Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 59(15): 7167-76, 2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27391254

RESUMEN

Ligand-target residence time is emerging as a key drug discovery parameter because it can reliably predict drug efficacy in vivo. Experimental approaches to binding and unbinding kinetics are nowadays available, but we still lack reliable computational tools for predicting kinetics and residence time. Most attempts have been based on brute-force molecular dynamics (MD) simulations, which are CPU-demanding and not yet particularly accurate. We recently reported a new scaled-MD-based protocol, which showed potential for residence time prediction in drug discovery. Here, we further challenged our procedure's predictive ability by applying our methodology to a series of glucokinase activators that could be useful for treating type 2 diabetes mellitus. We combined scaled MD with experimental kinetics measurements and X-ray crystallography, promptly checking the protocol's reliability by directly comparing computational predictions and experimental measures. The good agreement highlights the potential of our scaled-MD-based approach as an innovative method for computationally estimating and predicting drug residence times.


Asunto(s)
Glucoquinasa/química , Simulación de Dinámica Molecular , Cristalografía por Rayos X , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Glucoquinasa/antagonistas & inhibidores , Glucoquinasa/metabolismo , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/metabolismo , Cinética , Ligandos , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Factores de Tiempo
2.
Br J Pharmacol ; 168(2): 339-53, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22925001

RESUMEN

BACKGROUND AND PURPOSE: Small-molecule glucokinase activators (GKAs) are currently being investigated as therapeutic options for the treatment of type 2 diabetes (T2D). Because liver overexpression of glucokinase is thought to be associated with altered lipid profiles, this study aimed at assessing the potential lipogenic risks linked to oral GKA administration. EXPERIMENTAL APPROACH: Nine GKA candidates were qualified for their ability to activate recombinant glucokinase and to stimulate glycogen synthesis in rat hepatocytes and insulin secretion in rat INS-1E cells. In vivo activity was monitored by plasma glucose and HbA1c measurements after oral administration in rodents. Risk-associated effects were assessed by measuring hepatic and plasma triglycerides and free fatty acids, as well as plasma aminotransferases, and alkaline phosphatase. KEY RESULTS: GKAs, while efficiently decreasing glycaemia in acute conditions and HbA1c levels after chronic administration in hyperglycemic db/db mice, were potent inducers of hepatic steatosis. This adverse outcome appeared as soon as 4 days after daily oral administration at pharmacological doses and was not transient. GKA treatment similarly increased hepatic triglycerides in diabetic and normoglycaemic rats, together with a pattern of metabolic phenotypes including different combinations of increased plasma triglycerides, free fatty acids, alanine and aspartyl aminotransferases, and alkaline phosphatase. GKAs belonging to three distinct structural families induced hepatic steatosis in db/db mice, arguing in favour of a target-mediated, rather than a chemical class-mediated, effect. CONCLUSION AND IMPLICATIONS: Given the risks associated with fatty liver disease in the general population and furthermore in patients with T2D, these findings represent a serious warning for the use of GKAs in humans. LINKED ARTICLE: This article is commented on by Rees and Gloyn, pp. 335-338 of this issue. To view this commentary visit http://dx.doi.org/10.1111/j.1476-5381.2012.02201.x.


Asunto(s)
Activadores de Enzimas/farmacología , Hígado Graso/inducido químicamente , Glucoquinasa/metabolismo , Hipoglucemiantes/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Animales , Glucemia/análisis , Células CACO-2 , Línea Celular Tumoral , Células Cultivadas , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Activadores de Enzimas/uso terapéutico , Hígado Graso/metabolismo , Hemoglobina Glucada/análisis , Hepatocitos/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Hipoglucemiantes/uso terapéutico , Absorción Intestinal , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Ratas Zucker
3.
Life Sci ; 89(7-8): 259-68, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21736880

RESUMEN

AIMS: We have developed biochemical and cell based assays to characterize small therapeutic molecules that inhibit the DNA damage checkpoint enzyme, Chk1 (Checkpoint kinase 1). MAIN METHODS: To prepare a screen of large chemical libraries, we purified the full-length and the catalytic domain versions of human Chk1. We characterized their properties and then selected full-length Chk1 as the variant most suitable for screening. We then identified and characterized structurally different Chk1 inhibitors in cell based-assays by measuring cytotoxicity and checkpoint bypass activity. KEY FINDINGS: We treated human cells with topoisomerase I inhibitors and demonstrated that at the time of Chk1 inhibitor addition, the cells have damaged DNA and activated Chk1. One Chk1 inhibitor, the indolocarbazole S27888, was active in the checkpoint bypass assay. SIGNIFICANCE: Knowing that the protein kinase inhibitory properties are different for each inhibitor, it seems that only a limited range of inhibitory activity is tolerated by cells. Chk1 has an essential role in determining how cancer cells respond to genotoxic treatments, therefore, inhibitors of this protein kinase are of great medical interest.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Carbazoles/farmacología , Neoplasias del Colon/tratamiento farmacológico , Proteínas Quinasas/metabolismo , Inhibidores de Topoisomerasa I/farmacología , Adenocarcinoma/enzimología , Adenocarcinoma/genética , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Neoplasias del Colon/enzimología , Neoplasias del Colon/genética , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Proteínas Quinasas/genética , Spodoptera/citología
4.
Cell Metab ; 13(4): 376-388, 2011 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-21459323

RESUMEN

AMPK has emerged as a critical mechanism for salutary effects of polyphenols on lipid metabolic disorders in type 1 and type 2 diabetes. Here we demonstrate that AMPK interacts with and directly phosphorylates sterol regulatory element binding proteins (SREBP-1c and -2). Ser372 phosphorylation of SREBP-1c by AMPK is necessary for inhibition of proteolytic processing and transcriptional activity of SREBP-1c in response to polyphenols and metformin. AMPK stimulates Ser372 phosphorylation, suppresses SREBP-1c cleavage and nuclear translocation, and represses SREBP-1c target gene expression in hepatocytes exposed to high glucose, leading to reduced lipogenesis and lipid accumulation. Hepatic activation of AMPK by the synthetic polyphenol S17834 protects against hepatic steatosis, hyperlipidemia, and accelerated atherosclerosis in diet-induced insulin-resistant LDL receptor-deficient mice in part through phosphorylation of SREBP-1c Ser372 and suppression of SREBP-1c- and -2-dependent lipogenesis. AMPK-dependent phosphorylation of SREBP may offer therapeutic strategies to combat insulin resistance, dyslipidemia, and atherosclerosis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aterosclerosis/tratamiento farmacológico , Hígado Graso/tratamiento farmacológico , Resistencia a la Insulina , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Animales , Benzopiranos/uso terapéutico , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Humanos , Lipogénesis , Masculino , Metformina/uso terapéutico , Ratones , Fosforilación , Receptores de LDL/genética , Receptores de LDL/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 2 de Unión a Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Transcripción Genética
5.
Thromb Res ; 124(4): 439-46, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19540572

RESUMEN

INTRODUCTION: Plasminogen Activator Inhibitor-1 (PAI-1) is the most potent endogenous inhibitor of fibrinolysis which is implicated in the pathogenesis of myocardial infarction and metabolic syndrome. The formation of reactive oxygen species (ROS) plays an important role in the pathology of vascular disorders and has been shown to increase PAI-1 expression by endothelial cells. Growing evidence indicates that NADPH oxidase and in particular the constitutively active Nox4-p22(phox) complexes are major sources of ROS in endothelial cells. The aim of the present study was to characterize the role of NADPH oxidase and in particular Nox4 in the regulation of PAI-1 expression in cultured Human Umbilical Venous Endothelial Cells (HUVECs). METHODS AND RESULTS: N-acetylcysteine (NAC, scavenger of ROS), diphenylene iodonium chloride (DPI, inhibitor of flavoproteins), M40403 (superoxyde dismutase mimic) and S17834 (inhibitor of NADPH oxidase) inhibited PAI-1 release and promoter activity in HUVECs. Specific knock down of Nox4 mRNA by siRNA caused a decrease in ROS production and NADPH oxidase activity. Moreover, Nox4 silencing decreased PAI-1 expression, release and activity as well as p38 MAPK pathways and NFkappaB activation. These signalling pathways are also involved in PAI-1 release. CONCLUSIONS: The NADPH oxidase inhibitors DPI and S 17834 as well as Nox4 silencing decreased PAI-1 synthesis in human cultured endothelial cells demonstrating the involvement of the constitutively active Nox4-containing NADPH oxidase in ROS-mediated PAI-1 transcription via p38 MAPK pathways. NADPH oxidase targeting with inhibitors such as S17834 could be an interesting strategy to decrease both oxidative stress and PAI-1 synthesis.


Asunto(s)
Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , NADPH Oxidasas/metabolismo , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Acetilcisteína/farmacología , Benzopiranos/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Humanos , Manganeso , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Compuestos Onio/farmacología , Compuestos Organometálicos/farmacología , Inhibidor 1 de Activador Plasminogénico/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/metabolismo
6.
J Biol Chem ; 283(29): 20015-26, 2008 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-18482975

RESUMEN

Resveratrol may protect against metabolic disease through activating SIRT1 deacetylase. Because we have recently defined AMPK activation as a key mechanism for the beneficial effects of polyphenols on hepatic lipid accumulation, hyperlipidemia, and atherosclerosis in type 1 diabetic mice, we hypothesize that polyphenol-activated SIRT1 acts upstream of AMPK signaling and hepatocellular lipid metabolism. Here we show that polyphenols, including resveratrol and the synthetic polyphenol S17834, increase SIRT1 deacetylase activity, LKB1 phosphorylation at Ser(428), and AMPK activity. Polyphenols substantially prevent the impairment in phosphorylation of AMPK and its downstream target, ACC (acetyl-CoA carboxylase), elevation in expression of FAS (fatty acid synthase), and lipid accumulation in human HepG2 hepatocytes exposed to high glucose. These effects of polyphenols are largely abolished by pharmacological and genetic inhibition of SIRT1, suggesting that the stimulation of AMPK and lipid-lowering effect of polyphenols depend on SIRT1 activity. Furthermore, adenoviral overexpression of SIRT1 stimulates the basal AMPK signaling in HepG2 cells and in the mouse liver. AMPK activation by SIRT1 also protects against FAS induction and lipid accumulation caused by high glucose. Moreover, LKB1, but not CaMKKbeta, is required for activation of AMPK by polyphenols and SIRT1. These findings suggest that SIRT1 functions as a novel upstream regulator for LKB1/AMPK signaling and plays an essential role in the regulation of hepatocyte lipid metabolism. Targeting SIRT1/LKB1/AMPK signaling by polyphenols may have potential therapeutic implications for dyslipidemia and accelerated atherosclerosis in diabetes and age-related diseases.


Asunto(s)
Hepatocitos/enzimología , Metabolismo de los Lípidos , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Sirtuinas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP , Animales , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Línea Celular , Activación Enzimática/efectos de los fármacos , Flavonoides/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación Enzimológica de la Expresión Génica , Glucosa/farmacología , Humanos , Ratones , Complejos Multienzimáticos/genética , Fenoles/farmacología , Polifenoles , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Sirtuina 1 , Sirtuinas/genética
7.
Diabetes ; 55(8): 2180-91, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16873680

RESUMEN

Because polyphenols may have beneficial effects on dyslipidemia, which accelerates atherosclerosis in diabetes, we examined the effect of polyphenols on hepatocellular AMP-activated protein kinase (AMPK) activity and lipid levels, as well as hyperlipidemia and atherogenesis in type 1 diabetic LDL receptor-deficient mice (DMLDLR(-/-)). In HepG2 hepatocytes, polyphenols, including resveratrol (a major polyphenol in red wine), apigenin, and S17834 (a synthetic polyphenol), increased phosphorylation of AMPK and its downstream target, acetyl-CoA carboxylase (ACC), and they increased activity of AMPK with 200 times the potency of metformin. The polyphenols also prevented the lipid accumulation that occurred in HepG2 cells exposed to high glucose, and their ability to do so was mimicked and abrogated, respectively, by overexpression of constitutively active and dominant-negative AMPK mutants. Furthermore, treatment of DMLDLR(-/-) mice with S17834 prevented the decrease in AMPK and ACC phosphorylation and the lipid accumulation in the liver, and it also inhibited hyperlipidemia and the acceleration of aortic lesion development. These studies 1) reveal that inactivation of hepatic AMPK is a key event in the pathogenesis of hyperlipidemia in diabetes, 2) point to a novel mechanism of action of polyphenols to lower lipids by activating AMPK, and 3) emphasize a new therapeutic avenue to benefit hyperlipidemia and atherosclerosis specifically in diabetes via activating AMPK.


Asunto(s)
Aterosclerosis/prevención & control , Diabetes Mellitus Experimental/complicaciones , Flavonoides/administración & dosificación , Lípidos/sangre , Complejos Multienzimáticos/metabolismo , Fenoles/administración & dosificación , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de LDL/deficiencia , Proteínas Quinasas Activadas por AMP , Acetil-CoA Carboxilasa/metabolismo , Animales , Apigenina/farmacología , Benzopiranos/administración & dosificación , Carcinoma Hepatocelular , Línea Celular Tumoral , Diabetes Mellitus Experimental/tratamiento farmacológico , Activación Enzimática/efectos de los fármacos , Glucosa/farmacología , Humanos , Hipolipemiantes/administración & dosificación , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Polifenoles , Receptores de LDL/fisiología , Resveratrol , Estilbenos/administración & dosificación
8.
Eur J Med Chem ; 41(3): 306-20, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16414149

RESUMEN

The third melatonin binding site, MT3 is a non-classical one since it is not a seven transmembrane domains receptor, but an enzyme, quinone reductase 2. A major concern for the study of the physiological role of this site is the lack of specific ligands, permitting to more accurately dissect the pathways linked to the activation of MT3. Indeed, in the course of finding new ligands, we identified a new series of compounds with affinity to the binding site in the nM range, particularly 2,3-dimethoxy 7-hydroxy 10-methyl 5H 10H indeno(1,2-b)indol-10-one (DMHMIO), with a Ki of 190 pM. Based on slightly different and novel synthons compared to most of the compounds used in melatonin pharmacology studies, these compounds offer new perspective for the description of the melatonin pathways, so much more by not having any affinity towards the MT1 and MT2 'classical' melatonin receptors.


Asunto(s)
Indenos/química , Indoles/química , Melatonina/metabolismo , Quinona Reductasas/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Cricetinae , Indenos/metabolismo , Indenos/farmacología , Indoles/metabolismo , Indoles/farmacología , Ligandos , Melatonina/química , Estructura Molecular , Nanotecnología , Quinona Reductasas/química , Quinona Reductasas/efectos de los fármacos , Receptores de Melatonina/química , Receptores de Melatonina/efectos de los fármacos , Receptores de Melatonina/metabolismo , Relación Estructura-Actividad
9.
Biochem Pharmacol ; 68(10): 1911-22, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15476662

RESUMEN

We have identified a novel series of indenoindole derivatives endowed with potent cytotoxic activities toward cancer cells. Five compounds containing a 8-[2-(dialkylamino)ethoxy]-2,3-dimethoxy-5H-10H-indeno[1,2-b]indol-10-one-O-propynyl-oxime core substituted with a phenyl, furanyl, or a methyl substituent on the propynyl side chain have been synthesized and their mechanism of action was investigated using a panel of complementary biophysical and biochemical techniques. The compounds were shown to intercalate into DNA with a preference for AT-rich sequences. They have no effect on topoisomerase I but they strongly stimulate DNA cleavage by topoisomerase II. Their capacity to stabilize topoisomerase II-DNA covalent complexes is comparable to that of the reference drug etoposide. The nature and orientation of the substituent on the propynyl chain modulate the DNA binding and topoisomerase II inhibitory properties of the compounds and, apparently, there is a correlation between the cytotoxic potential and the molecular action at the DNA-topoisomerase II level. The growth of human K562 leukemia cells is strongly reduced in the presence of the indenoindoles (IC(50) in the 50nM range) which maintain a high cytotoxic activity toward the adriamycin-resistant K562adr cells line in vitro. The low resistance indexes measured with the indenoindoles (RRI = 10-30) compared to adriamycin (RRI = 1000) suggest that our new compounds are weakly or not sensitive to drug efflux mediated by glycoprotein-P and/or multidrug resistance (MDR) protein pumps. Finally, we also show that these indenoindoles arrest K562 cells in the G2/M phase of the cell cycle and promote apoptosis, as indicated by the appearance of internucleosomal DNA cleavage. One compound in the series was tested for in vivo antitumor activity against the colon 38 model and at 25mg/kg it showed 100% complete tumor regression in the treated mice, without significant body weight loss. Altogether, the results reported here establish that our indenoindole derivatives represent a novel interesting series of DNA-targeted cytotoxic agents.


Asunto(s)
Antineoplásicos/farmacología , ADN/efectos de los fármacos , Indoles/farmacología , Inhibidores de Topoisomerasa II , Animales , Antineoplásicos/uso terapéutico , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , ADN/metabolismo , Modelos Animales de Enfermedad , Humanos , Indoles/uso terapéutico , Células K562 , Ratones , Trasplante de Neoplasias , Neoplasias Experimentales/tratamiento farmacológico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Arzneimittelforschung ; 53(11): 774-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14677373

RESUMEN

The general synthetic pathway and the interactions with arachidonic acid metabolism of the new compound S 19812 (N-hydroxy-N-methyl 4-(2,3-bis-(4-methoxyphenyl)-thiophen-5-yl)butanamide, CAS 181308-68-9) were investigated. S 19812 was selected for its well balanced dual inhibition of cyclooxygenase and 5-lipoxygenase pathways in vitro in rat polymorphonuclear neutrophils (PMNs) (IC50 PGE2: 0.10 mumol/l, LTB4: 0.07 mumol/l) and ex vivo in mice blood (ED50 PGE2: 13.1 mg/kg, LTB4: 20.8 mg/kg). These properties make this drug a promising therapeutic agent for pain and inflammation associated with osteoarthritis.


Asunto(s)
Amidas/síntesis química , Amidas/farmacología , Inhibidores de la Ciclooxigenasa/síntesis química , Inhibidores de la Ciclooxigenasa/farmacología , Inhibidores de la Lipooxigenasa/síntesis química , Inhibidores de la Lipooxigenasa/farmacología , Tiofenos/síntesis química , Tiofenos/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Calcimicina/farmacología , Ciclooxigenasa 1 , Dinoprostona/biosíntesis , Indicadores y Reactivos , Isoenzimas/metabolismo , Leucotrieno B4/biosíntesis , Masculino , Proteínas de la Membrana , Ratones , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ratas
11.
FEBS Lett ; 553(1-2): 157-62, 2003 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-14550565

RESUMEN

The binding profile of [(3)H]BHDP ([(3)H]N-benzyl-N'-(2-hydroxy-3,4-dimethoxybenzyl)-piperazine) was evaluated. [(3)H]BHDP labelled a single class of binding sites with high affinity (K(d)=2-3 nM) in rat liver mitochondria and synaptic membranes. The pharmacological characterization of these sites using sigma reference compounds revealed that these sites are sigma receptors and, more particularly, sigma1 receptors. Indeed, BHDP inhibited [(3)H]pentazocine binding, a marker for sigma1 receptors, with high affinity in a competitive manner. BHDP is selective for sigma1 receptors since it did not show any relevant affinity for most of the other receptors, ion channels or transporters tested. Moreover, in an in vitro model of cellular hypoxia, BHDP prevented the fall in adenosine triphosphate (ATP) levels caused by 24 h hypoxia in cultured astrocytes. Taken together, these results demonstrate that [(3)H]BHDP is a potent and selective ligand for sigma1 receptors showing cytoprotective effects in astrocytes.


Asunto(s)
Encéfalo/metabolismo , Mitocondrias Hepáticas/metabolismo , Piperazinas/metabolismo , Receptores sigma/metabolismo , Sinaptosomas/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/citología , Células Cultivadas , Concentración de Iones de Hidrógeno , Hipoxia/metabolismo , Ligandos , Piperazinas/farmacología , Unión Proteica , Ratas , Especificidad por Sustrato , Factores de Tiempo , Tritio/metabolismo
12.
Arzneimittelforschung ; 53(12): 844-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14732965

RESUMEN

S 19812 (N-hydroxy-N-methyl-4-(2,3-bis-(4-methoxyphenyl)-thiophen-5-yl) butanamide, CAS 181308-68-9), a dual inhibitor of cyclooxygenase and lipoxygenase pathways, was evaluated in different models of pain and inflammation. Its gastric tolerance was also investigated. After acute oral treatment S 19812 exhibited a non-opioid analgesic activity observed in the phenylbenzoquinone-induced writhing model in mice (ED50 = 2.1 mg/kg) and in the carrageenan-induced hyperalgesia model in rats (ED50 = 9.1 mg/kg, preventive treatment; 8.3 mg/kg, curative treatment). Anti-inflammatory activity was observed in the adjuvant-induced arthritis in rat (inhibition of edema ED50 = 11 mg/kg/day p.o., day 28). In rats and mice, S 19812 exhibited an excellent gastric tolerance at doses up to 800 mg/kg p.o.


Asunto(s)
Amidas/farmacología , Analgésicos , Inhibidores de la Ciclooxigenasa/farmacología , Inhibidores de la Lipooxigenasa/farmacología , Tiofenos/farmacología , Ácido Acético , Aire , Amidas/toxicidad , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/toxicidad , Artritis Experimental/tratamiento farmacológico , Carragenina , Inhibidores de la Ciclooxigenasa/toxicidad , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Calor , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Inhibidores de la Lipooxigenasa/toxicidad , Masculino , Ratones , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Gastropatías/inducido químicamente , Gastropatías/patología , Tiofenos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA