Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Bioorg Chem ; 147: 107393, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38691908

RESUMEN

Cyclooxygenase-2 plays a vital role in inflammation by catalyzing arachidonic acid conversion toward prostaglandins, making it a prime therapeutic objective. Selective COX-2 inhibitors represent significant progress in anti-inflammatory therapy, offering improved efficacy and fewer side effects. This study describes the synthesis of novel anti-inflammatory compounds from established pharmaceutically marketed agents like fenamates III-V and ibuprofen VI. Through rigorous in vitro testing, compounds 7b-c, and 12a-b demonstrated substantial in vitro selective inhibition, with IC50 values of 0.07 to 0.09 µM, indicating potent pharmacological activity. In vivo assessment, particularly focusing on compound 7c, revealed significant anti-inflammatory effects. Markedly, it demonstrated the highest inhibition of paw thickness (58.62 %) at the 5-hr mark compared to the carrageenan group, indicating its potency in mitigating inflammation. Furthermore, it exhibited a rapid onset of action, with a 54.88 % inhibition observed at the 1-hr mark. Subsequent comprehensive evaluations encompassing analgesic efficacy, histological characteristics, and toxicological properties indicated that compound 7c did not induce gastric ulcers, in contrast to the ulcerogenic tendency associated with mefenamic acid. Moreover, compound 7c underwent additional investigations through in silico methodologies such as molecular modelling, field alignment, and density functional theory. These analyses underscored the therapeutic potential and safety profile of this novel compound, warranting further exploration and development in the realm of pharmaceutical research.


Asunto(s)
Antiinflamatorios no Esteroideos , Carragenina , Inhibidores de la Ciclooxigenasa 2 , Ciclooxigenasa 2 , Fenamatos , Ibuprofeno , Ibuprofeno/farmacología , Ibuprofeno/química , Ibuprofeno/síntesis química , Ciclooxigenasa 2/metabolismo , Animales , Inhibidores de la Ciclooxigenasa 2/farmacología , Inhibidores de la Ciclooxigenasa 2/síntesis química , Inhibidores de la Ciclooxigenasa 2/química , Estructura Molecular , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/síntesis química , Relación Estructura-Actividad , Fenamatos/farmacología , Fenamatos/química , Fenamatos/síntesis química , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Edema/tratamiento farmacológico , Edema/inducido químicamente , Simulación del Acoplamiento Molecular , Ratas , Masculino
2.
J Inorg Biochem ; 228: 111696, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35030390

RESUMEN

Through the reaction of copper(II) acetate with nicotinamide (pyridine-3-carboxylic acid amide, niacinamide) and some derivatives of N-phenylanthranilic acid (fenamates), seven new mixed-ligand copper(II) compounds were isolated: [Cu(tolf-O)(tolf-O,O')nia-N)2(EtOH)] (1), [Cu(tolf-O)(tolf-O,O')(nia-N)2(MeOH)] (2), [Cu(meclf-O)(meclf-O,O')(nia-N)2(EtOH)] (3), [Cu(meclf-O)(meclf-O,O')(nia-N)2(MeOH)] (4), [Cu(meclf-O)(meclf-O,O')(nia-N)2(ACN)] (5), [Cu(mef-O)(mef-O,O')(nia-N)2(EtOH)] (6) and [Cu(mef-O)(mef-O,O')(nia-N)2(ACN)] (7) containing a molecule of relevant solvent as ligand in their primary crystal structure (tolf = tolfenamate, meclf = meclofenamate, mef = mefenamate, nia = nicotinamide, EtOH = ethanol, MeOH = methanol, ACN = acetonitrile). The structures of the complexes were determined by single-crystal X-ray analysis. The intermolecular interactions were studied by Hirshfeld surface analysis. The complexes were characterized by IR, UV-vis and EPR spectroscopy and their redox properties were determined by cyclic voltammetry. The interaction of the complexes with bovine serum albumin was studied by fluorescence emission spectroscopy and the albumin-binding constants of the compounds were calculated. The interaction of the complexes with calf-thymus DNA was monitored by diverse techniques (UV-vis spectroscopy, cyclic voltammetry, viscosity measurements) suggesting intercalation as the most possible mode of binding. DNA-competitive studies of the complexes with ethidium bromide were monitored by fluorescence emission spectroscopy. The cytotoxic effects of copper(II) complexes on lung carcinoma cells and healthy cells were determined by the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] colorimetric technique.


Asunto(s)
Antiinflamatorios no Esteroideos/química , Complejos de Coordinación/química , Cobre/química , ADN/química , Niacinamida/química , Albúmina Sérica Bovina/química , Células A549 , Animales , Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Cristalografía por Rayos X/métodos , Espectroscopía de Resonancia por Spin del Electrón/métodos , Etidio/química , Fenamatos/química , Humanos , Sustancias Intercalantes/química , Oxidación-Reducción
3.
J Inorg Biochem ; 218: 111410, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33721718

RESUMEN

The interaction of FeCl3 with the fenamate non-steroidal anti-inflammatory drugs has led to the formation and isolation of trinuclear iron(III) complexes, while in the presence of the nitrogen-donors 2,2'-bipyridine or pyridine tetranuclear iron(III) complexes were derived. The five resultant complexes were characterized by diverse techniques (including infrared, electronic and Mössbauer spectroscopy) and their crystal structures were determined by single-crystal X-ray crystallography. These complexes are the first structurally characterized Fe(III)-fenamato complexes. The complexes were evaluated for their ability to scavenge in vitro free radicals such as hydroxyl, 1,1-diphenyl-2-picrylhydrazyl and 2,2΄-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid). The in vitro binding affinity of the complexes to calf-thymus (CT) DNA was examined and their interaction with serum albumins was also investigated. In total, the complexes present promising activity against the radicals tested, and they may bind tightly to CT DNA possibly via intercalation and reversibly to serum albumins.


Asunto(s)
Complejos de Coordinación/metabolismo , ADN/metabolismo , Fenamatos/metabolismo , Hierro/química , Hierro/metabolismo , Albúmina Sérica Bovina/metabolismo , Albúmina Sérica Humana/metabolismo , Animales , Complejos de Coordinación/química , Cristalografía por Rayos X , Fenamatos/química , Humanos , Sustancias Intercalantes/metabolismo , Estructura Molecular
4.
Eur Biophys J ; 49(7): 591-607, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32940715

RESUMEN

GABA and glycine act as inhibitory neurotransmitters in the CNS. Inhibitory neurotransmission is mediated via activation of ionotropic GABAA and glycine receptors. We used a modeling approach to explain the opposite effects of the general anesthetic etomidate (ETM) and fenamate mefenamic acid (MFA) on GABA- and glycine-activated currents recorded in isolated cerebellar Purkinje cells and hippocampal pyramidal neurons, respectively. These drugs potentiated GABAARs but blocked GlyRs. We built a homology model of α1ß GlyR based on the cryo-EM structure of open α1 GlyR, used the α1ß3γ2 GABAAR structure from the PDB, and applied Monte-Carlo energy minimization to optimize models of receptors and ligand-receptor complexes. In silico docking suggests that ETM/MFA bind at the transmembrane ß( +)/α( -) intersubunit interface in GABAAR. Our models predict that the bulky side chain of the highly conserved Arg19' residue at the plus interface side wedges the interface and maintains the conducting receptor state. We hypothesized that MFA/ETM binding at the ß( +)/α( -) interface leads to prolongation of receptor life-time in the open state. Having analyzed different GABAAR and GlyR structures available in the PDB, we found that mutual arrangement of the Arg19' and Gln-26' side chains at the plus and minus interface sides, respectively, plays an important role when the receptor switches from the open to closed state. We show that this process is accompanied by narrowing of the intersubunit interfaces, leading to extrusion of the Arg19' side chain from the interface. Our models allow us to explain the lack of GlyR potentiation in our electrophysiological experiments.


Asunto(s)
Etomidato/química , Ácido Mefenámico/química , Neuronas/metabolismo , Proteínas Nucleares/química , Oxidorreductasas/química , Receptores de GABA-A/química , Anestésicos Generales/farmacología , Animales , Sitios de Unión , Simulación por Computador , Bases de Datos de Proteínas , Electrofisiología , Fenamatos/química , Glicina/química , Ligandos , Conformación Molecular , Simulación del Acoplamiento Molecular , Método de Montecarlo , Unión Proteica , Ratas , Ratas Wistar , Receptores de Glicina/química , Transmisión Sináptica
5.
J Med Chem ; 62(17): 8274-8283, 2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31393717

RESUMEN

The wild type protein, transthyretin (TTR), and over 120 genetic TTR variants are amyloidogenic and cause, respectively, sporadic and hereditary systemic TTR amyloidosis. The homotetrameric TTR contains two identical thyroxine binding pockets, occupation of which by specific ligands can inhibit TTR amyloidogenesis in vitro. Ligand binding stabilizes the tetramer, inhibiting its proteolytic cleavage and its dissociation. Here, we show with solution-state NMR that ligand binding induces long-distance conformational changes in the TTR that have not previously been detected by X-ray crystallography, consistently with the inhibition of the cleavage of the DE loop. The NMR findings, coupled with surface plasmon resonance measurements, have identified dynamic exchange processes underlying the negative cooperativity of binding of "monovalent" ligand tafamidis. In contrast, mds84, our prototypic "bivalent" ligand, which is a more potent stabilizer of TTR in vitro that occupies both thyroxine pockets and the intramolecular channel between them, has greater structural effects.


Asunto(s)
Fenamatos/química , Prealbúmina/química , Sitios de Unión , Fenamatos/síntesis química , Humanos , Ligandos , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Prealbúmina/síntesis química , Relación Estructura-Actividad
6.
Org Biomol Chem ; 17(13): 3409-3415, 2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30869715

RESUMEN

Rare multiple fluorescence properties including aggregation-induced emission and polymorphism/shape/size-dependent emission were found coexisting in a class of typical non-steroidal anti-inflammatory analgesic drugs, fenamates, which could provide a new approach toward future drug evaluation. Different from the complexity and biological incompatibility of the traditional AIE molecular design, this work opens new avenues to the development of new AIE systems.


Asunto(s)
Analgésicos/química , Antiinflamatorios no Esteroideos/química , Fenamatos/química , Colorantes Fluorescentes/química , Evaluación de Medicamentos , Fluorescencia , Estructura Molecular , Tamaño de la Partícula , Teoría Cuántica
7.
Environ Sci Process Impacts ; 19(5): 656-665, 2017 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-28401228

RESUMEN

Fenamates are a class of nonsteroidal anti-inflammatory drugs (NSAIDs) that are not fully removed during wastewater treatment and can be released to surface waters. Here, near-surface photochemical half-lives were evaluated to range from minutes to hours of four fenamates and the closely related diclofenac. While quantum yields for direct photochemical reactions at the water surface vary widely from 0.071 for diclofenac to <0.001 for mefenamic acid, all fenamates showed significant reactivity towards singlet oxygen and hydroxyl radical with bimolecular reaction rate constants of 1.3-2.8 × 107 M-1 s-1 and 1.1-2.7 × 1010 M-1 s-1, respectively. Photodecay rates increased in the presence of dissolved organic matter (DOM) for diclofenac (+19%), tolfenamic acid (+9%), and mefenamic acid (+95%), but decreased for flufenamic acid (-2%) and meclofenamic acid (-14%) after accounting for light screening effects. Fast reaction rate constants of all NSAIDs with model triplet sensitizers were quantified by laser flash photolysis. Here, the direct observation of diphenylamine radical intermediates by transient absorption spectroscopy demonstrates one-electron oxidation of all fenamates. Quenching rate constants of these radical intermediates by ascorbic acid, a model antioxidant, were also quantified. These observations suggest that the balance of oxidation by photoexcited triplet DOM and quenching of the formed radical intermediates by antioxidant moieties determines whether net sensitization or net quenching by DOM occurs in the photochemical degradation of fenamates.


Asunto(s)
Antiinflamatorios no Esteroideos/análisis , Fenamatos/análisis , Sustancias Húmicas/análisis , Luz , Contaminantes Químicos del Agua/análisis , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/efectos de la radiación , Restauración y Remediación Ambiental , Fenamatos/química , Fenamatos/efectos de la radiación , Agua Dulce/química , Radical Hidroxilo/química , Modelos Teóricos , Oxidación-Reducción , Fotoquímica , Oxígeno Singlete/química , Análisis Espectral , Aguas Residuales/química , Contaminantes Químicos del Agua/química , Contaminantes Químicos del Agua/efectos de la radiación
8.
J Biol Chem ; 291(29): 15069-81, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27226593

RESUMEN

Cyclooxygenase-2 (COX-2) catalyzes the oxygenation of arachidonic acid (AA) and endocannabinoid substrates, placing the enzyme at a unique junction between the eicosanoid and endocannabinoid signaling pathways. COX-2 is a sequence homodimer, but the enzyme displays half-of-site reactivity, such that only one monomer of the dimer is active at a given time. Certain rapid reversible, competitive nonsteroidal anti-inflammatory drugs (NSAIDs) have been shown to inhibit COX-2 in a substrate-selective manner, with the binding of inhibitor to a single monomer sufficient to inhibit the oxygenation of endocannabinoids but not arachidonic acid. The underlying mechanism responsible for substrate-selective inhibition has remained elusive. We utilized structural and biophysical methods to evaluate flufenamic acid, meclofenamic acid, mefenamic acid, and tolfenamic acid for their ability to act as substrate-selective inhibitors. Crystal structures of each drug in complex with human COX-2 revealed that the inhibitor binds within the cyclooxygenase channel in an inverted orientation, with the carboxylate group interacting with Tyr-385 and Ser-530 at the top of the channel. Tryptophan fluorescence quenching, continuous-wave electron spin resonance, and UV-visible spectroscopy demonstrate that flufenamic acid, mefenamic acid, and tolfenamic acid are substrate-selective inhibitors that bind rapidly to COX-2, quench tyrosyl radicals, and reduce higher oxidation states of the heme moiety. Substrate-selective inhibition was attenuated by the addition of the lipid peroxide 15-hydroperoxyeicosatertaenoic acid. Collectively, these studies implicate peroxide tone as an important mechanistic component of substrate-selective inhibition by flufenamic acid, mefenamic acid, and tolfenamic acid.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/química , Inhibidores de la Ciclooxigenasa 2/farmacología , Ciclooxigenasa 2/metabolismo , Fenamatos/farmacología , Sustitución de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Ciclooxigenasa 2/química , Ciclooxigenasa 2/genética , Endocannabinoides/metabolismo , Fenamatos/química , Hemo/química , Humanos , Técnicas In Vitro , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Peróxidos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Triptófano/química
9.
Phys Chem Chem Phys ; 17(12): 7936-48, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25720865

RESUMEN

In traditional molecular mechanics force fields, intramolecular non-bonded interactions are modelled as intermolecular interactions, and the form of the torsion potential is based on the conformational profiles of small organic molecules. We investigate how a separate model for the intramolecular forces in pharmaceuticals could be more realistic by analysing the low barrier to rotation of the phenyl ring in the fenamates (substituted N-phenyl-aminobenzoic acids), that results in a wide range of observed angles in the numerous fenamate crystal structures. Although the conformational energy changes by significantly less than 10 kJ mol(-1) for a complete rotation of the phenyl ring for fenamic acid, the barrier is only small because of small correlated changes in the other bond and torsion angles. The maxima for conformations where the two aromatic rings approach coplanarity arise from steric repulsion, but the maxima when the two rings are approximately perpendicular arise from a combination of an electronic effect and intramolecular dispersion. Representing the ab initio conformational energy profiles as a cosine series alone is ineffective; however, combining a cos 2ξ term to represent the electronic barrier with an intramolecular atom-atom exp-6 term for all atom pairs separated by three or more bonds (1-4 interactions) provides a very effective representation. Thus we propose a new, physically motivated, generic analytical model of conformational energy, which could be combined with an intermolecular model to form more accurate force-fields for modelling the condensed phases of pharmaceutical-like organic molecules.


Asunto(s)
Fenamatos/química , Preparaciones Farmacéuticas/química , Electrones , Conformación Molecular , Simulación de Dinámica Molecular , Termodinámica
10.
Artículo en Inglés | MEDLINE | ID: mdl-22954807

RESUMEN

Solid-state Raman and IR spectra of two polymorphic forms of each of three fenamates (flufenamic acid, mefenamic acid and tolfenamic acid) display subtle but highly reproducible differences. Many of these spectral differences can be ascribed to different conformations of these molecules, involving two of four possible orientations of one substituted benzene ring with respect to the other. Interpretation of the vibrational spectra in terms of conformational differences has been facilitated by DFT calculations at the B3LYP/cc-pVDZ level for each conformer. The calculated spectra are compared with the experimental spectra in order to identify the conformers present in two polymorphic forms in each case, and detailed band assignments are obtained from the DFT calculations.


Asunto(s)
Fenamatos/química , Conformación Molecular , Espectrometría Raman , Ácido Flufenámico/química , Ácido Mefenámico/química , Modelos Moleculares , Teoría Cuántica , Espectrofotometría Infrarroja , Termodinámica , Vibración , ortoaminobenzoatos/química
11.
Mol Pharmacol ; 82(5): 795-802, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22851714

RESUMEN

Niflumic acid, 2-{[3-(trifluoromethyl)phenyl]amino}pyridine-3-carboxylic acid (NFA), a nonsteroidal anti-inflammatory drug that blocks cyclooxygenase (COX), was shown previously to activate [Na(+)](i)-regulated Slo2.1 channels. In this study, we report that other fenamates, including flufenamic acid, mefenamic acid, tolfenamic acid, meclofenamic acid, and a phenyl acetic acid derivative, diclofenac, also are low-potency (EC(50) = 80 µM to 2.1 mM), partial agonists of human Slo2.1 channels heterologously expressed in Xenopus oocytes. Substituent analysis determined that N-phenylanthranilic acid was the minimal pharmacophore for fenamate activation of Slo2.1 channels. The effects of fenamates were biphasic, with an initial rapid activation phase followed by a slow phase of current inhibition. Ibuprofen, a structurally dissimilar COX inhibitor, did not activate Slo2.1. Preincubation of oocytes with ibuprofen did not significantly alter the effects of NFA, suggesting that neither channel activation nor inhibition is associated with COX activity. A point mutation (A278R) in the pore-lining S6 segment of Slo2.1 increased the sensitivity to activation and reduced the inhibition induced by NFA. Together, our results suggest that fenamates bind to two sites on Slo2.1 channels: an extracellular accessible site to activate and a cytoplasmic accessible site in the pore to inhibit currents.


Asunto(s)
Fenamatos/farmacología , Canales de Potasio/agonistas , Animales , Inhibidores de la Ciclooxigenasa/farmacología , Femenino , Fenamatos/química , Humanos , Ibuprofeno/farmacología , Técnicas In Vitro , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/agonistas , Ácido Niflúmico/farmacología , Oocitos/efectos de los fármacos , Oocitos/fisiología , Técnicas de Placa-Clamp , Mutación Puntual , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/genética , Canales de potasio activados por Sodio , Relación Estructura-Actividad , Xenopus laevis , ortoaminobenzoatos/farmacología
12.
Biochem Pharmacol ; 83(7): 923-31, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22285229

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) are widely used anti-inflammatory therapeutic agents, among which the fenamate analogues play important roles in regulating intracellular Ca²âº transient and ion channels. However, the effect of NSAIDs on TRPC4 and TRPC5 is still unknown. To understand the structure-activity of fenamate analogues on TRPC channels, we have synthesized a series of fenamate analogues and investigated their effects on TRPC4 and TRPC5 channels. Human TRPC4 and TRPC5 cDNAs in tetracycline-regulated vectors were transfected into HEK293 T-REx cells. The whole cell current and Ca²âº movement were recorded by patch clamp and calcium imaging, respectively. Flufenamic acid (FFA), mefenamic acid (MFA), niflumic acid (NFA) and diclofenac sodium (DFS) showed inhibition on TRPC4 and TRPC5 channels in a concentration-dependent manner. The potency was FFA>MFA>NFA>DFS. Modification of 2-phenylamino ring by substitution of the trifluoromethyl group in FFA with F, CH3, OCH3, OCH2CH3, COOH, and NO2 led to the changes in their channel blocking activity. However, 2-(2'-methoxy-5'-methylphenyl)aminobenzoic acid stimulated TRPC4 and TRPC5 channels. Selective COX1-3 inhibitors (aspirin, celecoxib, acetaminophen, and indomethacin) had no effect on the channels. Longer perfusion (> 5 min) with FFA (100 µM) and MFA (100 µM) caused a potentiation of TRPC4 and TRPC5 currents after their initial blocking effects that appeared to be partially mediated by the mitochondrial Ca²âº release. Our results suggest that fenamate analogues are direct modulators of TRPC4 and TRPC5 channels. The substitution pattern and conformation of the 2-phenylamino ring could alter their blocking activity, which is important for understanding fenamate pharmacology and new drug development targeting the TRPC channels.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Calcio/metabolismo , Fenamatos/farmacología , Canales Catiónicos TRPC/antagonistas & inhibidores , Animales , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/química , Técnicas de Cultivo de Célula , Fenamatos/síntesis química , Fenamatos/química , Colorantes Fluorescentes/química , Fura-2/análogos & derivados , Fura-2/química , Células HEK293 , Humanos , Masculino , Microscopía Fluorescente , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Músculo Liso Vascular/citología , Técnicas de Placa-Clamp , Ratas , Relación Estructura-Actividad , Canales Catiónicos TRPC/genética , Transfección
13.
J Med Chem ; 55(5): 2311-23, 2012 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-22263837

RESUMEN

Aldo-keto reductase 1C3 (AKR1C3; type 5 17ß-hydroxysteroid dehydrogenase) is overexpressed in castration resistant prostate cancer (CRPC) and is implicated in the intratumoral biosynthesis of testosterone and 5α-dihydrotestosterone. Selective AKR1C3 inhibitors are required because compounds should not inhibit the highly related AKR1C1 and AKR1C2 isoforms which are involved in the inactivation of 5α-dihydrotestosterone. NSAIDs, N-phenylanthranilates in particular, are potent but nonselective AKR1C3 inhibitors. Using flufenamic acid, 2-{[3-(trifluoromethyl)phenyl]amino}benzoic acid, as lead compound, five classes of structural analogues were synthesized and evaluated for AKR1C3 inhibitory potency and selectivity. Structure-activity relationship (SAR) studies revealed that a meta-carboxylic acid group relative to the amine conferred pronounced AKR1C3 selectivity without loss of potency, while electron withdrawing groups on the phenylamino B-ring were optimal for AKR1C3 inhibition. Lead compounds did not inhibit COX-1 or COX-2 but blocked the AKR1C3 mediated production of testosterone in LNCaP-AKR1C3 cells. These compounds offer promising leads toward new therapeutics for CRPC.


Asunto(s)
3-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Antineoplásicos/síntesis química , Fenamatos/síntesis química , Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , 20-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , 3-Hidroxiesteroide Deshidrogenasas/genética , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Ciclooxigenasa 1/metabolismo , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa/síntesis química , Inhibidores de la Ciclooxigenasa/química , Inhibidores de la Ciclooxigenasa/farmacología , Fenamatos/química , Fenamatos/farmacología , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , Isoenzimas/antagonistas & inhibidores , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Relación Estructura-Actividad , Testosterona/antagonistas & inhibidores , Testosterona/biosíntesis
14.
Eur J Med Chem ; 46(2): 497-508, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21167625

RESUMEN

In this work we have developed an in silico model to predict the inhibition of ß-amyloid aggregation by small organic molecules. In particular we have explored the inhibitory activity of a series of 62 N-phenylanthranilic acids using Kohonen maps and Counterpropagation Artificial Neural Networks. The effects of various structural modifications on biological activity are investigated and novel structures are designed using the developed in silico model. More specifically a search for optimized pharmacophore patterns by insertions, substitutions, and ring fusions of pharmacophoric substituents of the main building block scaffolds is described. The detection of the domain of applicability defines compounds whose estimations can be accepted with confidence.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Fenamatos/farmacología , Redes Neurales de la Computación , Análisis por Conglomerados , Fenamatos/química , Ensayos Analíticos de Alto Rendimiento , Ligandos , Modelos Moleculares , Estructura Molecular , Valor Predictivo de las Pruebas , Estereoisomerismo , Relación Estructura-Actividad
15.
Proc Natl Acad Sci U S A ; 107(47): 20483-8, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21059958

RESUMEN

Transthyretin (TTR) amyloidosis is a fatal disease for which new therapeutic approaches are urgently needed. We have designed two palindromic ligands, 2,2'-(4,4'-(heptane-1,7-diylbis(oxy))bis(3,5-dichloro-4,1-phenylene)) bis(azanediyl)dibenzoic acid (mds84) and 2,2'-(4,4'-(undecane-1,11-diylbis(oxy))bis(3,5-dichloro-4,1-phenylene)) bis(azanediyl)dibenzoic acid (4ajm15), that are rapidly bound by native wild-type TTR in whole serum and even more avidly by amyloidogenic TTR variants. One to one stoichiometry, demonstrable in solution and by MS, was confirmed by X-ray crystallographic analysis showing simultaneous occupation of both T4 binding sites in each tetrameric TTR molecule by the pair of ligand head groups. Ligand binding by native TTR was irreversible under physiological conditions, and it stabilized the tetrameric assembly and inhibited amyloidogenic aggregation more potently than other known ligands. These superstabilizers are orally bioavailable and exhibit low inhibitory activity against cyclooxygenase (COX). They offer a promising platform for development of drugs to treat and prevent TTR amyloidosis.


Asunto(s)
Amiloide/biosíntesis , Amiloidosis/metabolismo , Fenamatos/metabolismo , Ligandos , Prealbúmina/metabolismo , Amiloide/metabolismo , Amiloidosis/tratamiento farmacológico , Animales , Rastreo Diferencial de Calorimetría , Cromatografía en Gel , Cristalografía por Rayos X , Fenamatos/síntesis química , Fenamatos/química , Fenamatos/farmacocinética , Fluorometría , Espectrometría de Masas , Ratones , Modelos Moleculares , Estructura Molecular , Ultracentrifugación
16.
Biol Pharm Bull ; 33(5): 886-90, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20460771

RESUMEN

A human aldose reductase-like protein, AKR1B10 in the aldo-keto reductase (AKR) superfamily, was recently identified as a tumor marker of several types of cancer. Tolrestat, an aldose reductase inhibitor (ARI), is known to be the most potent inhibitor of the enzyme. In this study, we compared the inhibitory effects of other ARIs including flavonoids on AKR1B10 and aldose reductase to evaluate their specificity. However, ARIs showed lower inhibitory potency for AKR1B10 than for aldose reductase. In the search for potent and selective inhibitors of AKR1B10 from other drugs used clinically, we found that non-steroidal antiinflammatory N-phenylanthranilic acids, diclofenac and glycyrrhetic acid competitively inhibited AKR1B10, showing K(i) values of 0.35-2.9 microM and high selectivity to this enzyme (43-57 fold versus aldose reductase). Molecular docking studies of mefenamic acid and glycyrrhetic acid in the AKR1B10-nicotinamide adenine dinucleotide phosphate (NADP(+)) complex and site-directed mutagenesis of the putative binding residues suggest that the side chain of Val301 and a hydrogen-bonding network among residues Val301, Gln114 and Ser304 are important for determining the inhibitory potency and selectivity of the non-steroidal antiinflammatory drugs. Thus, the potent and selective inhibition may be related to the cancer chemopreventive roles of the drugs, and their structural features may facilitate the design of new anti-cancer agents targeting AKR1B10.


Asunto(s)
Aldehído Reductasa/antagonistas & inhibidores , Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos Fitogénicos/farmacología , Biomarcadores de Tumor/antagonistas & inhibidores , Fenamatos/farmacología , Ácido Glicirretínico/farmacología , Extractos Vegetales/farmacología , Aldo-Ceto Reductasas , Aminoácidos/química , Antiinflamatorios no Esteroideos/química , Antineoplásicos Fitogénicos/química , Diclofenaco/química , Diclofenaco/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Fenamatos/química , Flavonoides/química , Flavonoides/farmacología , Ácido Glicirretínico/química , Humanos , Ácido Mefenámico/química , Ácido Mefenámico/farmacología , Mutación , NADP/química , Extractos Vegetales/química , Especificidad por Sustrato
17.
Mol Pharm ; 6(2): 557-70, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19265444

RESUMEN

Intestinal fatty acid binding protein (I-FABP) is present at high levels in the absorptive cells of the intestine (enterocytes), where it plays a role in the intracellular solubilization of fatty acids (FA). However, I-FABP has also been shown to bind to a range of non-FA ligands, including some lipophilic drug molecules. Thus, in addition to its central role in FA trafficking, I-FABP potentially serves as an important intracellular carrier of lipophilic drugs. In this study we provide a detailed thermodynamic analysis of the binding and stability properties of I-FABP in complex with a series of fibrate and fenamate drugs to provide an insight into the forces driving drug binding to I-FABP. Drug binding and selectivity for I-FABP are driven by the interplay of protein-ligand interactions and solvent processes. The Gibbs free energies (deltaGo) determined from dissociation constants at 25 degrees C ranged from -6.2 to -10 kcal/mol. The reaction energetics indicate that drug binding to I-FABP is an enthalpy-entropy driven process. The relationship between I-FABP stability and drug binding affinity was examined by pulse proteolysis. There is a strong coupling between drug binding and I-FABP stability. The effect of an I-FABP protein sink on the kinetics and thermodynamics of tolfenamic acid permeation across an artificial phospholipid membrane were investigated. I-FABP significantly decreased the energy barrier for desorption of tolfenamic acid from the membrane into the acceptor compartment. Taken together, these data suggest that the formation of stable drug-I-FABP complexes is thermodynamically viable under conditions simulating the reactant concentrations likely observed in vivo and maybe a significant biochemical process that serves as a driving force for passive intestinal absorption of lipophilic drugs.


Asunto(s)
Ácido Clofíbrico/farmacología , Proteínas de Unión a Ácidos Grasos/química , Proteínas de Unión a Ácidos Grasos/metabolismo , Fenamatos/farmacología , Hipolipemiantes/farmacología , Animales , Sitios de Unión , Ácido Clofíbrico/química , Enterocitos/metabolismo , Ácidos Grasos/metabolismo , Fenamatos/química , Hipolipemiantes/química , Absorción Intestinal , Mucosa Intestinal/metabolismo , Membranas Artificiales , Conformación Proteica , Ratas , Termodinámica
18.
Bioorg Med Chem Lett ; 19(3): 654-7, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19121939

RESUMEN

It is believed that beta-amyloid aggregation is an important event in the development of Alzheimer's disease. In the course of our studies to identify beta-amyloid aggregation inhibitors, a series of N-phenyl anthranilic acid analogs were synthesized and studied for beta-amyloid inhibition activity. The synthesis, structure-activity relationship, and in vivo activity of these analogs are discussed.


Asunto(s)
Amiloide/química , Química Farmacéutica/métodos , Fenamatos/química , Enfermedad de Alzheimer , Animales , Modelos Animales de Enfermedad , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Fenamatos/síntesis química , Humanos , Ratones , Microscopía de Fuerza Atómica , Modelos Químicos , Estructura Molecular , Péptidos/química , Relación Estructura-Actividad
19.
Acta Crystallogr C ; 64(Pt 7): o367-71, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18599979

RESUMEN

The structures of 2-[(2,3-dimethylphenyl)carbamoyl]benzenesulfonamide, 2-[(3,4-dimethylphenyl)carbamoyl]benzenesulfonamide and 2-[(2,6-dimethylphenyl)carbamoyl]benzenesulfonamide, all C(15)H(16)N(2)O(3)S, are stabilized by extensive intra- and intermolecular hydrogen bonds. In all three structures, the sulfonamide and carbamoyl groups are involved in hydrogen bonding. In the 2,3-dimethyl and 2,6-dimethyl derivatives, dimeric units and chains of molecules are formed parallel to the c axis. In the 3,4-dimethyl derivative, the hydrogen bonding creates tetrameric units, resulting in macrocyclic R(4)(4)(22) rings that form sheets in the ab plane. The three analogues are closely related to the fenamate class of nonsteroidal anti-inflammatory drugs.


Asunto(s)
Antiinflamatorios no Esteroideos/química , Benzamidas/química , Fenamatos/química , Sulfonamidas/química , Cristalografía , Dimerización , Enlace de Hidrógeno , Estructura Molecular
20.
Artículo en Inglés | MEDLINE | ID: mdl-17433768

RESUMEN

Spectral characteristics of N-phenylanthranilic acid (NPAA) have been studied in different solvents, pH and beta-cyclodextrin (beta-CD) and compared with anthranilic acid (2-aminobenzoic acid, 2ABA). In all solvents a dual fluorescence is observed in NPAA, whereas 2ABA gives single emission. Combining the results observed in the absorption, fluorescence emission and fluorescence excitation spectra, it is found that strong intramolecular hydrogen bonding (IHB) interactions present in NPAA molecule. The inclusion complex of NPAA with beta-CD is analysed by UV-vis, fluorimetry, FT-IR, (1)H NMR, scanning electron microscope and AM 1 method. The above spectral studies show that NPAA forms a 1:1 inclusion complex with beta-CD and COOH group present in the beta-CD cavity. A mechanism is proposed to explain the inclusion process.


Asunto(s)
Fenamatos/química , Solventes/química , beta-Ciclodextrinas/química , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética , Microscopía Electrónica de Rastreo , Protones , Espectrometría de Fluorescencia , Espectroscopía Infrarroja por Transformada de Fourier
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA