Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 172(16): 4078-88, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25988595

RESUMO

BACKGROUND AND PURPOSE: Small-molecule inhibitors of prolyl hydroxylase (PHD) enzymes are a novel target for the treatment of anaemia and functional iron deficiency (FID). Other than being orally bioavailable, the differentiation of PHD inhibitors from recombinant human erythropoietin (rhEPO) has not been demonstrated. EXPERIMENTAL APPROACH: JNJ-42905343 was identified and characterized as a novel inhibitor of PHD and its action was compared with rhEPO in healthy rats and in a rat model of inflammation-induced anaemia and FID [peptidoglycan-polysaccharide (PGPS) model]. KEY RESULTS: Oral administration of JNJ-42905343 to healthy rats increased the gene expression of cytochrome b (DcytB) and divalent metal-ion transporter 1 (DMT1) in the duodenum, and increased plasma EPO. Repeated administration of JNJ-42905343 for 28 days increased blood haemoglobin, mean corpuscular haemoglobin (MCH) and mean corpuscular volume (MCV). The serum iron concentration was increased with low doses (0.3 mg·kg(-1) ) but reduced at high doses (6 mg·kg(-1) ). In PGPS-treated rats, administration of JNJ-42905343 for 28 days corrected FID and anaemia, as reflected by increased blood haemoglobin, MCH and MCV. Increased expression of DcytB and DMT1 genes in the duodenum resulting in increased iron availability was defined as the mechanism for these effects. rhEPO did not affect DcytB and DMT1 and was not effective in PGPS-treated rats. CONCLUSIONS AND IMPLICATIONS: PHD inhibition has a beneficial effect on iron metabolism in addition to stimulating the release of EPO. Small-molecule inhibitors of PHD such as JNJ-42905343 represent a mechanism distinct from rhEPO to treat anaemia and FID.


Assuntos
Anemia/tratamento farmacológico , Inibidores de Prolil-Hidrolase/uso terapêutico , Pirazóis/uso terapêutico , Quinazolinonas/uso terapêutico , Anemia/sangue , Anemia/genética , Anemia/metabolismo , Animais , Proteínas de Transporte de Cátions/genética , Citocromos b/genética , Duodeno/efeitos dos fármacos , Duodeno/metabolismo , Índices de Eritrócitos/efeitos dos fármacos , Eritropoetina/sangue , Eritropoetina/genética , Eritropoetina/farmacologia , Eritropoetina/uso terapêutico , Feminino , Expressão Gênica/efeitos dos fármacos , Hemoglobinas/análise , Inflamação/sangue , Inflamação/induzido quimicamente , Inflamação/complicações , Inflamação/genética , Inflamação/metabolismo , Ferro/sangue , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Peptidoglicano , Inibidores de Prolil-Hidrolase/farmacocinética , Inibidores de Prolil-Hidrolase/farmacologia , Pirazóis/farmacocinética , Pirazóis/farmacologia , Quinazolinonas/farmacocinética , Quinazolinonas/farmacologia , Ratos Endogâmicos Lew
3.
J Pharmacol Exp Ther ; 338(1): 328-36, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21493750

RESUMO

JNJ-26070109 [(R)4-bromo-N-[1-(2,4-difluoro-phenyl)-ethyl]-2-(quinoxaline-5-sulfonylamino)-benzamide] is a representative of a new chemical class of competitive antagonists of cholecystokinin 2 (CCK2) receptors. In this study, the primary in vitro pharmacology of JNJ-26070109 was evaluated along with the pharmacokinetic and pharmacodynamic properties of this compound in rat and canine models of gastric acid secretion. JNJ-26070109 expressed high affinity for human (pK(I) = 8.49 ± 0.13), rat (pK(I) = 7.99 ± 0.08), and dog (pK(I) = 7.70 ± 0.14) CCK2 receptors. The selectivity of JNJ-26070109 at the CCK2 receptor versus the CCK1 receptor was species-dependent, with the greatest degree of selectivity (>1200-fold) measured at the human isoforms of the CCK1 receptor (selectivity at CCK2 versus CCK1 receptors: human, ∼1222-fold; rat, ∼324-fold; dog ∼336-fold). JNJ-26070109 behaved as a surmountable, competitive, antagonist of human CCK2 receptors in a calcium mobilization assay (pK(B) = 8.53 ± 0.05) and in pentagastrin-stimulated gastric acid secretion in the isolated, lumen-perfused, mouse stomach assay (pK(B) = 8.19 ± 0.13). The pharmacokinetic profile of this compound was determined in vivo in rats and dogs. JNJ-26070109 was shown to have high oral bioavailability (%F rat = 73 ± 16; %F dog = 92 ± 12) with half lives of 1.8 ± 0.3 and 1.2 ± 0.1 h in rat and dog, respectively. The pharmacodynamic properties of this compound were investigated using two in vivo models. In conscious rat and dog chronic gastric fistula models of pentagastrin-stimulated acid secretion, JNJ-26070109 had oral EC(50) values of 1.5 and 0.26 µM, respectively. Overall, we have demonstrated that JNJ-26070109 is a high-affinity, selective CCK2 receptor antagonist with good pharmacokinetic properties.


Assuntos
Benzodiazepinonas/administração & dosagem , Benzodiazepinonas/metabolismo , Compostos de Fenilureia/administração & dosagem , Compostos de Fenilureia/metabolismo , Quinoxalinas/administração & dosagem , Receptor de Colecistocinina B/antagonistas & inibidores , Receptor de Colecistocinina B/metabolismo , Sulfonamidas/administração & dosagem , Administração Oral , Animais , Benzodiazepinonas/química , Disponibilidade Biológica , Células CHO , Células CACO-2 , Cricetinae , Cricetulus , Cães , Relação Dose-Resposta a Droga , Feminino , Cobaias , Humanos , Masculino , Camundongos , Compostos de Fenilureia/química , Quinoxalinas/química , Quinoxalinas/metabolismo , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Sulfonamidas/química , Sulfonamidas/metabolismo
4.
Mol Pharmacol ; 79(6): 910-20, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21372172

RESUMO

The hypoxia-inducible factor (HIF) prolyl hydroxylase (PHD) enzymes represent novel targets for the treatment of anemia, ulcerative colitis, and ischemic and metabolic disease inter alia. We have identified a novel small-molecule inhibitor of PHD, 1-(5-chloro-6-(trifluoromethoxy)-1H-benzoimidazol-2-yl)-1H-pyrazole-4-carboxylic acid (JNJ-42041935), through structure-based drug design methods. The pharmacology of JNJ-42041935 was investigated in enzyme, cellular, and whole-animal systems and was compared with other compounds described in the literature as PHD inhibitors. JNJ-42041935, was a potent (pK(I) = 7.3-7.9), 2-oxoglutarate competitive, reversible, and selective inhibitor of PHD enzymes. In addition, JNJ-42041935 was used to compare the effect of selective inhibition of PHD to intermittent, high doses (50 µg/kg i.p.) of an exogenous erythropoietin receptor agonist in an inflammation-induced anemia model in rats. JNJ-42041935 (100 µmol/kg, once a day for 14 days) was effective in reversing inflammation-induced anemia, whereas erythropoietin had no effect. The results demonstrate that JNJ-42041935 is a new pharmacological tool, which can be used to investigate PHD inhibition and demonstrate that PHD inhibitors offer great promise for the treatment of inflammation-induced anemia.


Assuntos
Benzimidazóis/farmacologia , Inibidores Enzimáticos/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pirazóis/farmacologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Dados de Sequência Molecular , Pró-Colágeno-Prolina Dioxigenase/química , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Ligação Proteica , Ratos , Ratos Endogâmicos Lew
5.
ACS Med Chem Lett ; 1(9): 526-9, 2010 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24900242

RESUMO

HIF prolyl 4-hydroxylases (PHD) are a family of enzymes that mediate key physiological responses to hypoxia by modulating the levels of hypoxia inducible factor 1-α (HIF1α). Certain benzimidazole-2-pyrazole carboxylates were discovered to be PHD2 inhibitors using ligand- and structure-based methods and found to be potent, orally efficacious stimulators of erythropoietin secretion in vivo.

6.
Adv Hematol ; 2009: 251915, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19946423

RESUMO

The heme-regulated inhibitor (HRI) negatively regulates protein synthesis by phosphorylating eukaryotic initiation factor-2alpha (eIF2alpha) thereby inhibiting protein translation. The importance of HRI in regulating hemoglobin synthesis in erythroid cells makes it an attractive molecular target in need of further characterization. In this work, we have cloned and expressed the canine form of the HRI kinase. The canine nucleotide sequence has 86%, 82%, and 81% identity to the human, mouse, and rat HRI, respectively. It was noted that an isoleucine residue in the ATP binding site of human, rat, and mouse HRI is replaced by a valine in the canine kinase. The expression of canine HRI protein by in vitro translation using wheat germ lysate or in Sf9 cells using a baculovirus expression system was increased by the addition of hemin. Following purification, the canine protein was found to be 72 kD and showed kinase activity determined by its ability to phosphorylate a synthetic peptide substrate. Quercetin, a kinase inhibitor known to inhibit mouse and human HRI, inhibits canine HRI in a concentration-dependent manner. Additionally, quercetin is able to increase de novo protein synthesis in canine reticulocytes. We conclude that the canine is a suitable model species for studying the role of HRI in erythropoiesis.

9.
Bioorg Med Chem Lett ; 19(22): 6376-8, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19815410

RESUMO

In the previous article we demonstrated how certain CCK2R-selective anthranilic amides could be structurally modified to afford high-affinity, selective CCK1R activity. We now describe our efforts at modulating and optimizing the CCK1R and CCK2R affinities aimed at producing compounds with good pharmacokinetics properties and in vivo efficacy in rat models of gastric acid and pancreatic amylase secretion.


Assuntos
Isoxazóis/química , Receptor de Colecistocinina A/antagonistas & inibidores , Receptor de Colecistocinina B/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Substituição de Aminoácidos , Animais , Modelos Moleculares , Ratos , Ratos Wistar , Resultado do Tratamento
10.
J Biomol Screen ; 14(6): 627-35, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19498081

RESUMO

The prolyl-4-hydroxylase proteins regulate the hypoxia-inducible transcription factors (HIFs) by hydroxylation of proline residues targeting HIF-1alpha for proteasomal degradation. Using the purified catalytic domain of prolyl hydroxylase 2 (PHD2(181-417)), an enzymatic assay has been developed to test inhibitors of the enzyme in vitro. Because PHD2 hydroxylates HIF-1alpha, with succinic acid produced as an end product, radiolabeled [5-(14)C]-2-oxoglutaric acid was used and formation of [14C]-succinic acid was measured to quantify PHD2(181-417) enzymatic activity. Comparison of the separation of 2-oxoglutaric acid and succinic acid by either ion exchange chromatography or precipitation with phenylhydrazine showed similar results, but the quantification and throughput were vastly increased using the latter method. The PHD2 reaction was substrate and concentration dependent. The addition of iron to the enzyme reaction mix resulted in an increase in enzymatic activity. The Km value for 2-oxoglutaric acid was determined to be 0.9 microM, and known PHD2 inhibitors were used to validate the assay. In addition, the authors demonstrate that this assay can be applied to other 2-oxoglutaric acid-dependent enzymes, including the asparaginyl hydroxylase, factor-inhibiting HIF-1alpha (FIH). A concentration-dependent increase in succinic acid production using recombinant FIH enzyme with a synthetic peptide substrate was observed. The authors conclude that a by-product enzyme assay measuring the conversion of 2-oxoglutaric acid to succinic acid using the catalytic domain of the human PHD2 provides a convenient method for the biochemical evaluation of inhibitors of the 2-oxoglutaric acid-dependent hydroxylases.


Assuntos
Bioensaio/métodos , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/farmacologia , Ácidos Cetoglutáricos/metabolismo , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Precipitação Química , Cromatografia por Troca Iônica , Humanos , Hidrazinas/metabolismo , Hidroxilação/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia , Ácidos Cetoglutáricos/química , Ácidos Cetoglutáricos/isolamento & purificação , Cinética , Peptídeos/metabolismo , Proteínas Repressoras/metabolismo , Especificidade por Substrato/efeitos dos fármacos , Ácido Succínico/química , Ácido Succínico/isolamento & purificação , Titulometria
11.
Bioorg Med Chem ; 16(7): 3917-25, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18289861

RESUMO

A novel series of cholecystokinin-2 receptor (CCK-2R) antagonists has been identified, as exemplified by anthranilic sulfonamide 1 (pK(i)=7.6). Pharmacokinetic and stability studies indicated that this series of compounds suffered from metabolic degradation, and that both the benzothiadiazole and piperidine rings were rapidly oxidized by liver enzymes. A combination of synthesis, computational methods, (1)H NMR conformational studies, and X-ray crystallographic analyses were applied to elucidate key pharmacophore elements, and to discover analogs with improved pharmacokinetic profiles, and high receptor binding affinity and selectivity.


Assuntos
Receptor de Colecistocinina B/antagonistas & inibidores , Animais , Benzotiazóis/síntese química , Benzotiazóis/química , Benzotiazóis/farmacologia , Cristalografia por Raios X , Humanos , Espectroscopia de Ressonância Magnética , Microssomos Hepáticos/efeitos dos fármacos , Modelos Moleculares , Estrutura Molecular , Receptor de Colecistocinina A/metabolismo , Receptor de Colecistocinina B/metabolismo , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia
12.
Bioorg Med Chem Lett ; 17(24): 6905-9, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18029172

RESUMO

A novel strategy for the synthesis of cholecystokinin-2 receptor ligands was developed. The route employs a solution-phase synthesis of a series of anthranilic sulfonamides followed by a resin capture purification strategy to produce multi-milligram quantities of compounds for bioassay. The synthesis was used to produce >100 compounds containing various functional groups, highlighting the general applicability of this strategy and to address specific metabolism issues in our CCK-2 program.


Assuntos
Colecistocinina/metabolismo , Técnicas de Química Combinatória , Receptor de Colecistocinina B/metabolismo , Sulfonamidas/síntese química , Sulfonamidas/farmacologia , ortoaminobenzoatos/síntese química , ortoaminobenzoatos/farmacologia , Animais , Desenho de Fármacos , Humanos , Ligantes , Microssomos Hepáticos/efeitos dos fármacos , Estrutura Molecular , Ratos , Relação Estrutura-Atividade , Sulfonamidas/química , ortoaminobenzoatos/química
13.
J Pharmacol Exp Ther ; 323(2): 562-9, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17684117

RESUMO

3-[5-(3,4-Dichloro-phenyl)-1-(4-methoxy-phenyl)-1H-pyrazol-3-yl]-2-m-tolyl-propionate (JNJ-17156516) is a novel, potent, and selective cholecystokinin (CCK)1-receptor antagonist. In this study, the pharmacology of JNJ-17156516 was investigated both in vitro and in vivo, and the pharmacokinetic profile was evaluated in rats. JNJ-17156516 expressed high-affinity at the cloned human (pK(I) = 7.96 +/- 0.11), rat (pK(I) = 8.02 +/- 0.11), and canine (pK(I) = 7.98 +/- 0.04) CCK1 receptors, and it was also highly selective for the CCK1 receptor compared with the CCK2 receptor across the same species ( approximately 160-, approximately 230-, and approximately 75-fold, respectively). The high affinity of JNJ-17156516 at CCK1 receptors in vitro was confirmed in radioligand binding studies on fresh human gallbladder tissue (pK(I) = 8.22 +/- 0.05). In a functional in vitro assay of guinea pig gallbladder contraction, JNJ-17156516 behaved as a competitive antagonist, with a pK(B) value of 8.00 +/- 0.07. In vivo, JNJ-17156516 produced a parallel, rightward shift in the CCK-8S-evoked contraction of the guinea pig gallbladder. The dose required to shift the CCK-8S dose-response curve was 240 nmol kg(-1) i.v. In the anesthetized rat, JNJ-17156516 produced a dose-related decrease in the number of duodenal contractions evoked by infusion of CCK-8S, with an ED(50) = 484 nmol kg(-1). Pharmacokinetic analysis of JNJ-17156516 in rats, revealed that JNJ-17156516 had a half-life of 3.0 +/- 0.5 h and a very high bioavailability (108 +/- 10%) in this species. Overall, we have demonstrated that JNJ-17156516 is a high-affinity selective human CCK1 receptor antagonist with good pharmacokinetic properties in rats.


Assuntos
Ácidos Pentanoicos/farmacologia , Fenilpropionatos/farmacologia , Pirazóis/farmacologia , Receptor de Colecistocinina A/antagonistas & inibidores , Animais , Cricetinae , Cães , Relação Dose-Resposta a Droga , Vesícula Biliar/efeitos dos fármacos , Vesícula Biliar/fisiologia , Cobaias , Humanos , Técnicas In Vitro , Masculino , Contração Muscular/efeitos dos fármacos , Fenilpropionatos/metabolismo , Pirazóis/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina A/metabolismo
14.
J Pharmacol Exp Ther ; 323(2): 665-74, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17690251

RESUMO

Transient receptor potential vanilloid 1 (TRPV1) plays an integral role in modulating the cough reflex, and it is an attractive antitussive drug target. The purpose of this study was to characterize a TRPV1 antagonist, 4-(3-trifluoromethyl-pyridin-2-yl)-piperazine-1-carboxylic acid (5-trifluoromethyl-pyridin-2-yl)-amide (JNJ17203212), against the guinea pig TRPV1 receptor in vitro followed by a proof-of-principle study in an acid-induced model of cough. The affinity of JNJ17203212 for the recombinant guinea pig TRPV1 receptor was estimated by radioligand binding, and it was functionally characterized by antagonism of low-pH and capsaicin-induced activation of the ion channel (fluorometric imaging plate reader and electrophysiology). The nature of antagonism was further tested against the native channel in isolated guinea pig tracheal rings. Following pharmacokinetic characterization of JNJ17203212 in guinea pigs, pharmacodynamic and efficacy studies were undertaken to establish the antitussive efficacy of the TRPV1 antagonist. The pK(i) of JNJ17203212 for recombinant guinea pig TRPV1 was 7.14 +/- 0.06. JNJ17203212 inhibited both pH (pIC(50) of 7.23 +/- 0.05) and capsaicin (pIC(50) of 6.32 +/- 0.06)-induced channel activation. In whole-cell patch clamp, the pIC(50) for inhibition of guinea pig TRPV1 was 7.3 +/- 0.01. JNJ17203212 demonstrated surmountable antagonism in isolated trachea, with a pK(B) value of 6.2 +/- 0.1. Intraperitoneal administration of 20 mg/kg JNJ17203212 achieved a maximal plasma exposure of 8.0 +/- 0.4 microM, and it attenuated capsaicin evoked coughs with similar efficacy to codeine (25 mg/kg). Last, JNJ17203212 dose-dependently produced antitussive efficacy in citric acid-induced experimental cough in guinea pigs. Our data provide preclinical support for developing TRPV1 antagonists for the treatment of cough.


Assuntos
Aminopiridinas/farmacologia , Antitussígenos/farmacologia , Piperazinas/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Aminopiridinas/sangue , Animais , Células CHO , Tosse/tratamento farmacológico , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Feminino , Cobaias , Masculino , Piperazinas/sangue
15.
Biochem Biophys Res Commun ; 357(1): 264-9, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17418097

RESUMO

Obestatin was recently described as a bioactive peptide encoded for by the same gene as ghrelin but with opposite actions on food intake. Although some groups have confirmed these findings others find no effect. We investigated the effect of obestatin on feeding in rodents over a wide range of doses. Acute administration of obestatin inhibited feeding at doses of 10-100 nmol/kg i.p. in mice and 100-300 nmol/kg i.p. in lean and Zucker fatty rats. Interestingly, the dose-response relationship was U-shaped such that both low and high doses were without effect in either species. Treatment of mice with obestatin over a 7-day period decreased body weight gain and food consumption. Overall, obestatin suppressed food intake and body weight gain in rodent and an unusual dose-response relationship was found. These findings may explain the difficulties in reproducing the effects of obestatin on feeding reported by some groups.


Assuntos
Peso Corporal/fisiologia , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Hormônios Peptídicos/administração & dosagem , Aumento de Peso/fisiologia , Administração Oral , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Grelina , Masculino , Ratos , Ratos Sprague-Dawley , Aumento de Peso/efeitos dos fármacos
16.
J Med Chem ; 49(21): 6371-90, 2006 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17034143

RESUMO

A high throughput screening approach to the identification of selective cholecystokinin-2 receptor (CCK-2R) ligands resulted in the discovery of a novel series of antagonists, represented by 1-[2-[(2,1,3-benzothiadiazol-4-ylsulfonyl)amino]-5-chlorobenzoyl]-piperidine (1; CCK-2R, pK(I) = 6.4). Preliminary exploration of the structure-activity relationships around the anthranilic ring and the amide and sulfonamide moieties led to a nearly 50-fold improvement of receptor affinity and showed a greater than 1000-fold selectivity over the related cholecystokinin-1 receptor. Pharmacokinetic evaluation led to the identification of 4-[4-iodo-2-[(5-quinoxalinylsulfonyl)amino]benzoyl]-morpholine, 26d, a compound that demonstrates promising pharmacokinetic properties in the rat and dog with respect to plasma clearance and oral bioavailability and is a potent inhibitor in vivo of pentagastrin-stimulated acid secretion in the rat when dosed orally.


Assuntos
Morfolinas/síntese química , Quinoxalinas/síntese química , Receptor de Colecistocinina B/antagonistas & inibidores , Sulfonamidas/síntese química , ortoaminobenzoatos/síntese química , Animais , Ligação Competitiva , Disponibilidade Biológica , Cães , Estabilidade de Medicamentos , Vesícula Biliar/efeitos dos fármacos , Vesícula Biliar/fisiologia , Ácido Gástrico/metabolismo , Cobaias , Humanos , Técnicas In Vitro , Masculino , Microssomos Hepáticos/metabolismo , Morfolinas/química , Morfolinas/farmacologia , Contração Muscular , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiologia , Quinoxalinas/química , Quinoxalinas/farmacologia , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina A/antagonistas & inibidores , Estereoisomerismo , Estômago/efeitos dos fármacos , Estômago/fisiologia , Relação Estrutura-Atividade , Sulfonamidas/química , Sulfonamidas/farmacologia , ortoaminobenzoatos/química , ortoaminobenzoatos/farmacologia
17.
Regul Pept ; 136(1-3): 109-16, 2006 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-16782214

RESUMO

The neuropeptide Neuromedin U (NMU) stimulates smooth muscle contraction, and modulates local blood flow and adrenocortical function via two endogenous receptors, NMU1 and NMU2. Although its amino-acid sequence is highly conserved across species, the physiological effects of NMU are variable between species and little is known of its effects on human tissues. We have examined the contractile effects of NMU-25 on human smooth muscles of the gastrointestinal (GI) tract (ascending colon, gallbladder) and long saphenous vein (LSV) using in vitro organ bath bioassays. From LSV, ileum, gallbladder, caecum and colon, NMU receptor transcripts were amplified by RT-PCR and expression levels were determined by semi-quantitative scanning densitometry. NMU-25 produced a concentration-dependent, sustained contraction of isolated smooth muscle (p[A](50)+/-s.e.m., ascending colon, 8.93+/-0.18; gallbladder, 7.01+/-0.15; LSV, 8.67+/-0.09). NMU1 and NMU2 receptor transcription was detected in all tissues; transcription of both receptors was similar in gallbladder, but NMU1 receptor transcription was predominant in the sigmoid colon and LSV. In summary, these studies indicate that NMU may control tone in the human GI tract and LSV through an action on smooth muscle. Development of NMU receptor subtype-selective ligands will aid the further elucidation of the physiological roles of NMU and its two receptors.


Assuntos
Músculo Liso/patologia , Neuropeptídeos/farmacologia , Veia Safena/patologia , Colo/metabolismo , Densitometria , Vesícula Biliar/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Contração Muscular , Músculo Liso/metabolismo , Neuropeptídeos/química , Neuropeptídeos/metabolismo , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica
18.
Br J Pharmacol ; 147(8): 886-96, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16474416

RESUMO

The peptide ligand neuromedin U (NMU) has been implicated in an array of biological activities, including contraction of uterine, intestinal and urinary bladder smooth muscle. However, many of these responses appear to be species-specific. This study was undertaken to fully elucidate the range of smooth muscle-stimulating effects of NMU in rats, mice and guinea-pigs, and to examine the extent of the species differences. In addition, the NMU1 receptor knockout mouse was used to determine which receptor subtype mediates the contractile responses generated by NMU in the mouse. A range of isolated organ in vitro bioassays were carried out, which were chosen to re-confirm previous literature reports (uterine and stomach fundus contraction) and also to explore potentially novel smooth muscle responses to NMU. This investigation uncovered a number of previously unidentified NMU-mediated responses: contraction of rat lower esophageal sphinster (LES), rat ileum, mouse gallbladder, enhancement of electrically evoked contractions in rat and mouse vas deferens, and a considerable degree of cross-species differences. Studies using the NMU1 receptor knockout mice revealed that in the mouse fundus and gallbladder assays the NMU contractile response was mediated entirely through the NMU1 receptor subtype, whereas, in assays of mouse uterus and vas deferens, the response to NMU was unchanged in the NMU1 receptor knockout mouse, suggesting that the NMU response may be mediated through the NMU2 receptor subtype. NMU receptor subtype-selective antagonists are required to further elucidate the role of the individual receptor subtypes.


Assuntos
Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Neuropeptídeos/farmacologia , Receptores de Neurotransmissores/genética , Receptores de Neurotransmissores/metabolismo , Animais , Feminino , Cobaias , Masculino , Camundongos , Camundongos Knockout , Contração Muscular/fisiologia , Músculo Liso/metabolismo , Ratos , Ratos Sprague-Dawley
19.
J Pharmacol Toxicol Methods ; 54(1): 36-41, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16246596

RESUMO

INTRODUCTION: Cholecystokinin type-1 (CCK(1)) receptors mediate many of the physiological functions of CCK including delay of gastric emptying, pancreatic enzyme secretion, intestinal motility and gallbladder contractility. Existing in-vivo assays for the quantitative measurement of CCK(1) receptor mediated function are generally variable, limited in precision and require a relatively large number of animals to obtain statistically meaningful data. We found that they did not provide robust pharmacokinetic-pharmacodynamic data for profiling compounds acting at these receptors. Accordingly, here we describe a novel rat duodenal contractility assay that addresses these problems. METHODS: Rats were anaesthetised and a saline-filled balloon was inserted through the body of the stomach and secured in the duodenum approximately 1 cm from the pyloric sphincter for measurement of intra-lumenal pressure. Studies were performed to determine a dose, rate and frequency of administration of CCK8S that produced a readily quantifiable response. RESULTS: Initial experiments revealed that sustained exposure to CCK8S resulted in the rapid development of tachyphylaxis. After investigating different dosing paradigms, it was found that pulsatile delivery of CCK8S (intravenous infusion for 1 min every 10 min) produced a readily quantifiable contractile response that did not exhibit tachyphylaxis. The assay response output was defined as the number of contractions >5 mm Hg over baseline. The contractions were blocked in a dose-dependent manner by intravenous bolus injections of the CCK(1) receptor antagonists, dexloxiglumide (2 and 20 micromol/kg), and devazepide (3-100 nmol/kg) but not by the CCK(2) receptor antagonist gastrazole (10 micromol/kg). CONCLUSION: A novel, simple, high quality assay for the quantification of the in-vivo activity of CCK(1) receptor ligands is described. CCK8S delivered by pulsatile intravenous infusion to anesthetized rats produced a burst of contractile activity of the duodenum mediated by CCK(1) receptors. This activity was highly reproducible and sustained for more than 3 h providing an assay that circumvents problems associated with agonist-induced tachyphylaxis.


Assuntos
Anestesia/métodos , Bioensaio/métodos , Receptor de Colecistocinina A/metabolismo , Animais , Relação Dose-Resposta a Droga , Duodeno/efeitos dos fármacos , Duodeno/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina A/agonistas , Receptor de Colecistocinina A/antagonistas & inibidores , Sincalida/farmacologia
20.
J Med Chem ; 48(22): 6790-802, 2005 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-16250638

RESUMO

A new molecular modeling approach has been used to derive a pharmacophore of the potent and selective cholecystokinin-2 (CCK(2)) receptor antagonist 5 (JB93182), based on features shared with two related series. The technique uses "field points" as simple and effective descriptions of the electrostatic and van der Waals maxima and minima surrounding a molecule equipped with XED (extended electron distribution) charges. Problems associated with the high levels of biliary elimination of 5 in vivo required us to design a compound with significantly lower molecular weight without sacrificing its nanomolar levels of in vitro activity. Two new series of compounds were designed to mimic the arrangement of field points present in the pharmacophore rather than its structural elements. In a formal sense, two of the three amides in 5 were replaced with either a simple pyrrole or imidazole, while some features thought to be essential for the high levels of in vitro activity of the parent compounds were retained and others deleted. These compounds maintained activity and selectivity for this receptor over CCK(1). In addition, the reduction in molecular weight coupled with lower polarities greatly reduced levels of biliary elimination associated with 5. This makes them good lead compounds for development of drug candidates whose structures are not obviously related to those of the parents and represents the first example of scaffold hopping using molecular field points.


Assuntos
Imidazóis/química , Modelos Moleculares , Pirróis/química , Receptor de Colecistocinina B/agonistas , Receptor de Colecistocinina B/antagonistas & inibidores , Animais , Ligação Competitiva , Córtex Cerebral/metabolismo , Desenho de Fármacos , Imidazóis/síntese química , Imidazóis/farmacologia , Técnicas In Vitro , Camundongos , Conformação Molecular , Pirróis/síntese química , Pirróis/farmacologia , Relação Quantitativa Estrutura-Atividade , Ensaio Radioligante , Receptor de Colecistocinina B/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA