Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Br J Pharmacol ; 176(24): 4625-4638, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31404942

RESUMEN

BACKGROUND AND PURPOSE: Microsomal PGE synthase-1 (mPGES-1), the inducible synthase that catalyses the terminal step in PGE2 biosynthesis, is of high interest as therapeutic target to treat inflammation. Inhibition of mPGES-1 is suggested to be safer than traditional NSAIDs, and recent data demonstrate anti-constrictive effects on vascular tone, indicating new therapeutic opportunities. However, there is a lack of potent mPGES-1 inhibitors lacking interspecies differences for conducting in vivo studies in relevant preclinical disease models. EXPERIMENTAL APPROACH: Potency was determined based on the reduction of PGE2 formation in recombinant enzyme assays, cellular assay, human whole blood assay, and air pouch mouse model. Anti-inflammatory properties were assessed by acute paw swelling in a paw oedema rat model. Effect on vascular tone was determined with human ex vivo wire myography. KEY RESULTS: We report five new mPGES-1 inhibitors (named 934, 117, 118, 322, and 323) that selectively inhibit recombinant human and rat mPGES-1 with IC50 values of 10-29 and 67-250 nM respectively. The compounds inhibited PGE2 production in a cellular assay (IC50 values 0.15-0.82 µM) and in a human whole blood assay (IC50 values 3.3-8.7 µM). Moreover, the compounds blocked PGE2 formation in an air pouch mouse model and reduced acute paw swelling in a paw oedema rat model. Human ex vivo wire myography analysis showed reduced adrenergic vasoconstriction after incubation with the compounds. CONCLUSION AND IMPLICATIONS: These mPGES-1 inhibitors can be used as refined tools in further investigations of the role of mPGES-1 in inflammation and microvascular disease.


Asunto(s)
Antiinflamatorios/farmacología , Arterias/efectos de los fármacos , Dinoprostona/biosíntesis , Edema/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Tono Muscular/efectos de los fármacos , Prostaglandina-E Sintasas/antagonistas & inhibidores , Células A549 , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacocinética , Arterias/enzimología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Edema/inmunología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Escherichia coli/genética , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Miografía , Prostaglandina-E Sintasas/sangre , Prostaglandina-E Sintasas/genética
2.
Prostaglandins Other Lipid Mediat ; 107: 26-34, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24045148

RESUMEN

Microsomal prostaglandin E synthase-1 (mPGES-1) inhibition has been suggested as an alternative to cyclooxygenase (COX) inhibition in the treatment of pain and inflammation. We characterized a selective inhibitor of mPGES-1 activity (compound III) and studied its impact on the prostanoid profile in various models of inflammation. Compound III is a benzoimidazole, which has a submicromolar IC50 in both human and rat recombinant mPGES-1. In cellular assays, it reduced PGE2 production in A549 cells, mouse macrophages and blood, causing a shunt to the prostacyclin pathway in the former two systems. Lastly, we assayed compound III in the air pouch model to verify its impact on the prostanoid profile and compare it to the profile obtained in mPGES-1 k.o. mice. As opposed to mPGES-1 genetic deletion, which attenuated PGE2 production and caused a shunt to the thromboxane pathway, mPGES-1 inhibition with compound III reduced PGE2 production and tended to decrease the levels of other prostanoids.


Asunto(s)
Bencimidazoles/farmacología , Inhibidores Enzimáticos/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Ácidos Isonipecóticos/farmacología , Animales , Línea Celular Tumoral , Dinoprostona/metabolismo , Evaluación Preclínica de Medicamentos , Técnicas de Inactivación de Genes , Humanos , Concentración 50 Inhibidora , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos DBA , Ratones Noqueados , Prostaglandina H2/metabolismo , Prostaglandina-E Sintasas , Ratas , Tromboxano B2/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-23528231

RESUMEN

Microsomal prostaglandin E synthase (mPGES)-1 inhibition has been proposed as an alternative to cyclooxygenase (COX) inhibition in the treatment of pain and inflammation. This novel approach could potentially mitigate the gastro-intestinal and cardiovascular side effects seen after long-term treatment with traditional non-steroidal anti-inflammatory drugs (NSAIDs) and Coxibs respectively. Several human mPGES-1 inhibitors have been developed in the recent years. However, they were all shown to be considerably less active on rodent mPGES-1, precluding the study of mPGES-1 inhibition in rodent models of inflammation and pain. The aim of this study was to characterize the new mPGES-1 inhibitor compound II, a pyrazolone that has similar potency on rat and human recombinant mPGES-1, in experimental models of inflammation. In cell culture, compound II inhibited PGE2 production in synovial fibroblasts from patients with rheumatoid arthritis (RASF) and in rat peritoneal macrophages. In vivo, compound II was first characterized in the rat air pouch model of inflammation where treatment inhibited intra-pouch PGE2 production. Compound II was also investigated in a rat adjuvant-induced arthritis model where it attenuated both the acute and delayed inflammatory responses. In conclusion, compound II represents a valuable pharmacological tool for the study of mPGES-1 inhibition in rat models.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Inflamación/tratamiento farmacológico , Prostaglandina-Endoperóxido Sintasas/metabolismo , Pirazolonas/administración & dosificación , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/enzimología , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Dinoprostona/biosíntesis , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Humanos , Inflamación/enzimología , Inflamación/patología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/patología , Dolor/tratamiento farmacológico , Dolor/patología , Prostaglandina-E Sintasas , Prostaglandina-Endoperóxido Sintasas/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/genética , Ratas , Líquido Sinovial/efectos de los fármacos , Líquido Sinovial/enzimología
4.
Assay Drug Dev Technol ; 9(5): 487-95, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21561373

RESUMEN

Microsomal prostaglandin E(2) synthase-1 (MPGES1) catalyzes the formation of prostaglandin E(2) from the endoperoxide prostaglandin H(2). MPGES1 expression is induced in inflammatory diseases, and this enzyme is regarded as a potential drug target. To aid in the drug discovery effort, a simple method for determination of inhibition mechanism and potency toward both prostaglandin H(2) and glutathione (GSH) has been developed. Using an assay with thiobarbituric acid-based detection, the inhibitory effects of six MPGES1 inhibitors were evaluated. The IC(50) values obtained at three substrate (S) concentrations ([S]K(M)) were used to estimate inhibition modality and inhibition constant values. This facilitated strategy is a useful and general screening method to evaluate the inhibitory effects of new drug compounds.


Asunto(s)
Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Interacciones Farmacológicas , Inhibidores Enzimáticos/metabolismo , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Fluorescencia , Glutatión/análisis , Humanos , Indoles/análisis , Indoles/farmacocinética , Indoles/farmacología , Concentración 50 Inhibidora , Oxidorreductasas Intramoleculares/análisis , Oxidorreductasas Intramoleculares/fisiología , Malondialdehído/metabolismo , Modelos Teóricos , Terapia Molecular Dirigida , Farmacocinética , Prostaglandina H2/antagonistas & inhibidores , Prostaglandina H2/metabolismo , Prostaglandina-E Sintasas , Tiobarbitúricos/metabolismo
5.
J Biol Chem ; 285(38): 29254-61, 2010 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-20605783

RESUMEN

Microsomal prostaglandin E synthase-1 (MPGES1) is induced during an inflammatory reaction from low basal levels by pro-inflammatory cytokines and subsequently involved in the production of the important mediator of inflammation, prostaglandin E(2). Nonsteroidal anti-inflammatory drugs prevent prostaglandin E(2) production by inhibiting the upstream enzymes cyclooxygenases 1 and 2. In contrast to these conventional drugs, a new generation of NSAIDs targets the terminal enzyme MPGES1. Some of these compounds potently inhibit human MPGES1 but do not have an effect on the rat orthologue. We investigated this interspecies difference in a rat/human chimeric form of the enzyme as well as in several mutants and identified key residues Thr-131, Leu-135, and Ala-138 in human MPGES1, which play a crucial role as gate keepers for the active site of MPGES1. These residues are situated in transmembrane helix 4, lining the entrance to the cleft between two subunits in the protein trimer, and regulate access of the inhibitor in the rat enzyme. Exchange toward the human residues in rat MPGES1 was accompanied with a gain of inhibitor activity, whereas exchange in human MPGES1 toward the residues found in rat abrogated inhibitor activity. Our data give evidence for the location of the active site at the interface between subunits in the homotrimeric enzyme and suggest a model of how the natural substrate PGH(2), or competitive inhibitors of MPGES1, enter the active site via the phospholipid bilayer of the membrane.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Immunoblotting , Concentración 50 Inhibidora , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Oxidorreductasas Intramoleculares/genética , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutagénesis Sitio-Dirigida , Prostaglandina-E Sintasas , Unión Proteica , Estructura Secundaria de Proteína , Ratas , Homología de Secuencia de Aminoácido
6.
Clin Pharmacokinet ; 41(11): 877-99, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12190333

RESUMEN

The clinical development of new drugs is often terminated because of unfavourable pharmacokinetic properties such as poor intestinal absorption after oral administration. Intestinal permeability and solubility are two of the most important factors that determine the absorption properties of a compound. Efficient and reliable computational models that predict these properties as early as possible in drug discovery and development are therefore desirable. In this review, we first discuss the implementation of predictive models of intestinal drug permeability and solubility in drug discovery and development. Secondly, we discuss the mechanisms of intestinal drug permeability and computational methods that can be used to predict it. We then discuss factors influencing drug solubility and models for predicting it. We finally speculate that once these and other predictive computational models are implemented in drug discovery and development, these processes will become much more effective. Further, an increased fraction of drug candidates that are less likely to fail during clinical development will be selected.


Asunto(s)
Diseño de Fármacos , Absorción Intestinal/fisiología , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Transporte Biológico , Humanos , Modelos Biológicos , Permeabilidad , Solubilidad , Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA