

RESUMO

OBJECTIVE: The aim of this study was to compare a diverse set of peptide and small-molecule calcium channel blockers for inactivated-state block of native and recombinant N-type calcium channels using fluorescence-based and automated patch-clamp electrophysiology assays. METHODS: The pharmacology of calcium channel blockers was determined at N-type channels in IMR-32 cells and in HEK cells overexpressing the inward rectifying K(+) channel Kir2.1. N-type channels were opened by increasing extracellular KCl. In the Kir2.1/N-type cell line the membrane potential could be modulated by adjusting the extracellular KCl, allowing determination of resting and inactivated-state block of N-type calcium channels. The potency and degree of state-dependent inhibition of these blockers were also determined by automated patch-clamp electrophysiology. RESULTS: N-type-mediated calcium influx in IMR-32 cells was determined for a panel of blockers with IC(50) values of 0.001-7 µM and this positively correlated with inactivated-state block of recombinant channels measured using electrophysiology. The potency of several compounds was markedly weaker in the state-dependent fluorescence-based assay compared to the electrophysiology assay, although the degree of state-dependent blockade was comparable. CONCLUSIONS: The present data demonstrate that fluorescence-based assays are suitable for assessing the ability of blockers to selectively interact with the inactivated state of the N-type channel.


Assuntos
Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio Tipo N/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo N/farmacologia , Linhagem Celular , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Técnicas de Patch-Clamp
8.
Pain ; 150(2): 319-326, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20621685

RESUMO

The TRPV1 antagonist A-995662 demonstrates analgesic efficacy in monoiodoacetate-induced osteoarthritic (OA) pain in rat, and repeated dosing results in increased in vivo potency and a prolonged duration of action. To identify possible mechanism(s) underlying these observations, release of neuropeptides and the neurotransmitter glutamate from isolated spinal cord was measured. In OA rats, basal release of glutamate, bradykinin and calcitonin gene-related peptide (CGRP) was significantly elevated compared to naïve levels, whereas substance P (SP) levels were not changed. In vitro studies showed that capsaicin-evoked TRPV1-dependent CGRP release was 54.7+/-7.7% higher in OA, relative to levels measured for naïve rats, suggesting that TRPV1 activity was higher under OA conditions. The efficacy of A-995662 in OA corresponded with its ability to inhibit glutamate and CGRP release from the spinal cord. A single, fully efficacious dose of A-995662, 100 micromol/kg, reduced spinal glutamate and CGRP release, while a single sub-efficacious dose of A-995662 (25 micromol/kg) was ineffective. Multiple dosing with A-995662 increased the potency and duration of efficacy in OA rats. Changes in efficacy did not correlate with plasma concentrations of A-995662, but were accompanied with reductions in spinal glutamate release. These findings suggest that repeated dosing of TRPV1 antagonists enhances therapeutic potency and duration of action against OA pain, at least in part, by the sustained reduction in release of glutamate and CGRP from the spinal cord.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Ácido Glutâmico/metabolismo , Osteoartrite do Joelho/metabolismo , Dor/metabolismo , Medula Espinal/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Tetra-Hidronaftalenos/farmacologia , Análise de Variância , Animais , Bradicinina/metabolismo , Osteoartrite do Joelho/induzido quimicamente , Dor/induzido quimicamente , Medição da Dor , Ratos , Ratos Sprague-Dawley , Medula Espinal/metabolismo , Substância P/metabolismo
9.
Brain Res ; 1329: 55-66, 2010 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-20206612

