Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Xenobiotica ; 46(10): 868-74, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26796435

RESUMEN

1. The metabolism of dydrogesterone was investigated in human liver cytosol (HLC) and human liver microsomes (HLM). Enzymes involved in dydrogesterone metabolism were identified and their relative contributions were estimated. 2. Dydrogesterone clearance was clearly higher in HLC compared to HLM. The major active metabolite 20α-dihydrodydrogesterone (20α-DHD) was only produced in HLC. 3. The formation of 20α-DHD by cytosolic aldo-keto reductase 1C (AKR1C) was confirmed with isoenzyme-specific AKR inhibitors. 4. Using recombinantly expressed human cytochrome P450 (CYP) isoenzymes, dydrogesterone was shown to be metabolically transformed by CYP3A4 and CYP2C19. 5. A clear contribution of CYP3A4 to microsomal metabolism of dydrogesterone was demonstrated with HLM and isoenzyme-specific CYP inhibitors, and confirmed by a significant correlation between dydrogesterone clearance and CYP3A4 activity. 6. Contribution of CYP2C19 was shown to be clearly less than CYP3A4 and restricted to a small group of human individuals with very high CYP2C19 activity. Therefore, it is expected that CYP2C19 genetic variations will not affect dydrogesterone pharmacokinetics in man. 7. In conclusion, dydrogesterone metabolism in the liver is dominated primarily by cytosolic enzymes (particularly AKR1C) and secondarily by CYP3A4, with the former exclusively responsible for 20α-DHD formation.


Asunto(s)
Aldehído Reductasa/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Didrogesterona/metabolismo , Progestinas/metabolismo , Aldo-Ceto Reductasas , Humanos
2.
Int J Clin Pharmacol Ther ; 51(12): 976-85, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24120718

RESUMEN

OBJECTIVES: This study evaluated interaction potential between sugammadex and aspirin on platelet aggregation. METHODS: This was a randomized, double-blind, placebo-controlled, four-period crossover study in 26 healthy adult males. Treatments were i.v. placebo, i.v. sugammadex 4 mg/kg, and i.v. placebo/sugammadex with oncedaily oral aspirin 75 mg. Primary objective was to assess interaction between sugammadex and aspirin on platelet aggregation using collagen-induced whole-blood aggregometry. Effects on activated partial thromboplastin time (APTT) and cutaneous bleeding time were also evaluated. Platelet aggregation and APTT were evaluated by geometric mean ratios, using area-under-effect curves 3 - 30 minutes after sugammadex/placebo dosing. Bleeding time ratio was evaluated at 5 minutes post-dosing. Non-inferiority margins were pre-specified via literature review. Type I error was controlled using a hierarchical strategy. RESULTS: Ratio for platelet aggregation for aspirin with sugammadex vs. aspirin alone was 1.01, with lower limit of two-sided 90% CI of 0.91(above non-inferiority margin of 0.75). Ratio for statistical interaction between sugammadex and aspirin on APTT was 1.01, with upper 90% CI of 1.04 (below non-inferiority margin of 1.50), and for sugammadex vs. placebo alone was 1.06, with an upper 90% CI of 1.07 (below non-inferiority margin of 1.50). Ratio for bleeding time for aspirin with sugammadex vs. aspirin plus placebo was 1.20, with upper 90% CI of 1.45 (below non-inferiority margin of 1.50). Sugammadex was generally well tolerated. CONCLUSION: There was no clinically relevant reduction in platelet aggregation with addition of sugammadex 4 mg/kg to aspirin. Pre-determined non-inferiority margins were not exceeded for bleeding time and APTT.


Asunto(s)
Aspirina/farmacología , Coagulación Sanguínea/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , gamma-Ciclodextrinas/farmacología , Adolescente , Adulto , Estudios Cruzados , Método Doble Ciego , Interacciones Farmacológicas , Humanos , Masculino , Persona de Mediana Edad , Tiempo de Tromboplastina Parcial , Sugammadex
3.
J Clin Pharmacol ; 54(9): 1038-47, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24691799

RESUMEN

Simvastatin and fenofibrate are frequently co-prescribed at staggered intervals for the treatment of dyslipidemia. Since a drug-drug interaction has been reported when the two drugs are given simultaneously, it is of clinical interest to know whether the interaction differs between simultaneous and staggered combinations. A study, assessing the impact of both combinations on the interaction, was conducted with 7-day treatment regimens using simvastatin 40 mg and fenofibrate 145 mg: (A) simvastatin only (evening), (B) simvastatin and fenofibrate (both in evening), and (C) simvastatin (evening) and fenofibrate (morning). Eighty-five healthy subjects received the respective treatments in a randomized, 3-way cross-over study. The pharmacokinetics of simvastatin and the active metabolite simvastatin acid were determined. There was a limited reduction in the AUC0-24h of simvastatin acid of 21 and 29% for simultaneous and staggered combination, respectively. The geometric mean AUC0-24h ratio of simvastatin acid for the two combined dosing regimens (B/C) and 90% confidence interval were 111% (102-121). The interaction apparently had no impact on lipid markers. The findings imply that the observed pharmacokinetic interaction is unlikely clinically relevant, and support the combined use of simvastatin and fenofibrate not only given at staggered interval but also given simultaneously.


Asunto(s)
Fenofibrato/administración & dosificación , Fenofibrato/farmacocinética , Hipolipemiantes/administración & dosificación , Hipolipemiantes/farmacocinética , Simvastatina/administración & dosificación , Simvastatina/farmacocinética , Colesterol/sangre , Estudios Cruzados , Esquema de Medicación , Interacciones Farmacológicas , Quimioterapia Combinada , Femenino , Fenofibrato/sangre , Humanos , Hipolipemiantes/sangre , Transportador 1 de Anión Orgánico Específico del Hígado , Masculino , Transportadores de Anión Orgánico/genética , Polimorfismo de Nucleótido Simple , Simvastatina/sangre , Triglicéridos/sangre
4.
Int Immunopharmacol ; 9(10): 1218-27, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19616132

RESUMEN

Molecular and cellular assessment of dermal delayed-type hypersensitivity (DTH) responses is a useful approach for evaluating the mechanism of action (MOA) of immunomodulatory agents. In the present report, we characterized the delayed-type hypersensitivity response induced by keyhole limpet hemocyanin (KLH), and validated its utility by evaluating an immunomodulator, BIRB-796. Intradermal KLH challenge of the ear pinna following subcutaneous antigen sensitization resulted in a pronounced skin inflammation that peaked at 24-48h. At the molecular level, there was an activation of 3 mitogen-activated protein kinases (MAPKs: p38, JNK and ERK), an induction of the chemokines CCL2/JE, CXCL2/Mip-2, CXCL1/KC, CCL3/Mip-1alpha CCL4/Mip-1beta and CXCL10/IP-10, and expression of the cytokines IL-1beta and IL-10 in the ear parenchyma. Modulation of TNFalpha protein level was only detected in ex-vivo ear whole organ cultures (EWOC). Consistent with this inflammatory profile there was an infiltration of neutrophils and mononuclear cells into the ear parenchyma. BIRB-796, a potent allosteric p38 MAPK inhibitor attenuated the ear swelling response, which correlated with a reduced inflammatory profile. BIRB-796 inhibited p38 but not JNK or ERK kinase activation, decreased multiple chemokines which correlated with a decrease in the infiltration of neutrophils and macrophages; CD4 T cells were modesty reduced. Similarly, there was a decrease of levels of cytokines including IL-1beta, IL-10 and TNFalpha. These data support the utility of this model for evaluating immunomodulators on skin inflammation and suggest that modulation of p38 kinase may be of therapeutic value for the treatment of inflammatory skin conditions.


Asunto(s)
Hemocianinas/inmunología , Hipersensibilidad Tardía/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Oído/patología , Femenino , Hipersensibilidad Tardía/patología , Hipersensibilidad Tardía/fisiopatología , Inmunización , Inyecciones Intradérmicas , Ratones , Ratones Endogámicos BALB C , Naftalenos/administración & dosificación , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Técnicas de Cultivo de Órganos , Otitis , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirazoles/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
5.
Biol Pharm Bull ; 30(7): 1231-6, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17603159

RESUMEN

The CXC chemokine receptor CXCR2 has been implicated in the pathogenesis of several chronic diseases including atherosclerosis. To enable animal studies towards understanding the role of human CXCR2 (hCXCR2) in disease development, we previously generated hCXCR2 knockin (hCXCR2(+/+)) mice. We have demonstrated that the phenotype and the acute immune response of the hCXCR2(+/+) mice was identical to that of wild-type mice, indicating that hCXCR2 indeed takes over the function of endogenous mouse CXCR2 (mCXCR2). In the present paper, we extend these findings by studying whether hCXCR2 functionally replaces the role of mCXCR2 in a chronic disease model for atherosclerosis. We first defined which of two well-described atherosclerosis models (ApoE(-/-) or LDLR(-/-) mice) is most suited for this purpose. When expression of mCXCR2 and that of its ligands in atherosclerotic lesions were compared in these mice, increased expression levels were observed only in LDLR(-/-) mice. Further, cultured atherosclerotic aortas from LDLR(-/-) mice did secrete significantly higher levels of CXCR2 ligands compared to aortas from healthy controls. Since these results support the role of CXCR2 in the atherogenesis in the LDLR(-/-) mice, double mutant hCXCR2(+/+)/LDLR(-/-) mice were generated and diet-induced atherosclerosis in these mice was compared to that in LDLR(-/-) mice. Upon an atherogenic diet, the hCXCR2(+/+)/LDLR(-/-) mice developed plaque lesions in a similar manner to those in LDLR(-/-) mice, indicating successful functional replacement of mCXCR2 by hCXCR2 in this disease model. We conclude that hCXCR2(+/+)/LDLR(-/-) mice present an attractive model to study the role of hCXCR2 in atherosclerosis development and for future testing of novel pharmaceuticals designed to antagonize hCXCR2.


Asunto(s)
Aterosclerosis/etiología , Receptores de Interleucina-8B/fisiología , Receptores de LDL/fisiología , Animales , Apolipoproteínas E/fisiología , Masculino , Ratones , Ratones Noqueados
6.
Eur J Immunol ; 35(9): 2573-82, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16094689

RESUMEN

Human CXCR2 (hCXCR2) has been implicated in diverse inflammatory diseases. When roles of this receptor studied in animal models are extrapolated into men, large species differences in expression of the receptor and its ligands must be considered. These differences seriously weaken conclusions toward the role of hCXCR2 in the development of human diseases. It furthermore hampers straightforward testing of CXCR2 antagonists, especially when compounds discriminate between human and other species' receptors. Using gene targeting in embryonic stem cells, a hCXCR2 knockin mouse strain was generated in which endogenous murine CXCR2 (mCXCR2) sequences are replaced by the hCXCR2 gene. Correct targeting and expression on neutrophils were confirmed by Southern blot and immunohistochemical analyses. A phenotypic analysis of the hCXCR2 knockin mice, in comparison to wild-type and CXCR2 knockout mice, confirmed proper function of the hCXCR2 gene. In vivo migratory responses of neutrophils were intact in hCXCR2 knockin mice. Finally, an experiment with a CXCR2 antagonist demonstrated that the knockin model is indeed useful for in vivo evaluation of low-molecular weight compounds. In conclusion, our data unequivocally show that hCXCR2 can functionally replace mCXCR2, making this an attractive model to test novel pharmaceuticals designed to antagonize human CXCR2 in vivo.


Asunto(s)
Peritonitis/inmunología , Receptores de Interleucina-8B/inmunología , Animales , Calcio/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Quimiocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Marcación de Gen , Humanos , Inflamación/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neutrófilos/citología , Neutrófilos/inmunología , Peritonitis/inducido químicamente , Receptores de Interleucina-8B/antagonistas & inhibidores , Receptores de Interleucina-8B/genética , Células Madre , Tioglicolatos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA