Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 19(1): 268, 2022 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-36333747

RESUMEN

The pathophysiology of sepsis may involve the activation of the NOD-type receptor containing the pyrin-3 domain (NLPR-3), mitochondrial and oxidative damages. One of the primary essential oxidation products is 8-oxoguanine (8-oxoG), and its accumulation in mitochondrial DNA (mtDNA) induces cell dysfunction and death, leading to the hypothesis that mtDNA integrity is crucial for maintaining neuronal function during sepsis. In sepsis, the modulation of NLRP-3 activation is critical, and mefenamic acid (MFA) is a potent drug that can reduce inflammasome activity, attenuating the acute cerebral inflammatory process. Thus, this study aimed to evaluate the administration of MFA and its implications for the reduction of inflammatory parameters and mitochondrial damage in animals submitted to polymicrobial sepsis. To test our hypothesis, adult male Wistar rats were submitted to the cecal ligation and perforation (CLP) model for sepsis induction and after receiving an injection of MFA (doses of 10, 30, and 50 mg/kg) or sterile saline (1 mL/kg). At 24 h after sepsis induction, the frontal cortex and hippocampus were dissected to analyze the levels of TNF-α, IL-1ß, and IL-18; oxidative damage (thiobarbituric acid reactive substances (TBARS), carbonyl, and DCF-DA (oxidative parameters); protein expression (mitochondrial transcription factor A (TFAM), NLRP-3, 8-oxoG; Bax, Bcl-2 and (ionized calcium-binding adaptor molecule 1 (IBA-1)); and the activity of mitochondrial respiratory chain complexes. It was observed that the septic group in both structures studied showed an increase in proinflammatory cytokines mediated by increased activity in NLRP-3, with more significant oxidative damage and higher production of reactive oxygen species (ROS) by mitochondria. Damage to mtDNA it was also observed with an increase in 8-oxoG levels and lower levels of TFAM and NGF-1. In addition, this group had an increase in pro-apoptotic proteins and IBA-1 positive cells. However, MFA at doses of 30 and 50 mg/kg decreased inflammasome activity, reduced levels of cytokines and oxidative damage, increased bioenergetic efficacy and reduced production of ROS and 8-oxoG, and increased levels of TFAM, NGF-1, Bcl-2, reducing microglial activation. As a result, it is suggested that MFA induces protection in the central nervous system early after the onset of sepsis.


Asunto(s)
Ácido Mefenámico , Sepsis , Animales , Ratas , Masculino , Especies Reactivas de Oxígeno/metabolismo , Ácido Mefenámico/metabolismo , Ácido Mefenámico/farmacología , Ratas Wistar , Inflamasomas/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Mitocondrias , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , ADN Mitocondrial , Citocinas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
2.
J Assoc Physicians India ; 70(3): 11-12, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35438288

RESUMEN

Nucleotide-binding oligomerization domain like receptors (NLRs) -intracellular proteins, are a recently discovered class of innate immune receptors that play a crucial role in initiating the inflammatory response following pathogen recognition. The dysregulation of NLRP3 inflammasome can cause uncontrolled inflammation and drive the development of a wide variety of human diseases. Mefenamic acid which belongs to fenamate group inhibits the NLRP3 inflammasome by inhibiting efflux of chloride ions and influx of calcium ions through blocking VRAC and TRPM2 respectively. Thus, Mefenamic acid provides a potentially practical pharmacological approach for treating NLRP3- driven diseases.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Humanos , Inflamasomas/metabolismo , Inflamación/metabolismo , Iones/metabolismo , Macrófagos/metabolismo , Ácido Mefenámico/metabolismo , Ácido Mefenámico/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
3.
J Headache Pain ; 23(1): 36, 2022 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-35282834

RESUMEN

BACKGROUND/AIM: Certain constituents in migraine food triggers and non-steroidal anti-inflammatory drugs (NSAIDs) inhibit sulfotransferases (SULTs) that detoxify drugs/chemicals and play role in the metabolism of neurotransmitters. We aimed to dissect SULT1A1 modulation of CSD susceptibility and behavior in an in vivo experimental model using hesperidin, a SULT1A1 inhibitor found in citrus fruits (known migraine triggers) and mefenamic acid (SULT1A1 inhibitor), an NSAID to simulate medication overuse. METHODS: Hesperidin was used as SULT1A1 inhibitor found in citrus fruits, known migraine triggers and mefenamic acid (NSAID), another SULT1A1 inhibitor, was used to induce MO in rats. The groups were; 1) Hesperidin (ip) or its vehicle-DMSO (ip) 2) Chronic (4 weeks) mefenamic acid (ip) or its vehicle (ip) 3) Chronic mefenamic acid+hesperidin (ip) or DMSO (ip). CSD susceptibility was evaluated and behavioral testing was performed. SULT1A1 enzyme activity was measured in brain samples. RESULTS: Single-dose of hesperidin neither changed CSD susceptibility nor resulted in any behavioral change. Chronic mefenamic acid exposure resulted in increased CSD susceptibility, mechanical-thermal hypersensitivity, increased head shake, grooming and freezing and decreased locomotion. Single dose hesperidin administration after chronic mefenamic acid exposure resulted in increased CSD susceptibility and mechanical-thermal hypersensitivity, increased freezing and decreased locomotion. SULT1A1 enzyme activity was lower in mefenamic acid and mefenamic acid+hesperidin groups compared to their vehicles. CONCLUSION: Mefenamic acid and hesperidin have synergistic effect in modulating CSD susceptibility and pain behavior. Sulfotransferase inhibition may be the common mechanism by which food triggers and NSAIDs modulate migraine susceptibility. Further investigations regarding human provocation studies using hesperidin in migraine patients with medication overuse are needed.


Asunto(s)
Ácido Mefenámico , Trastornos Migrañosos , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Humanos , Ácido Mefenámico/metabolismo , Ácido Mefenámico/farmacología , Ácido Mefenámico/uso terapéutico , Trastornos Migrañosos/inducido químicamente , Trastornos Migrañosos/tratamiento farmacológico , Uso Excesivo de Medicamentos Recetados , Ratas , Sulfotransferasas/uso terapéutico
4.
Toxicology ; 448: 152648, 2021 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-33259822

RESUMEN

Mefenamic acid (MFA), one of the nonsteroidal anti-inflammatory drugs (NSAIDs), sometimes causes liver injury. Quinoneimines formed by cytochrome P450 (CYP)-mediated oxidation of MFA are considered to be causal metabolites of the toxicity and are detoxified by glutathione conjugation. A previous study reported that NAD(P)H:quinone oxidoreductase 1 (NQO1) can reduce the quinoneimines, but NQO1 is scarcely expressed in the human liver. The purpose is to identify enzyme(s) responsible for the decrease in MFA-quinoneimine formation in the human liver. The formation of MFA-quinoneimine by recombinant CYP1A2 and CYP2C9 was significantly decreased by the addition of human liver cytosol, and the extent of the decrease in the metabolite formed by CYP1A2 was larger than that by CYP2C9. By column chromatography, superoxide dismutase 1 (SOD1) was identified from the human liver cytosol as an enzyme decreasing MFA-quinoneimine formation. Addition of recombinant SOD1 into the reaction mixture decreased the formation of MFA-quinoneimine from MFA by recombinant CYP1A2. By a structure-activity relationship study, we found that SOD1 decreased the formation of quinoneimines from flufenamic acid and tolfenamic acid, but did not affect those produced from acetaminophen, amodiaquine, diclofenac, and lapatinib. Thus, SOD1 may selectively decrease the quinoneimine formation from fenamate-class NSAIDs. To examine whether SOD1 can attenuate cytotoxicity caused by MFA, siRNA for SOD1 was transfected into CYP1A2-overexpressed HepG2 cells. The leakage of lactate dehydrogenase caused by MFA treatment was significantly increased by knockdown of SOD1. In conclusion, we found that SOD1 can serve as a detoxification enzyme for quinoneimines to protect from drug-induced toxicity.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Ácido Mefenámico/metabolismo , Quinonas/metabolismo , Superóxido Dismutasa-1/biosíntesis , Adulto , Anciano , Femenino , Células Hep G2 , Humanos , Masculino , Ácido Mefenámico/antagonistas & inhibidores , Microsomas Hepáticos/metabolismo , Persona de Mediana Edad , Quinonas/antagonistas & inhibidores , Superóxido Dismutasa-1/antagonistas & inhibidores , Superóxido Dismutasa-1/deficiencia
5.
CPT Pharmacometrics Syst Pharmacol ; 10(2): 127-136, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33314761

RESUMEN

The sodium-glucose cotransporter 2 inhibitor ertugliflozin is metabolized by the uridine 5'-diphospho-glucuronosyltransferase (UGT) isozymes UGT1A9 and UGT2B4/2B7. This analysis evaluated the drug-drug interaction (DDI) following co-administration of ertugliflozin with the UGT inhibitor mefenamic acid (MFA) using physiologically-based pharmacokinetic (PBPK) modeling. The ertugliflozin modeling assumptions and parameters were verified using clinical data from single-dose and multiple-dose studies of ertugliflozin in healthy volunteers, and the PBPK fraction metabolized assignments were consistent with human absorption, distribution, metabolism, and excretion results. The model for MFA was developed using clinical data, and in vivo UGT inhibitory constant values were estimated using the results from a clinical DDI study with MFA and dapagliflozin, a UGT1A9 and UGT2B4/2B7 substrate in the same chemical class as ertugliflozin. Using the verified compound files, PBPK modeling predicted an ertugliflozin ratio of area under the plasma concentration-time curves (AUCR ) of 1.51 when co-administered with MFA. ClinicalTrials.gov identifier: NCT00989079.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Inhibidores de la Ciclooxigenasa/farmacocinética , Glucuronosiltransferasa/metabolismo , Ácido Mefenámico/farmacocinética , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacocinética , Adulto , Área Bajo la Curva , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Compuestos Bicíclicos Heterocíclicos con Puentes/metabolismo , Inhibidores de la Ciclooxigenasa/administración & dosificación , Inhibidores de la Ciclooxigenasa/metabolismo , Interacciones Farmacológicas , Femenino , Voluntarios Sanos , Humanos , Masculino , Ácido Mefenámico/administración & dosificación , Ácido Mefenámico/metabolismo , Persona de Mediana Edad , Modelos Biológicos , Inhibidores del Cotransportador de Sodio-Glucosa 2/administración & dosificación , Inhibidores del Cotransportador de Sodio-Glucosa 2/metabolismo , UDP Glucuronosiltransferasa 1A9 , Uridina/metabolismo
6.
Bioorg Med Chem Lett ; 30(10): 127112, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32209292

RESUMEN

An improved and rapid synthesis of mefenamic acid based indole derivatives has been achieved via the ligand free Cu-catalyzed coupling-cyclization method under ultrasound irradiation. This simple, straightforward and inexpensive one-pot method involved the reaction of a terminal alkyne derived from mefenamic acid with 2-iodosulfanilides in the presence of CuI and K2CO3 in PEG-400. The reaction proceeded via an initial CC bond formation (the coupling step) followed by CN bond formation (the intramolecular cyclization) to afford the mefenamic acid based indole derivatives in good to acceptable yields. Several of these compounds showed inhibition of PDE4 in vitro and the SAR (Structure Activity Relationship) within the series is discussed. The compound 3d has been identified as a promising and selective inhibitor of PDE4B (IC50 = 1.34 ± 0.46 µM) that showed TNF-α inhibition in vitro (IC50 = 5.81 ± 0.24 µM) and acceptable stability in the rat liver microsomes.


Asunto(s)
Cobre/química , Indoles/química , Ácido Mefenámico/química , Sonicación , Sitios de Unión , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Catálisis , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Ciclización , Semivida , Humanos , Indoles/metabolismo , Indoles/farmacología , Ácido Mefenámico/metabolismo , Ácido Mefenámico/farmacología , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa 4/química , Inhibidores de Fosfodiesterasa 4/metabolismo , Inhibidores de Fosfodiesterasa 4/farmacología , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
7.
Chembiochem ; 21(10): 1461-1472, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31919943

RESUMEN

The regio- (and stereo-)selectivity and specific activity of cytochrome P450s are determined by the accessibility of potential sites of metabolism (SOMs) of the bound substrate relative to the heme, and the activation barrier of the regioselective oxidation reaction(s). The accessibility of potential SOMs depends on the relative binding free energy (ΔΔGbind ) of the catalytically active substrate-binding poses, and the probability of the substrate to adopt a transition-state geometry. An established experimental method to measure activation energies of enzymatic reactions is the analysis of reaction rate constants at different temperatures and the construction of Arrhenius plots. This is a challenge for multistep P450-catalyzed processes that involve redox partners. We introduce a modified Arrhenius approach to overcome the limitations in studying P450 selectivity, which can be applied in multiproduct enzyme catalysis. Our approach gives combined information on relative activation energies, ΔΔGbind values, and collision entropies, yielding direct insight into the basis of selectivity in substrate conversion.


Asunto(s)
Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/metabolismo , Hemo/metabolismo , Ácido Mefenámico/metabolismo , Sitios de Unión , Catálisis , Hidroxilación , Simulación de Dinámica Molecular , Oxidación-Reducción , Unión Proteica , Especificidad por Sustrato , Termodinámica
8.
Neuropharmacology ; 160: 107795, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31560908

RESUMEN

Fenamates mefanamic and niflumic acids (MFA and NFA) induced dual potentiating and inhibitory effects on GABA currents recorded in isolated cerebellar Purkinje cells using the whole-cell patch-clamp and fast-application techniques. Regardless of the concentration, both drugs induced a pronounced prolongation of the current response. We demonstrated that the same concentration of drugs can produce both potentiating and inhibitory effects, depending on the GABA concentration, which indicates that both processes take place simultaneously and the net effect depends on the concentrations of both the agonist and fenamate. We found that the NFA-induced block is strongly voltage-dependent. The Woodhull analysis of the block suggests that NFA has two binding sites in the pore - shallow and deep. We built a homology model of the open GABAAR based on the cryo-EM structure of the open α1 GlyR and applied Monte-Carlo energy minimization to optimize the ligand-receptor complexes. A systematic search for MFA/NFA binding sites in the GABAAR pore revealed the existence of two sites, the location of which coincides well with predictions of the Woodhull model. In silico docking suggests that two fenamate molecules are necessary to occlude the pore. We showed that MFA, acting as a PAM, competes with an intravenous anesthetic etomidate for a common binding site. We built structural models of MFA and NFA binding at the transmembrane ß(+)/α(-) intersubunit interface. We suggested a hypothesis on the molecular mechanism underlying the prolongation of the receptor lifetime in open state after MFA/NFA binding and ß subunit specificity of the fenamate potentiation.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antagonistas de Receptores de GABA-A/farmacología , Ácido Mefenámico/farmacología , Ácido Niflúmico/farmacología , Receptores de GABA-A/metabolismo , Anestésicos Intravenosos/farmacología , Animales , Antiinflamatorios no Esteroideos/metabolismo , Sitios de Unión/efectos de los fármacos , Células Cultivadas , Sinergismo Farmacológico , Etomidato/farmacología , Antagonistas de Receptores de GABA-A/metabolismo , Ácido Mefenámico/metabolismo , Ácido Niflúmico/metabolismo , Células de Purkinje/efectos de los fármacos , Ratas
9.
Drug Dev Ind Pharm ; 45(10): 1610-1616, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31311329

RESUMEN

Mefenamic acid (MA) has been reported as a weakly soluble drug which presents weak in vivo absorption upon oral administration using conventional formulations. Solid dispersions (SDs) have been investigated extensively in literature for enhancing the solubility and bioavailability of weakly-soluble molecules. Hence, the aim of proposed study was to prepare MA novel formulations in the form of SDs using hot-melt extrusion technology in order to enhance its palatability, bioavailability, and pharmacodynamics effects/anti-inflammatory efficacy. Various SDs of MA were prepared using hot-melt extrusion technology, characterized physically and investigated for dissolution tests. Optimized SD formulations of MA were being subjected to palatability, pharmacodynamics, and pharmacokinetic studies in rats. Optimized SD of MA showed significant rat palatability tastes as compared with pure and marketed MA (p < .05). Anti-inflammatory efficacy of 20% SD and 25% SD of MA was found to be 86.44 and 89.83%, respectively, in comparison with 74.57 and 78.24% by pure MA and marketed MA, respectively. The anti-inflammatory efficacy of optimized SD was found to be significant as compared with pure and marketed MA (p < .05). The oral absorption of MA from optimized 20% SD was also noted as statistically significant as compared with pure MA (p < .05). The relative bioavailability of MA from 20 and 25% SDs was 2.97 and 2.24-folds higher than pure MA. The results of this study suggested that SDs prepared using hot-melt extrusion technology are capable to enhance palatability, anti-inflammatory efficacy, and oral bioavailability of MA in comparison with pure drug.


Asunto(s)
Ácido Mefenámico/química , Ácido Mefenámico/farmacología , Animales , Antiinflamatorios/química , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Disponibilidad Biológica , Rastreo Diferencial de Calorimetría/métodos , Química Farmacéutica/métodos , Portadores de Fármacos/química , Composición de Medicamentos/métodos , Femenino , Tecnología de Extrusión de Fusión en Caliente/métodos , Inflamación/tratamiento farmacológico , Ácido Mefenámico/metabolismo , Ratas , Ratas Wistar , Solubilidad/efectos de los fármacos
10.
J Appl Microbiol ; 127(3): 724-738, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31173436

RESUMEN

AIMS: We aimed to expand the microbial biocatalyst platform to generate essential oxyfunctionalized standards for pharmaceutical, toxicological and environmental research. In particular, we examined the production of oxyfunctionalized nonsteroidal anti-inflammatory drugs (NSAIDs) by filamentous-fungi. METHODS AND RESULTS: Four NSAIDs; diclofenac, ibuprofen, naproxen and mefenamic acid were used as substrates for oxyfunctionalization in a biocatalytic process involving three filamentous-fungi strains; Beauveria bassiana, Clitocybe nebularis and Mucor hiemalis. Oxyfunctionalized metabolites that are major degradation intermediates formed by Cytochrome P450 monooxygenases in human metabolism were produced in isolated yields of up to 99% using 1 g l-1 of substrate. In addition, a novel compound, 3',4'-dihydroxydiclofenac, was produced by B. bassiana. Proteomic analysis identified CYP548A5 that might be responsible for diclofenac oxyfunctionalization in B. bassiana. CONCLUSIONS: Efficient fungi catalysed oxyfunctionalization was achieved when using NSAIDs as substrates. High purities and isolated yields of the produced metabolites were achieved. SIGNIFICANCE AND IMPACT OF THE STUDY: The lack of current efficient synthetic strategies for oxyfunctionalization of NSAIDs is a bottleneck to perform pharmacokinetic, pharmacodynamic and toxicological analysis for the pharmaceutical industry. Additionally, oxyfunctionalized derivatives are needed for tracking the fate and impact of such metabolites in the environment. Herein, we described a fungi catalysed process that surpasses previously reported strategies in terms of efficiency, to synthesize oxyfunctionalized NSAIDs.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Hongos/metabolismo , Basidiomycota/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Diclofenaco/metabolismo , Ibuprofeno/metabolismo , Ácido Mefenámico/metabolismo , Mucor/metabolismo , Naproxeno/metabolismo , Proteómica
11.
J Inorg Biochem ; 194: 97-113, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30849593

RESUMEN

Copper(II) complexes containing non-steroidal anti-inflammatory drugs (NSAIDs) have been the subject of many research papers and reviews. Here we report the synthesis, spectroscopic study and biological activity of novel mixed copper(II) complexes with NSAIDs: tolfenamic (tolf), mefenamic (mef) and flufenamic (fluf) acids and phenanthroline (phen): [Cu(tolf-O,O')2(phen)] (1), [Cu(mef-O,O')2(phen)] (2), [Cu(fluf-O,O')2(phen)] (3). Complexes were characterized by X-ray analysis and EPR spectroscopy. Complexes 1-3 are monomeric, six-coordinate and crystallize in a monoclinic space group. Interaction of Cu(II) complexes with DNA was studied by means of absorption titrations, viscosity measurements and gel electrophoresis. The relative ability of the complexes to cleave DNA even in the absence of hydrogen peroxide is in the order 3 > 2 > 1. Application of the reactive oxygen species (ROS) scavengers, L-histidine, DMSO and SOD confirmed that singlet oxygen, hydroxyl radicals (Fenton reaction) and superoxide radical were formed, respectively. Thus, in addition to mechanism of intercalation, redox-cycling mechanism which in turn lead to the formation of ROS contribute to DNA damage. Cu(II) complexes exhibit excellent SOD-mimetic activity in the order 3~1 > 2. The fluorescence spectroscopy revealed that albumin may act as a targeted drug delivery vehicle for Cu(II) complexes (K~106). The anticancer activities of complexes 1-3 were investigated using an MTS assay (reduction of the tetrazolium compound) against three cancer cell lines (HT-29 human colon adenocarcinoma, HeLa and T-47D breast cancer cells) and mesenchymal stromal cells (MSC). The most promising compound, from the viewpoint of its NSAID biological activity is 3, due to the presence of the three fluorine atoms participating in the formation of weak hydrogen-bonds at the DNA surface.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , ADN/efectos de los fármacos , Fenamatos/farmacología , Sustancias Intercalantes/farmacología , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/metabolismo , Materiales Biomiméticos/farmacología , Línea Celular Tumoral , Complejos de Coordinación/síntesis química , Complejos de Coordinación/metabolismo , Cobre/química , ADN/metabolismo , Daño del ADN/efectos de los fármacos , Escherichia coli/química , Fenamatos/síntesis química , Fenamatos/metabolismo , Ácido Flufenámico/síntesis química , Ácido Flufenámico/metabolismo , Ácido Flufenámico/farmacología , Humanos , Sustancias Intercalantes/síntesis química , Sustancias Intercalantes/metabolismo , Ácido Mefenámico/síntesis química , Ácido Mefenámico/metabolismo , Ácido Mefenámico/farmacología , Oxidación-Reducción , Fenantrolinas/síntesis química , Fenantrolinas/metabolismo , Fenantrolinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Albúmina Sérica Humana , Superóxido Dismutasa/química , ortoaminobenzoatos/síntesis química , ortoaminobenzoatos/metabolismo , ortoaminobenzoatos/farmacología
12.
Xenobiotica ; 49(4): 397-403, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29543105

RESUMEN

Coumadin (R/S-warfarin) metabolism plays a critical role in patient response to anticoagulant therapy. Several cytochrome P450s oxidize warfarin into R/S-6-, 7-, 8-, 10, and 4'-hydroxywarfarin that can undergo subsequent glucuronidation by UDP-glucuronosyltransferases (UGTs); however, current studies on recombinant UGTs cannot be adequately extrapolated to microsomal glucuronidation capacities for the liver. Herein, we estimated the capacity of the average human liver to glucuronidate hydroxywarfarin and identified UGTs responsible for those metabolic reactions through inhibitor phenotyping. There was no observable activity toward R/S-warfarin, R/S-10-hydroxywarfarin or R/S-4'-hydroxywarfarin. The observed metabolic efficiencies (Vmax/Km) toward R/S-6-, 7-, and especially 8-hydroxywarfarin indicated a high glucuronidation capacity to metabolize these compounds. UGTs demonstrated strong regioselectivity toward the hydroxywarfarins. UGT1A6 and UGT1A1 played a major role in R/S-6- and 7-hydroxywarfarin glucuronidation, respectively, whereas UGT1A9 accounted for almost all of the generation of the R/S-8-hydroxywarfarin glucuronide. In summary, these studies expanded insights to glucuronidation of hydroxywarfarins by pooled human liver microsomes, novel roles for UGT1A6 and 1A9, and the overall degree of regioselectivity for the UGT reactions.


Asunto(s)
Glucurónidos/metabolismo , Microsomas Hepáticos/metabolismo , Warfarina/análogos & derivados , Bilirrubina/química , Bilirrubina/metabolismo , Glucuronosiltransferasa/antagonistas & inhibidores , Humanos , Concentración 50 Inhibidora , Cinética , Ácido Mefenámico/química , Ácido Mefenámico/metabolismo , Fenotipo , Serotonina/química , Serotonina/metabolismo , Estereoisomerismo , Warfarina/química , Warfarina/metabolismo
13.
Biochem Pharmacol ; 159: 25-31, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30423313

RESUMEN

The activities of hundreds, perhaps thousands, of metabolites are regulated by human cytosolic sulfotransferases (SULTs) - a 13-member family of disease relevant enzymes that catalyze transfer of the sulfuryl moiety (-SO3) from PAPS (3'-phosphoadenosine 5'-phosphosulfonate) to the hydroxyls and amines of acceptors. SULTs harbor two independent allosteric sites, one of which, the focus of this work, binds non-steroidal anti-inflammatory drugs (NSAIDs). The structure of the first NSAID-binding site - that of SULT1A1 - was elucidated recently and homology modeling suggest that variants of the site are present in all SULT isoforms. The objective of the current study was to assess whether the NSAID-binding site can be used to regulate sulfuryl transfer in humans in an isoform specific manner. Mefenamic acid (Mef) is a potent (Ki 27 nM) NSAID-inhibitor of SULT1A1 - the predominant SULT isoform in small intestine and liver. Acetaminophen (APAP), a SULT1A1 specific substrate, is extensively sulfonated in humans. Dehydroepiandrosterone (DHEA) is specific for SULT2A1, which we show here is insensitive to Mef inhibition. APAP and DHEA sulfonates are readily quantified in urine and thus the effects of Mef on APAP and DHEA sulfonation could be studied non-invasively. Compounds were given orally in a single therapeutic dose to a healthy, adult male human with a typical APAP-metabolite profile. Mef profoundly decreased APAP sulfonation during first pass metabolism and substantially decreased systemic APAP sulfonation without influencing DHEA sulfonation; thus, it appears the NSAID site can be used to control sulfonation in humans in a SULT-isoform specific manner.


Asunto(s)
Acetaminofén/farmacocinética , Arilsulfotransferasa/metabolismo , Ácido Mefenámico/farmacocinética , Sulfotransferasas/metabolismo , Acetaminofén/metabolismo , Acetaminofén/orina , Sitio Alostérico , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacocinética , Arilsulfotransferasa/antagonistas & inhibidores , Arilsulfotransferasa/química , Sitios de Unión , Deshidroepiandrosterona/administración & dosificación , Deshidroepiandrosterona/metabolismo , Deshidroepiandrosterona/orina , Interacciones Farmacológicas , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Espectroscopía de Resonancia Magnética , Ácido Mefenámico/metabolismo , Ácido Mefenámico/orina , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/química
14.
J Biol Chem ; 292(49): 20305-20312, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29038294

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most commonly prescribed drugs worldwide-more than 111 million prescriptions were written in the United States in 2014. NSAIDs allosterically inhibit cytosolic sulfotransferases (SULTs) with high specificity and therapeutically relevant affinities. This study focuses on the interactions of SULT1A1 and mefenamic acid (MEF)-a potent, highly specific NSAID inhibitor of 1A1. Here, the first structure of an NSAID allosteric site-the MEF-binding site of SULT1A1-is determined using spin-label triangulation NMR. The structure is confirmed by site-directed mutagenesis and provides a molecular framework for understanding NSAID binding and isoform specificity. The mechanism of NSAID inhibition is explored using molecular dynamics and equilibrium and pre-steady-state ligand-binding studies. MEF inhibits SULT1A1 turnover through an indirect (helix-mediated) stabilization of the closed form of the active-site cap of the enzyme, which traps the nucleotide and slows its release. Using the NSAID-binding site structure of SULT1A1 as a comparative model, it appears that 11 of the 13 human SULT isoforms harbor an NSAID-binding site. We hypothesize that these sites evolved to enable SULT isoforms to respond to metabolites that lie within their metabolic domains. Finally, the NSAID-binding site structure offers a template for developing isozyme-specific allosteric inhibitors that can be used to regulate specific areas of sulfuryl-transfer metabolism.


Asunto(s)
Sitio Alostérico , Antiinflamatorios no Esteroideos/metabolismo , Citosol/enzimología , Sulfotransferasas/química , Arilsulfotransferasa/antagonistas & inhibidores , Humanos , Isoenzimas/metabolismo , Espectroscopía de Resonancia Magnética , Ácido Mefenámico/metabolismo , Ácido Mefenámico/farmacología , Unión Proteica , Sulfotransferasas/antagonistas & inhibidores
15.
Eur J Pharmacol ; 801: 28-34, 2017 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-28259712

RESUMEN

Drug induced gastrointestinal ulceration, renal side effects and hepatotoxicity are the main causes of numerous Non-Steroidal Anti-inflammatory Drugs (NSAIDs). Cyclooxygenase-2 (COX-2) inhibitors discovered to decrease the gastrointestinal issues, but unfortunately, most of them are associated with major cardiovascular adverse effects. Along these lines, various new strategies and frameworks were developed wherein basic alterations of the present medications were accounted for. The aim of the study was to prepare derivatives of mefenamic acid to evaluate anti-inflammatory activity with fewer adverse reactions. In this study, molecular docking investigations of outlined derivatives were done utilizing Protein Data Bank (PDB ID-4PH9). Synthesis of heterocyclic compounds was carried out utilizing Dicyclohexylcarbodiimide/4-Dimethylaminopyridine (DCC/DMAP) coupling. Acute toxicity prediction was performed using free online GUSAR (General Unrestricted Structure-Activity Relationships) software. The study indicated most of the compounds under safe category. In-vitro pharmacological assessment of heterocyclic compounds was done for COX-1 and COX-2 enzymes for the determination of selectivity. In vivo pharmacological screening for anti-inflammatory activity and ED50 value were determined utilizing carrageenan induced rat paw edema. Gastro intestinal safety study was carried out on selected compounds and found to be devoid of any gastric ulcer toxicity. Most of the compounds indicated high scores as compared to standard during molecular modelling, analysis and displayed interactions with active amino acids of a COX-2 enzyme. The pharmacological screening uncovered that compound substituted with p-bromophenyl indicated maximum potency.


Asunto(s)
Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/farmacología , Ácido Mefenámico/síntesis química , Ácido Mefenámico/farmacología , Simulación del Acoplamiento Molecular , Amidas/química , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Técnicas de Química Sintética , Ciclooxigenasa 1/química , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/química , Ciclooxigenasa 2/metabolismo , Evaluación Preclínica de Medicamentos , Ácido Mefenámico/química , Ácido Mefenámico/metabolismo , Conformación Proteica , Ratas , Ratas Wistar , Relación Estructura-Actividad
16.
J Pharm Sci ; 106(4): 1117-1123, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28057544

RESUMEN

Transglycosylated stevia (stevia-G) can effectively improve the dissolution and bioavailability of poorly water-soluble drugs. Furthermore, addition of an ionic surfactant to stevia-G solution has been shown to enhance the dissolution effect of stevia-G on flurbiprofen. Herein, 4 surfactants, namely sodium dodecyl sulfate, sodium N-dodecanoylsarcosinate, sodium monododecyl phosphate, and lauryltrimethylammonium chloride (LTAC) were screened to investigate their synergistic effect with stevia-G in enhancing the solubility of mefenamic acid (MFA). The ternary formulation containing LTAC produced the highest increase in solubility, whereas the binary MFA/LTAC formulation did not increase the solubility of MFA. Surface tension was evaluated to analyze the interaction between stevia-G and each ionic surfactant, wherein the Rubingh model was applied to predict mixed micelle formation between stevia-G and LTAC. Interaction parameters calculated by the Rubingh model reflected mixed micelle formation between stevia-G and LTAC relative to the self-interactions of the 2 individual surfactants. All interaction parameters in this system showed negative values, indicating a favorable interaction (e.g., hydrogen bond or electrostatic and dipole) between binary components in the mixed micelles. Spray-dried particles of ternary formulations (MFA/stevia-G/LTAC) were prepared to evaluate the dissolution profile and physicochemical properties. Dissolution profiling showed that the concentration of MFA released from spray-dried particles was significantly higher than untreated MFA.


Asunto(s)
Ácido Mefenámico/metabolismo , Micelas , Stevia/metabolismo , Tensoactivos/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas/fisiología , Glicosilación , Ácido Mefenámico/química , Solubilidad , Stevia/química , Tensión Superficial , Tensoactivos/química , Difracción de Rayos X
17.
Med Chem ; 12(6): 585-91, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26740206

RESUMEN

BACKGROUND: Non steroidal anti-inflammatory drugs are the most widely prescribed drugs to manage pain and inflammatory conditions, but their long term use is associated with gastrointestinal toxicity. OBJECTIVES: The study aimed to synthesize an ester-based prodrug of a non steroidal anti-inflammatory agent, mefenamic acid in order to improve the therapeutic index vis a vis to overcome the side effects such as gastrointestinal irritation and bleeding associated with the use of mefenamic acid. METHODS: The ester prodrug (MA-NH) was prepared by condensing mefenamic acid with N-hydroxymethylsuccinimide in the presence of Phosphorus oxychloride. The pharmacokinetic profile, including stability and release of mefenamic acid and N-hydroxymethylsuccinimide from the ester prodrug (MA-NH) was studied by RP- HPLC in acidic medium (pH 1.2), basic medium (pH 7.4), 80 % v/v human plasma, 10 % w/v rat intestinal homogenate and 10 % w/v rat liver homogenate (pH 7.4). RESULTS: The chemical structure of the title compound was characterized by using modern spectroscopic techniques. The prodrug was found to be stable in acid medium, but it hydrolyzed and released sufficient quantities of the drug in alkaline medium. The prodrug produced lesser number of ulcers and showed improved analgesic and anti-inflammatory activity as compared to the parent drug. CONCLUSION: The results indicate that the synthesized prodrug (MA-NH) is better in terms of analgesic and antiinflammatory activities and with less GI toxicity than the parent drug.


Asunto(s)
Analgésicos/uso terapéutico , Antiinflamatorios no Esteroideos/uso terapéutico , Ácido Mefenámico/análogos & derivados , Ácido Mefenámico/metabolismo , Profármacos/uso terapéutico , Succinimidas/uso terapéutico , Analgésicos/síntesis química , Analgésicos/farmacocinética , Analgésicos/toxicidad , Animales , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/farmacocinética , Antiinflamatorios no Esteroideos/toxicidad , Femenino , Humanos , Hidrólisis , Masculino , Ácido Mefenámico/síntesis química , Ácido Mefenámico/farmacocinética , Ácido Mefenámico/uso terapéutico , Ácido Mefenámico/toxicidad , Profármacos/síntesis química , Profármacos/farmacocinética , Profármacos/toxicidad , Ratas , Ratas Wistar , Succinimidas/síntesis química , Succinimidas/química , Succinimidas/farmacocinética , Succinimidas/toxicidad , Úlcera/inducido químicamente
18.
Acta Pharm ; 65(4): 453-62, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26677901

RESUMEN

Mefenamic acid (MA) is a widely used non-steroidal antiinflammatory (NSAID) drug. The adverse effects typical of NSAIDs are also present in the case of MA, partly due to its low water solubility. The aim of this study was to increase the water solubility of MA in order to influence its absorption and bioavailability. Solid dispersions of MA were prepared by the melting method using polyethylene glycol 6000 and different types (laurate, D-1216; palmitate, P-1670; stearate, S-1670) and amounts of sucrose esters as carriers. The X-ray diffraction results show that MA crystals were not present in the products. Dissolution tests carried out in artificial intestinal juice showed that the product containing 10 % D-1216 increased water solubility about 3 times. The apparent permeability coefficient of MA across human Caco-2 intestinal epithelial cell layers was high and, despite the difference in solubility, there was no further increase in drug penetration in the presence of the applied additives.


Asunto(s)
Antiinflamatorios no Esteroideos/química , Portadores de Fármacos , Ésteres/química , Ácido Mefenámico/química , Polietilenglicoles/química , Sacarosa/química , Antiinflamatorios no Esteroideos/metabolismo , Células CACO-2 , Química Farmacéutica , Impedancia Eléctrica , Humanos , Absorción Intestinal , Mucosa Intestinal/metabolismo , Secreciones Intestinales/química , Cinética , Ácido Mefenámico/metabolismo , Permeabilidad , Solubilidad , Solventes/química , Sacarosa/análogos & derivados , Agua/química
19.
Biopharm Drug Dispos ; 36(8): 529-51, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26109199

RESUMEN

Mefenamic acid (MEF) is a widely prescribed non-steroidal anti-inflammatory drug that has been found associated with rare but severe cases of hepatotoxicity, nephrotoxicity and gastrointestinal toxicity. The formation of protein-reactive acylating metabolites such as 1-O-acyl-MEF glucuronide (MEFG) and 3'-hydroxymethyl-MEF 1-O-acyl-glucuronide is one proposed cause. In addition to the well-reported 3'-hydroxymethyl-MEF, two mono-hydroxyl-MEF (OH-MEFs) were recently identified in vitro. However, in vivo evidence is lacking and whether these OH-MEFs would be further glucuronidated to the potentially reactive 1-O-acyl-glucuronides (OH-MEFGs) is unknown. Utilizing UPLC-Q-TOF/MS and LC-MS/MS, the current study identified, for the first time, four OH-MEFs and their corresponding OH-MEFGs from plasma after a single oral administration of MEF (40 mg/kg) to rats, including an OH-MEF that has not been reported previously. The systemic exposure of these identified metabolites was high, with metabolic to parent AUC0 → 24 h ratios reaching 23-52% (OH-MEFs) and 8-29% (OH-MEFGs). These metabolites also had a long systemic exposure time in both single and 5 day multiple oral MEF-treated rats, with elimination half-lives between 9 h and > 24 h. In addition to these novel metabolites, the previously reported MEFG was also identified and its systemic exposure was found to be doubled after multiple MEF administrations. These pharmacokinetic results suggest that systemic toxicities caused by the potentially reactive MEFG and OH-MEFGs could be considerable, especially after repeated MEF treatment. Nevertheless, MEFG and OH-MEFGs had negligible uptake in the brain, indicating a minimal risk of brain toxicities. Furthermore, an in situ intestinal perfusion study revealed that during MEF absorption, it was extensively metabolized to MEFG while < 5% was metabolized to OH-MEFs and OH-MEFGs.


Asunto(s)
Encéfalo/metabolismo , Intestino Delgado/metabolismo , Ácido Mefenámico/análogos & derivados , Microsomas Hepáticos/metabolismo , Administración Oral , Animales , Biotransformación , Encéfalo/efectos de los fármacos , Cromatografía Liquida , Relación Dosis-Respuesta a Droga , Glucuronatos/sangre , Glucuronatos/metabolismo , Glucuronatos/farmacocinética , Glucuronatos/toxicidad , Técnicas In Vitro , Absorción Intestinal , Masculino , Ácido Mefenámico/sangre , Ácido Mefenámico/metabolismo , Ácido Mefenámico/farmacocinética , Ácido Mefenámico/toxicidad , Estructura Molecular , Perfusión , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Distribución Tisular
20.
Drug Test Anal ; 7(7): 626-33, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25209992

RESUMEN

A new model is presented that can be used to screen for bioactivation of drugs. The evaluation of toxicity is an important step in the development of new drugs. One way to detect possible toxic metabolites is to use trapping agents such as glutathione. Often human liver microsomes are used as a metabolic model in initial studies. However, there is a need for alternatives that are easy to handle, cheap, and can produce large amounts of metabolites. In the presented study, paracetamol, mefenamic acid, and diclofenac, all known to form reactive metabolites in humans, were incubated with the fungus Cunninghamella elegans and the metabolites formed were characterized with ultra high performance liquid chromatography coupled to a quadrupole time of flight mass spectrometer. Interestingly, glutathione conjugates formed by the fungus were observed for all three drugs and their retention times and MS/MS spectra matched those obtained in a comparative experiment with human liver microsomes. These findings clearly demonstrated that the fungus is a suitable trapping model for toxic biotransformation products. Cysteine conjugates of all three test drugs were also observed with high signal intensities in the fungal incubates, giving the model a further indicator of drug bioactivation. To our knowledge, this is the first demonstration of the use of a fungal model for the formation and trapping of reactive drug metabolites. The investigated model is cheap, easy to handle, it does not involve experimental animals and it can be scaled up to produce large amounts of metabolites.


Asunto(s)
Acetaminofén/metabolismo , Cunninghamella/metabolismo , Diclofenaco/metabolismo , Ácido Mefenámico/metabolismo , Espectrometría de Masas en Tándem/métodos , Acetaminofén/análisis , Animales , Cromatografía Líquida de Alta Presión/métodos , Cunninghamella/química , Diclofenaco/análisis , Humanos , Ácido Mefenámico/análisis , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...