RESUMO

Elevated temperature and decreased extracellular pH are hallmarks of inflammatory pain states. Dorsal root ganglia (DRG) neurons are integral in transferring painful stimuli from the periphery to central sites. This study investigated the effect of elevated temperatures on the response of DRG neurons to acute application of acidic solutions. At room temperature (22 degrees C), in response to pH 5.5, there were a variety of kinetic responses consistent with differential expression of TRPV1 and ASIC channels. Increasing the temperature resulted in a significant increase in the peak and total current mediated by TRPV1 in response to an acidic solution. In contrast, the amplitude of a fast activating, rapidly inactivating ASIC1-like current was not affected by increasing the temperature but did result in an increased rate of desensitization that reduced the total current level. This effect on the rate of desensitization was temperature-dependent and could be reversed by returning to 22 degrees C. Likewise, cells exhibiting slowly inactivating ASIC2-like responses also had temperature-dependent increase in the rate of desensitization. The ASIC2-like responses and the TRPV1 responses tended to decrease in amplitude with repetitive application of pH 5.5 even at 22 degrees C. The rate of desensitization of ASIC-like currents activated by less acidic solutions (pH 6.8) was also increased in a temperature-dependent manner. Finally, acidic pH reduced threshold to trigger action potentials, however, the pattern of action potential firing was shaped by the distribution of ASIC and TRPV1 channels. These results indicate that the ambient temperature at which acidosis occurs has a profound effect on the contribution of ASIC and TRPV1 channels, therefore, altering the neuronal excitability.


Assuntos
Gânglios Espinais/citologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Canais de Sódio/metabolismo , Canais de Cátion TRPV/metabolismo , Temperatura , Canais Iônicos Sensíveis a Ácido , Fatores Etários , Animais , Animais Recém-Nascidos , Células Cultivadas , Eletrofisiologia , Concentração de Íons de Hidrogênio , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley
10.
Protein Expr Purif ; 65(1): 38-50, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19121396

RESUMO

TRPV1 is a ligand-gated cation channel that is involved in acute thermal nociception and neurogenic inflammation. By using the GP67 signal peptide, high levels of full-length human TRPV1 was expressed in High Five insect cells using the baculovirus expression system. The functional activity of the expressed TRPV1 was confirmed by whole-cell ligand-gated ion flux recordings in the presence of capsaicin and low pH and via specific ligand binding to the isolated cellular membranes. Efficient solubilization and purification protocols have resulted in milligram amounts of detergent-solubilized channel at 80-90% purity after Ni2+ IMAC chromatography and size exclusion chromatography. Western blot analysis of amino and carboxyl terminal domains and MS of tryptic digestions of purified protein confirmed the presence of the full-length human TRPV1. Specific ligand binding experiments confirmed the protein integrity of the purified human TRPV1.


Assuntos
Baculoviridae , Expressão Gênica , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Canais de Cátion TRPV/biossíntese , Canais de Cátion TRPV/isolamento & purificação , Animais , Linhagem Celular , Humanos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Spodoptera , Canais de Cátion TRPV/química , Canais de Cátion TRPV/genética
11.
Eur J Pharmacol ; 596(1-3): 62-9, 2008 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-18755179

RESUMO

1-isoquinolin-5-yl-3-(4-trifluoromethyl-benzyl)-urea (A-425619), a novel, potent, and selective transient receptor potential type V1 (TRPV1) antagonist, attenuates pain associated with inflammation and tissue injury in rats. The purpose of this study was to extend the in vitro characterization of A-425619 to native TRPV1 receptors and to compare the pharmacological properties of TRPV1 receptors in the dorsal root ganglion with trigeminal ganglion neurons. A robust increase in intracellular Ca(2+) was elicited by a variety of TRPV1 agonists with similar rank order of potency between both cultures: resiniferatoxin>tinyatoxin>capsaicin>N-arachidonoyl-dopamine (NADA). A-425619 blocked the 500 nM capsaicin response in both dorsal root ganglion with trigeminal ganglion cultures with IC(50) values of 78 nM and 115 nM, respectively, whereas capsazepine was significantly less potent (dorsal root ganglia: IC(50)=2.63 microM; trigeminal ganglia: IC(50)=6.31 microM). Furthermore, A-425619 was more potent in blocking the 3 microM NADA-evoked response in both dorsal root ganglia (IC(50)=36 nM) and trigeminal ganglia (IC(50)=37 nM) than capsazepine (dorsal root ganglia, IC(50)=741 nM; trigeminal ganglia, IC(50)=708 nM). Electrophysiology studies showed that 100 nM A-425619 completely inhibited TRPV1-mediated acid activated currents in dorsal root ganglia and trigeminal ganglia neurons. In addition, A-425619 blocked capsaicin- and NADA-evoked calcitonin gene-related peptide (CGRP) release in both cultures more effectively than capsazepine. These data show that A-425619 is a potent TRPV1 antagonist at the native TRPV1 receptors, and suggest that the pharmacological profile for TRPV1 receptors on dorsal root ganglia and trigeminal ganglia is very similar.


Assuntos
Gânglios Espinais/efeitos dos fármacos , Isoquinolinas/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Gânglio Trigeminal/efeitos dos fármacos , Ureia/análogos & derivados , Aminobutiratos/farmacologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Cálcio/metabolismo , Capsaicina/farmacologia , Células Cultivadas , Gânglios Espinais/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/agonistas , Técnicas de Cultura de Tecidos , Gânglio Trigeminal/fisiologia , Ureia/farmacologia
12.
J Pharmacol Exp Ther ; 326(3): 879-88, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18515644

RESUMO

The transient receptor potential vanilloid (TRPV) 1 receptor, a nonselective cation channel expressed on peripheral sensory neurons and in the central nervous system, plays a key role in pain. TRPV1 receptor antagonism is a promising approach for pain management. In this report, we describe the pharmacological and functional characteristics of a structurally novel TRPV1 antagonist, (R)-(5-tert-butyl-2,3-dihydro-1H-inden-1-yl)-3-(1H-indazol-4-yl)-urea (ABT-102), which has entered clinical trials. At the recombinant human TRPV1 receptor ABT-102 potently (IC(50) = 5-7 nM) inhibits agonist (capsaicin, N-arachidonyl dopamine, anandamide, and proton)-evoked increases in intracellular Ca(2+) levels. ABT-102 also potently (IC(50) = 1-16 nM) inhibits capsaicin-evoked currents in rat dorsal root ganglion (DRG) neurons and currents evoked through activation of recombinant rat TRPV1 currents by capsaicin, protons, or heat. ABT-102 is a competitive antagonist (pA(2) = 8.344) of capsaicin-evoked increased intracellular Ca(2+) and shows high selectivity for blocking TRPV1 receptors over other TRP receptors and a range of other receptors, ion channels, and transporters. In functional studies, ABT-102 blocks capsaicin-evoked calcitonin gene-related peptide release from rat DRG neurons. Intraplantar administration of ABT-102 blocks heat-evoked firing of wide dynamic range and nociceptive-specific neurons in the spinal cord dorsal horn of the rat. This effect is enhanced in a rat model of inflammatory pain induced by administration of complete Freund's adjuvant. Therefore, ABT-102 potently blocks multiple modes of TRPV1 receptor activation and effectively attenuates downstream consequences of receptor activity. ABT-102 is a novel and selective TRPV1 antagonist with pharmacological and functional properties that support its advancement into clinical studies.


Assuntos
Potenciais de Ação/fisiologia , Temperatura Alta , Indazóis/farmacologia , Células do Corno Posterior/metabolismo , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo , Ureia/análogos & derivados , Potenciais de Ação/efeitos dos fármacos , Animais , Linhagem Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Indazóis/química , Masculino , Células do Corno Posterior/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ureia/química , Ureia/farmacologia
13.
Eur J Neurosci ; 27(3): 605-11, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18279313

RESUMO

Transient receptor potential (TRP)A1 channel has been implicated in various physiological processes, including thermosensation and pain. A recent study of TRPA1 knockout mice demonstrated deficits in sensing mechanical stimuli, suggesting a role for TRPA1 also in somatic mechanosensation. However, direct evidence of TRPA1 activation by mechanical forces has thus far been lacking. Here we show, using an intracellular calcium assay, that hypertonic solution (HTS) activates TRPA1 channels in human embryonic kidney 293 cells transiently expressing rat TRPA1. In contrast, hypotonic solution has no effect. Single-channel recordings reveal that HTS opens an ion channel that displays similar single-channel conductance to that evoked by the TRPA1 agonist allyl isothiocyanate (AITC) in both recombinant rat TRPA1 cell lines and rat dorsal root ganglia neurons. Ruthenium red reduces the open probability of the single-channel currents and blocks the whole-cell currents evoked by HTS. Camphor also blocks the whole-cell currents evoked by HTS. HTS-activated channel openings are only observed in patches that are also sensitive to AITC. Finally, like AITC, HTS depolarizes the membrane potential of dorsal root ganglia neurons leading to the generation of action potentials. Taken together, these findings indicate that TRPA1 mediates an osmotically-activated ion channel and support a role for TRPA1 in mechanosensation.


Assuntos
Canais de Cálcio/metabolismo , Canais Iônicos/metabolismo , Mecanorreceptores/metabolismo , Mecanotransdução Celular/fisiologia , Neurônios Aferentes/metabolismo , Tato/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Anquirinas , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/genética , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Cânfora/farmacologia , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Humanos , Soluções Hipertônicas/farmacologia , Canais Iônicos/efeitos dos fármacos , Isotiocianatos/farmacologia , Mecanorreceptores/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios Aferentes/efeitos dos fármacos , Concentração Osmolar , Técnicas de Patch-Clamp , Ratos , Rutênio Vermelho/farmacologia , Canal de Cátion TRPA1 , Canais de Cátion TRPC , Equilíbrio Hidroeletrolítico/efeitos dos fármacos , Equilíbrio Hidroeletrolítico/fisiologia
14.
J Pharmacol Exp Ther ; 323(1): 285-93, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17660385

RESUMO

1-((R)-5-tert-butyl-indan-1-yl)-3-isoquinolin-5-yl-urea (A-778317) is a novel, stereoselective, competitive antagonist that potently blocks transient receptor potential vanilloid-1 (TRPV1) receptor-mediated changes in intracellular calcium concentrations (pIC50 = 8.31 +/- 0.13). The (S)-stereoisomer, 1-((S)-5-tert-butyl-indan-1-yl)-3-isoquinolin-5-yl-urea (A-778316), is 6.8-fold less potent (pIC50 = 7.47 +/- 0.07). A-778317 also potently blocks capsaicin and acid activation of native rat TRPV1 receptors in dorsal root ganglion neurons. A-778317 was tritiated ([3H]A-778317; 29.3 Ci/mmol) and used to study recombinant human TRPV1 (hTRPV1) receptors expressed in Chinese ovary cells (CHO) cells. [3H]A-778317 labeled a single class of binding sites in hTRPV1-expressing CHO cell membranes with high affinity (KD = 3.4 nM; Bmax = 4.0 pmol/mg protein). Specific binding of 2 nM [3H]A-778317 to hTRPV1-expressing CHO cell membranes was reversible. The rank-order potency of TRPV1 receptor antagonists to inhibit binding of 2 nM [3H]A-778317 correlated well with their functional potencies in blocking TRPV1 receptor activation. The present data demonstrate that A-778317 blocks polymodal activation of the TRPV1 receptor by binding to a single high-affinity binding site and that [3H]A-778317 possesses favorable binding properties to facilitate further studies of hTRPV1 receptor pharmacology.


Assuntos
Isoquinolinas/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Ureia/análogos & derivados , Animais , Ligação Competitiva , Células CHO , Sinalização do Cálcio/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , Cricetulus , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Humanos , Isoquinolinas/síntese química , Isoquinolinas/química , Ligantes , Estrutura Molecular , Neurônios Aferentes/metabolismo , Ensaio Radioligante , Ratos , Proteínas Recombinantes/antagonistas & inibidores , Estereoisomerismo , Transfecção , Trítio , Ureia/síntese química , Ureia/química , Ureia/farmacologia
15.
J Neurochem ; 99(4): 1088-102, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17018028

RESUMO

Transient receptor potential channel type V (TRPV) 1 is a non-selective cation channel that can be activated by capsaicin, endogenous vanilloids, heat and protons. The human TRPV1 splice variant, TRPV1b, lacking exon 7, was cloned from human dorsal root ganglia (DRG) RNA. The expression profile and relative abundance of TRPV1b and TRPV1 in 35 different human tissues were determined by quantitative RT-PCR using isoform-specific probes. TRPV1b was most abundant in fetal brain, adult cerebellum and DRG. Functional studies using electrophysiological techniques showed that recombinant TRPV1b was not activated by capsaicin (1 microM), protons (pH 5.0) or heat (50 degrees C). However, recombinant TRPV1b did form multimeric complexes and was detected on the plasma membrane of cells, demonstrating that the lack of channel function was not due to defects in complex formation or cell surface expression. These results demonstrate that exon 7, which encodes the third ankyrin domain and 44 amino acids thereafter, is required for normal channel function of human TRPV1. Moreover, when co-expressed with TRPV1, TRPV1b formed complexes with TRPV1, and inhibited TRPV1 channel function in response to capsaicin, acidic pH, heat and endogenous vanilloids, dose-dependently. Taken together, these data support the hypothesis that TRPV1b is a naturally existing inhibitory modulator of TRPV1.


Assuntos
Sistema Nervoso/metabolismo , Neurônios/metabolismo , Nociceptores/metabolismo , Dor/metabolismo , Canais de Cátion TRPV/metabolismo , Sensação Térmica/genética , Processamento Alternativo/genética , Analgésicos não Narcóticos , Animais , Capsaicina/farmacologia , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Éxons/genética , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Perfilação da Expressão Gênica , Temperatura Alta/efeitos adversos , Humanos , Concentração de Íons de Hidrogênio , Substâncias Macromoleculares/metabolismo , Masculino , Dados de Sequência Molecular , Sistema Nervoso/citologia , Neurônios/citologia , Nociceptores/efeitos dos fármacos , Dor/induzido quimicamente , Dor/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo , Estrutura Terciária de Proteína/genética , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Cátion TRPV/genética , Sensação Térmica/efeitos dos fármacos
16.
Mol Pain ; 1: 28, 2005 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-16191202

RESUMO

TRPV1 (vanilloid receptor 1) receptors are activated by a variety of ligands such as capsaicin, as well as by acidic conditions and temperatures above 42 degrees C. These activators can enhance the potency of one another, shifting the activation curve for TRPV1 to the left. In this study, for example, we observed an approximately 10-fold shift in the capsaicin EC50 (640 nM to 45 nM) for rat TRPV1 receptors expressed in HEK-293 cells when the pH was lowered from 7.4 to 5.5. To investigate potential causes for this shift in capsaicin potency, the rates of current activation and deactivation of whole-cell currents were measured in individual cells exposed to treatments of pH 5.5, 1 microM capsaicin or in combination. Acidic pH was found to both increase the activation rate and decrease the deactivation rate of capsaicin-activated currents providing a possible mechanism for the enhanced potency of capsaicin under acidic conditions. Utilizing a paired-pulse protocol, acidic pH slowed the capsaicin deactivation rate and was readily reversible. Moreover, the effect could occur under modestly acidic conditions (pH 6.5) that did not directly activate TRPV1. When TRPV1 was maximally activated by capsaicin and acidic pH, the apparent affinity of the novel and selective capsaicin-site competitive TRPV1 antagonist, A-425619, was reduced approximately 35 fold. This shift was overcome by reducing the capsaicin concentration co-applied with acidic pH. Since inflammation is associated with tissue acidosis, these findings enhance understanding of TRPV1 receptor responses in inflammatory pain where tissue acidosis is prevalent.


Assuntos
Ácidos/metabolismo , Capsaicina/farmacologia , Canais de Cátion TRPV/metabolismo , Animais , Linhagem Celular , Humanos , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Isoquinolinas/farmacologia , Cinética , Prótons , Ratos , Canais de Cátion TRPV/antagonistas & inibidores , Fatores de Tempo , Ureia/análogos & derivados , Ureia/farmacologia
17.
J Pharmacol Exp Ther ; 314(1): 400-9, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15837819

RESUMO

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.


Assuntos
Temperatura Alta , Canais Iônicos/antagonistas & inibidores , Isoquinolinas/farmacologia , Ureia/análogos & derivados , Ácidos , Animais , Cálcio/metabolismo , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Eletrofisiologia , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/efeitos dos fármacos , Canais de Cátion TRPV , Ureia/farmacologia
18.
Br J Pharmacol ; 140(1): 202-10, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12967950

RESUMO

(1) Rapid desensitization of ligand-gated ion channel receptors can alter the apparent activity of receptor modulators, as well as make detection of fast-channel activation difficult. Investigation of the antagonist pharmacology of ATP-sensitive homomeric P2X3 receptors is limited by agonist-evoked fast-desensitization kinetics. (2) In the present studies, chimeric receptors were created using the coding sequence for the N-terminus and the first transmembrane domain of either the nondesensitizing human P2X2a or fast-desensitizing P2X3 receptor joined to the sequence encoding the extracellular loop, second transmembrane domain, and C-terminus of the other receptor (designated P2X2-3 and P2X3-2, respectively). These clones were stably transfected into 1321N1 astrocytoma cells for biophysical and pharmacological experiments using both electrophysiological and calcium-imaging methods. (3) Chimeric P2X2-3 and P2X3-2 receptors were inwardly rectifying and agonist responses showed desensitization properties similar to the wild-type human P2X2a and P2X3 receptors, respectively. (4) The P2X2-3 chimera displayed an agonist pharmacological profile similar to the P2X3 wild-type receptor being activated by low concentrations of both ATP and alpha,beta-meATP. In contrast, the P2X3-2 chimera had markedly reduced sensitivity to both agonists. (5) The P2X3 receptor antagonists TNP-ATP and A-317491 were shown to be potent, competitive antagonists of the P2X2-3 chimera (Ki=2.2 and 52.1 nm, respectively), supporting the hypothesis that rapid receptor desensitization can mask the competitive antagonism of wild-type homomeric P2X3 receptors.


Assuntos
Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Fenóis/farmacologia , Compostos Policíclicos/farmacologia , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Ligação Competitiva/efeitos dos fármacos , Ligação Competitiva/fisiologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Fenóis/química , Fenóis/metabolismo , Compostos Policíclicos/química , Compostos Policíclicos/metabolismo , Agonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2X3
19.
Muscle Nerve ; 26(6): 817-22, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12451607

RESUMO

Skeletal muscle hyperexcitability is characteristically associated with denervation. Expression of SK3, a small conductance Ca(2+)-activated K(+) channel (SK channel) in skeletal muscle is induced by denervation, and direct application of apamin, a peptide blocker of SK channels, dramatically reduces hyperexcitability. To investigate the role of SK3 channels in denervation- induced hyperexcitability, SK3 expression was manipulated using a transgenic mouse that harbors a tetracycline-regulated SK3 gene. Electromyographic (EMG) recordings from anterior tibial (AT) muscle showed that denervated muscle from transgenic or wild-type animals had equivalent hyperexcitability that was blocked by apamin. In contrast, denervated skeletal muscle from SK3tTA mice lacking SK3 channels showed little or no hyperexcitability, similar to results from wild-type innervated skeletal muscle. However, innervated skeletal muscle from SK3tTA mice containing SK3 channels did not show hyperexcitability. The results demonstrate that SK3 channels are necessary but not sufficient for denervation-induced skeletal muscle hyperexcitability.


Assuntos
Potenciais de Ação/genética , Fasciculação/fisiopatologia , Denervação Muscular/efeitos adversos , Músculo Esquelético/inervação , Músculo Esquelético/fisiopatologia , Doenças Musculares/fisiopatologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/deficiência , Animais , Eletromiografia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Contração Muscular/genética , Canais de Potássio/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...