Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pestic Biochem Physiol ; 199: 105805, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38458670

RESUMEN

Diquat (DQ) poisoning has garnered attention in recent years, primarily due to the rising incidence of cases worldwide, coupled with the absence of a viable antidote for its treatment. Despite the fact that diquat monopyridone (DQ-M) has been identified as a significant metabolite of DQ, the enzyme responsible for its formation remains unknown. In this study, we have identified aldehyde oxidase (AOX) as a vital enzyme involved in DQ oxidative metabolism. The metabolism of DQ to DQ-M was significantly inhibited by AOX inhibitors including raloxifene and hydralazine. The source of oxygen incorporated into DQ-M was proved to be from water through a H218O incubation experiment which further corroborated DQ-M formation via AOX metabolism. The product of DQ-M in vitro generated by fresh rat tissues co-incubation was consistent with its AOX expression. The result of the molecular docking analysis of DQ and AOX protein showed that DQ is capable of binding to AOX. Furthermore, the cytotoxicity of DQ was significantly higher than DQ-M at the same concentration tested in six cell types. This work is the first to uncover the involvement of aldehyde oxidase, a non-cytochrome P450 enzyme, in the oxidative metabolic pathway of diquat, thus providing a potential target for the development of detoxification treatment.


Asunto(s)
Aldehído Oxidasa , Diquat , Ratas , Animales , Diquat/farmacología , Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Simulación del Acoplamiento Molecular , Estrés Oxidativo , Redes y Vías Metabólicas , Sistema Enzimático del Citocromo P-450/metabolismo
2.
J Chem Theory Comput ; 19(24): 9302-9317, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38085599

RESUMEN

Aldehyde oxidase (AOX) and other related molybdenum-containing enzymes are known to oxidize the C-H bonds of aromatic rings. This process contributes to the metabolism of pharmaceutical compounds and, therefore, is of vital importance to drug pharmacokinetics. The present work describes an automated computational workflow and its use for the prediction of intrinsic reactivity of small aromatic molecules toward a minimal model of the active site of AOX. The workflow is based on quantum chemical transition state searches for the underlying single-step oxidation reaction, where the automated protocol includes identification of unique aromatic C-H bonds, creation of three-dimensional reactant and product complex geometries via a templating approach, search for a transition state, and validation of reaction end points. Conformational search on the reactants, products, and the transition states is performed. The automated procedure has been validated on previously reported transition state barriers and was used to evaluate the intrinsic reactivity of nearly three hundred heterocycles commonly found in approved drug molecules. The intrinsic reactivity of more than 1000 individual aromatic carbon sites is reported. Stereochemical and conformational aspects of the oxidation reaction, which have not been discussed in previous studies, are shown to play important roles in accurate modeling of the oxidation reaction. Observations on structural trends that determine the reactivity are provided and rationalized.


Asunto(s)
Aldehído Oxidasa , Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Dominio Catalítico , Oxidación-Reducción
3.
Eur J Pharm Sci ; 191: 106603, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37827455

RESUMEN

Aldehyde oxidase (AOX) is a cytosolic drug-metabolizing enzyme which has attracted increasing attention in drug development due to its high hepatic expression, broad substrate profile and species differences. In contrast, there is limited information on the presence and activity of AOX in extrahepatic tissues including ocular tissues. Because several ocular drugs are potential substrates for AOX, we performed a comprehensive analysis of the AOX1 expression and activity profile in seven ocular tissues from humans, rabbits, and pigs. AOX activities were determined using optimized assays for the established human AOX1 probe substrates 4-dimethylamino-cinnamaldehyde (DMAC) and phthalazine. Inhibition studies were undertaken in conjunctival and retinal homogenates using well-established human AOX1 inhibitors menadione and chlorpromazine. AOX1 protein contents were quantitated with targeted proteomics and confirmed by immunoblotting. Overall, DMAC oxidation rates varied over 10-fold between species (human ˃˃ rabbit ˃ pig) and showed 2- to 6-fold differences between tissues from the same species. Menadione seemed a more potent inhibitor of DMAC oxidation across species than chlorpromazine. Human AOX1 protein levels were highest in the conjunctiva, followed by most posterior tissues, whereas anterior tissues showed low levels. The rabbit AOX1 expression was high in the conjunctiva, retinal pigment epithelial (RPE), and choroid while lower in the anterior tissues. Quantification of pig AOX1 was not successful but immunoblotting confirmed the presence of AOX1 in all species. DMAC oxidation rates and AOX1 contents correlated quite well in humans and rabbits. This study provides, for the first time, insights into the ocular expression and activity of AOX1 among multiple species.


Asunto(s)
Aldehído Oxidasa , Vitamina K 3 , Humanos , Conejos , Animales , Porcinos , Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Vitamina K 3/metabolismo , Clorpromazina , Oxidación-Reducción , Hígado/metabolismo
4.
Arch Toxicol ; 96(8): 2145-2246, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35648190

RESUMEN

This is an overview of the metabolic reactions of drugs, natural products, physiological compounds, and other (general) chemicals catalyzed by flavin monooxygenase (FMO), monoamine oxidase (MAO), NAD(P)H quinone oxidoreductase (NQO), and molybdenum hydroxylase enzymes (aldehyde oxidase (AOX) and xanthine oxidoreductase (XOR)), including roles as substrates, inducers, and inhibitors of the enzymes. The metabolism and bioactivation of selected examples of each group (i.e., drugs, "general chemicals," natural products, and physiological compounds) are discussed. We identified a higher fraction of bioactivation reactions for FMO enzymes compared to other enzymes, predominately involving drugs and general chemicals. With MAO enzymes, physiological compounds predominate as substrates, and some products lead to unwanted side effects or illness. AOX and XOR enzymes are molybdenum hydroxylases that catalyze the oxidation of various heteroaromatic rings and aldehydes and the reduction of a number of different functional groups. While neither of these two enzymes contributes substantially to the metabolism of currently marketed drugs, AOX has become a frequently encountered route of metabolism among drug discovery programs in the past 10-15 years. XOR has even less of a role in the metabolism of clinical drugs and preclinical drug candidates than AOX, likely due to narrower substrate specificity.


Asunto(s)
Productos Biológicos , Oxidorreductasas , Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Humanos , Molibdeno , Monoaminooxidasa/metabolismo , Oxidorreductasas/metabolismo
5.
Methods Mol Biol ; 2342: 257-284, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34272698

RESUMEN

Aldehyde oxidase (AO) has emerged as an important drug metabolizing enzyme over the last decade. Several compounds have failed in the clinic because the clearance or toxicity was underestimated by preclinical species. Human AO is much more active than rodent AO, and dogs do not have functional AO. Metabolic products from AO-catalyzed oxidation are generally nonreactive and often they have much lower solubility. AO metabolism is not limited to oxidation as AO can also catalyze reduction of oxygen and nitrite. Reduction of oxygen leads to the reactive oxygen species (ROS) superoxide radical anion and hydrogen peroxide. Reduction of nitrite leads to the formation of nitric oxide with potential pharmacological implications. AO is also reported to catalyze the reductive metabolism of nitro-compounds, N-oxides, sulfoxides, isoxazoles, isothiazoles, nitrite, and hydroxamic acids. These reductive transformations may cause toxicity due to the formation of reactive metabolites. Moreover, the inhibition kinetics are complex, and multiple probe substrates should be used when assessing the potential for DDIs. Finally, AO appears to be amenable to computational predictions of both regioselectivity and rates of reaction, which holds promise for virtual screening.


Asunto(s)
Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Inhibidores Enzimáticos/química , Aldehído Oxidasa/antagonistas & inhibidores , Animales , Catálisis , Perros , Diseño de Fármacos , Inhibidores Enzimáticos/farmacocinética , Humanos , Peróxido de Hidrógeno/metabolismo , Modelos Moleculares , Oxidación-Reducción , Conformación Proteica , Relación Estructura-Actividad , Superóxidos/metabolismo
6.
Drug Metab Dispos ; 49(9): 729-735, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34183377

RESUMEN

Mammalian aldehyde oxidases (AOX) are molybdo-flavoenzymes of pharmacological and pathophysiologic relevance that are involved in phase I drug metabolism and, as a product of their enzymatic activity, are also involved in the generation of reactive oxygen species. So far, the physiologic role of aldehyde oxidase 1 in the human body remains unknown. The human enzyme hAOX1 is characterized by a broad substrate specificity, oxidizing aromatic/aliphatic aldehydes into their corresponding carboxylic acids, and hydroxylating various heteroaromatic rings. The enzyme uses oxygen as terminal electron acceptor to produce hydrogen peroxide and superoxide during turnover. Since hAOX1 and, in particular, some natural variants produce not only H2O2 but also high amounts of superoxide, we investigated the effect of both ROS molecules on the enzymatic activity of hAOX1 in more detail. We compared hAOX1 to the high-O2 .--producing natural variant L438V for their time-dependent inactivation with H2O2/O2 .- during substrate turnover. We show that the inactivation of the hAOX1 wild-type enzyme is mainly based on the production of hydrogen peroxide, whereas for the variant L438V, both hydrogen peroxide and superoxide contribute to the time-dependent inactivation of the enzyme during turnover. Further, the level of inactivation was revealed to be substrate-dependent: using substrates with higher turnover numbers resulted in a faster inactivation of the enzymes. Analysis of the inactivation site of the enzyme identified a loss of the terminal sulfido ligand at the molybdenum active site by the produced ROS during turnover. SIGNIFICANCE STATEMENT: This work characterizes the substrate-dependent inactivation of human aldehyde oxidase 1 under turnover by reactive oxygen species and identifies the site of inactivation. The role of ROS in the inhibition of human aldehyde oxidase 1 will have a high impact on future studies.


Asunto(s)
Aldehído Oxidasa , Especificidad por Sustrato/fisiología , Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Dominio Catalítico , Activación Enzimática , Pruebas de Enzimas/métodos , Humanos , Peróxido de Hidrógeno/análisis , Peróxido de Hidrógeno/metabolismo , Inactivación Metabólica/fisiología , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/análisis , Superóxidos/metabolismo
7.
J Insect Sci ; 20(6)2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-33295983

RESUMEN

Aldehyde oxidases (AOXs) are a subfamily of cytosolic molybdo-flavoenzymes that play critical roles in the detoxification and degradation of chemicals. Active AOXs, such as AOX1 and AOX2, have been identified and functionally analyzed in insect antennae but are rarely reported in other tissues. This is the first study to isolate and characterize the cDNA that encodes aldehyde oxidase 5 (BmAOX5) in the pheromone gland (PG) of the silkworm, Bombyx mori. The size of BmAOX5 cDNA is 3,741 nucleotides and includes an open reading frame, which encodes a protein of 1,246 amino acid residues. The theoretical molecular weight and isoelectric point of BmAOX5 are approximately 138 kDa and 5.58, respectively. BmAOX5 shares a similar primary structure with BmAOX1 and BmAOX2, containing two [2Fe-2S] redox centers, a FAD-binding domain, and a molybdenum cofactor (MoCo)-binding domain. RT-PCR revealed BmAOX5 to be particularly highly expressed in the PG (including ovipositor) of the female silkworm moth, and the expression was further confirmed by in situ hybridization, AOX activity staining, and anti-BmAOX5 western blotting. Further, BmAOX5 was shown to metabolize aromatic aldehydes, such as benzaldehyde, salicylaldehyde, and vanillic aldehyde, and fatty aldehydes, such as heptaldehyde and propionaldehyde. The maximum reaction rate (Vmax) of benzaldehyde as substrate was 21 mU and Km was 1.745 mmol/liter. These results suggested that BmAOX5 in the PG could metabolize aldehydes in the cytoplasm for detoxification or participate in the degradation of aldehyde pheromone substances and odorant compounds to identify mating partners and locate suitable spawning sites.


Asunto(s)
Aldehído Oxidasa , Bombyx , Feromonas/metabolismo , Glándulas Odoríferas/metabolismo , Aldehído Oxidasa/química , Aldehído Oxidasa/genética , Aldehído Oxidasa/aislamiento & purificación , Aldehído Oxidasa/metabolismo , Animales , Antenas de Artrópodos/metabolismo , Bombyx/genética , Bombyx/metabolismo , Genes de Insecto , Mariposas Nocturnas/genética , Mariposas Nocturnas/metabolismo
8.
J Pharmacol Exp Ther ; 374(2): 295-307, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32393528

RESUMEN

Gefitinib and erlotinib are epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) with activity against metastatic non-small cell lung cancer. Aldehyde oxidase-1 (AOX1) is a cytosolic drug-metabolizing enzyme. We conducted an experimental and molecular docking study on the effect of gefitinib, erlotinib, and select metabolites on the in vitro catalytic activity of AOX1, as assessed by carbazeran 4-oxidation, and determined the impact of AOX1 inhibition on hepatic metabolism of zaleplon and methotrexate. Gefitinib, desmorpholinopropylgefitinib, erlotinib, desmethylerlotinib, and didesmethylerlotinib inhibited human hepatic cytosolic carbazeran 4-oxidation by a competitive mode, with inhibition constants in submicromolar or low micromolar concentrations. Desmethylgefitinib did not affect AOX1 catalytic activity. A similar pattern was obtained when investigated with human kidney cytosol or recombinant AOX1. The differential effect of gefitinib on human, rat, and mouse hepatic AOX1 catalytic activity suggests species-dependent chemical inhibition of AOX1. Erlotinib was considerably more potent than gefitinib in decreasing hepatic cytosolic zaleplon 5-oxidation and methotrexate 7-oxidation. Molecular docking analyses provided structural insights into the interaction between EGFR-TKIs and AOX1, with key residues and bonds identified, which provided favorable comparison and ranking of potential inhibitors. Based on the US Food and Drug Administration guidance to assess the risk of drug-drug interactions, the calculated R1 values indicate that further investigations are warranted to determine whether gefitinib and erlotinib impact AOX1-mediated drug metabolism in vivo. Overall, erlotinib desmethylerlotinib, didesmethylerlotinib, gefitinib, and desmorpholinopropylgefitinib are potent inhibitors of human AOX1 catalytic function and hepatic metabolism of zaleplon and methotrexate, potentially affecting drug efficacy or toxicity. SIGNIFICANCE STATEMENT: As epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs), gefitinib and erlotinib are first-line pharmacotherapy for metastatic non-small cell lung cancer. Our experimental findings indicate that clinically relevant concentrations of gefitinib, desmorpholinopropylgefitinib, erlotinib, desmethylerlotinib, and didesmethylerlotinib, but not desmethylgefitinib, inhibit human aldehyde oxidase (AOX1) catalytic activity and hepatic cytosolic metabolism of zaleplon and methotrexate. Molecular docking analysis provide structural insights into the key AOX1 interactions with these EGFR-TKIs. Our findings may trigger improved strategies for new EGFR-TKI design and development.


Asunto(s)
Acetamidas/metabolismo , Aldehído Oxidasa/antagonistas & inhibidores , Clorhidrato de Erlotinib/farmacología , Gefitinib/farmacología , Hígado/efectos de los fármacos , Metotrexato/metabolismo , Simulación del Acoplamiento Molecular , Pirimidinas/metabolismo , Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Clorhidrato de Erlotinib/metabolismo , Gefitinib/metabolismo , Humanos , Hígado/metabolismo , Conformación Proteica
9.
J Biol Chem ; 295(16): 5377-5389, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32144208

RESUMEN

Aldehyde oxidases (AOXs) are a small group of enzymes belonging to the larger family of molybdo-flavoenzymes, along with the well-characterized xanthine oxidoreductase. The two major types of reactions that are catalyzed by AOXs are the hydroxylation of heterocycles and the oxidation of aldehydes to their corresponding carboxylic acids. Different animal species have different complements of AOX genes. The two extremes are represented in humans and rodents; whereas the human genome contains a single active gene (AOX1), those of rodents, such as mice, are endowed with four genes (Aox1-4), clustering on the same chromosome, each encoding a functionally distinct AOX enzyme. It still remains enigmatic why some species have numerous AOX enzymes, whereas others harbor only one functional enzyme. At present, little is known about the physiological relevance of AOX enzymes in humans and their additional forms in other mammals. These enzymes are expressed in the liver and play an important role in the metabolisms of drugs and other xenobiotics. In this review, we discuss the expression, tissue-specific roles, and substrate specificities of the different mammalian AOX enzymes and highlight insights into their physiological roles.


Asunto(s)
Aldehído Oxidasa/metabolismo , Evolución Molecular , Hígado/enzimología , Aldehído Oxidasa/química , Aldehído Oxidasa/genética , Animales , Humanos , Especificidad por Sustrato
10.
Anal Chem ; 92(5): 3981-3989, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32037799

RESUMEN

The in vivo monitoring of ascorbic acid (AA) following physiological and pathological events is of great importance because AA plays a critical role in brain functions. The conventional electrochemical sensors (ECSs) usually suffered from poor selectivity and sluggish electron transfer kinetics for cerebral AA oxidation. The exploitation of ECSs adapt to the electrochemical detection (ECD)-microdialysis system, here we reported a facile ratiometric electrochemical sensor (RECS) for in vivo/online repetitive measurements of cerebral AA in brain microdiaysate. The sensor were constructed by careful electrodeposition of graphene oxide (GO) onto glassy carbon (GC) electrodes. Methylene blue (MB) was electrostatically adsorbed onto the GO surface as a built-in reference to achieve ratiometric detection of AA. The subsequent proper electroreduction treatment was able to readily facilitate the oxidation of AA at a relatively negative potential (-100 mV) and the oxidation of MB at separated potential (-428 mV). The in vitro experiments demonstrated that the RECS exhibited high sensitivity (detection limit: 10 nM), selectivity, and stability toward AA determination, enabling the in vivo/online repetitive measurement of cerebral AA in brain microdiaysate with high reliability. As a result, the designed RECS was successfully applied in the ECD-microdialysis system to in vivo/online repetitive monitoring the dynamic change of cerebral AA in the progress of the global cerebral ischemia/reperfusion events. More, the microinjection of endogenous AA and AA oxidase (AAOx) verified the reliability of the proposed RECS for in vivo/online repetitive cerebral AA detection. This proposed sensor filled the gap that no rational electrochemical sensor has been developed for the ECD-microdialysis system since its creation by the Mao group in 2005, which provided a reliable and effective method for brain chemistry research.


Asunto(s)
Ácido Ascórbico/análisis , Encéfalo/metabolismo , Técnicas Electroquímicas/métodos , Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Animales , Diálisis , Electrodos , Grafito/química , Límite de Detección , Masculino , Azul de Metileno/química , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley
11.
Drug Metab Rev ; 51(4): 428-452, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31549868

RESUMEN

Human AOX1 is a member of the mammalian aldehyde oxidase (AOX) family of enzymes and it is an emerging cytosolic enzyme involved in phase I drug-metabolism, bio-transforming a number of therapeutic agents and xenobiotics. The current trend in drug-development is to design molecules which are not recognized and inactivated by CYP450 monooxygenases, the main drug-metabolizing system, to generate novel therapeutic agents characterized by optimal pharmacokinetic and pharmacodynamic properties. Unfortunately, this has resulted in a substantial enrichment in molecules which are recognized and metabolized by AOXs. The observation has raised interest in the generation of tools capable of predicting AOX-dependent drug-metabolism of novel molecules during the early phases of drug development. Such tools are likely to reduce the number of failures occurring at the clinical and late phase of the drug development process. The current review describes different in silico, in vitro and in vivo methods for the prediction of AOX metabolizing ability and focuses on the existing drawbacks and challenges associated with these approaches.


Asunto(s)
Aldehído Oxidasa/metabolismo , Preparaciones Farmacéuticas/metabolismo , Aldehído Oxidasa/antagonistas & inhibidores , Aldehído Oxidasa/química , Animales , Evaluación Preclínica de Medicamentos , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Especificidad de la Especie
12.
J Pharmacol Exp Ther ; 371(1): 75-86, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31289113

RESUMEN

Tamoxifen, raloxifene, and nafoxidine are selective estrogen receptor modulators (SERMs) reported to inhibit the catalytic activity of human aldehyde oxidase 1 (AOX1). How these drugs interact with AOX1 and whether other SERMs inhibit this drug-metabolizing enzyme are not known. Therefore, a detailed in vitro and in silico study involving parent drugs and their analogs was conducted to investigate the effect of specific SERMs, particularly acolbifene, bazedoxifene, and lasofoxifene on AOX1 catalytic activity, as assessed by carbazeran 4-oxidation, an AOX1-selective catalytic marker. The rank order in the potency (based on IC50 values) of AOX1 inhibition by SERMs was raloxifene > bazedoxifene ∼ lasofoxifene > tamoxifen > acolbifene. Inhibition of liver cytosolic AOX1 by bazedoxifene, lasofoxifene, and tamoxifen was competitive, whereas that by raloxifene was noncompetitive. Loss of 1-azepanylethyl group increased the inhibitory potency of bazedoxifene, whereas the N-oxide group decreased it. The 7-hydroxy group and the substituted pyrrolidine ring attached to the tetrahydronaphthalene structure contributed to AOX1 inhibition by lasofoxifene. These results are supported by molecular-docking simulations in terms of predicted binding modes, encompassing binding orientation and efficiency, and analysis of key interactions, particularly hydrogen bonds. The extent of AOX1 inhibition by bazedoxifene was increased by estrone sulfate and estrone. In summary, SERMs differentially inhibited human AOX1 catalytic activity. Structural features of bazedoxifene and lasofoxifene contributed to AOX1 inhibition, whereas those of acolbifene rendered it considerably less susceptible to AOX1 inhibition. Overall, our novel biochemical findings and molecular-docking analyses provide new insights into the interaction between SERMs and AOX1. SIGNIFICANCE STATEMENT: Aldehyde oxidase (AOX1) is a molybdo-flavoprotein and has emerged as a drug-metabolizing enzyme of potential therapeutic importance because drugs have been identified as AOX1 substrates. Selective estrogen receptor modulators (SERM), which are drugs used to treat and prevent various conditions, differentially inhibit AOX1 catalytic activity. Structural features of bazedoxifene and lasofoxifene contribute to AOX1 inhibition, whereas those of acolbifene render it considerably less susceptible to AOX1 inhibition. Our novel biochemical findings, together with molecular- docking analyses, provide new insights into the differential inhibitory effect of SERMs on the catalytic activity of human AOX1, how SERMs bind to AOX1, and increase our understanding of the AOX1 pharmacophore in the inhibition of AOX1 by drugs and other chemicals.


Asunto(s)
Aldehído Oxidasa/antagonistas & inhibidores , Indoles/farmacología , Simulación del Acoplamiento Molecular , Pirrolidinas/farmacología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tetrahidronaftalenos/farmacología , Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Sitios de Unión , Femenino , Humanos , Hígado/enzimología , Masculino , Unión Proteica
13.
Drug Metab Dispos ; 47(8): 874-882, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31182423

RESUMEN

(R)-2-(2-methylimidazo[2,1-b]thiazol-6-yl)-1-(2-(5-(6-methylpyrimidin-4-yl)-2,3-dihydro-1H-inden-1-yl)-2,7-diazaspiro[3.5]nonan-7-yl)ethan-1-one (PF-5190457) was identified as a potent and selective inverse agonist of the ghrelin receptor [growth hormone secretagogue receptor 1a (GHS-R1a)]. The present translational bed-to-bench work characterizes the biotransformation of this compound in vivo and then further explores in vitro metabolism in fractions of human liver and primary hepatocytes. Following oral administration of PF-5190457 in a phase 1b clinical study, hydroxyl metabolites of the compound were observed, including one that had not been observed in previously performed human liver microsomal incubations. PF-6870961 was biosynthesized using liver cytosol, and the site of hydroxylation was shown to be on the pyrimidine using nuclear magnetic resonance spectroscopy. The aldehyde oxidase (AO) inhibitor raloxifene and the xanthine oxidase inhibitor febuxostat inhibited the formation of PF-6870961 in human liver cytosol, suggesting both enzymes were involved in the metabolism of the drug. However, greater inhibition was observed with raloxifene, indicating AO is a dominant enzyme in the biotransformation. The intrinsic clearance of the drug in human liver cytosol was estimated to be 0.002 ml/min per milligram protein. This study provides important novel information at three levels: 1) it provides additional new information on the recently developed novel compound PF-5190457, the first GHS-R1a blocker that has moved to development in humans; 2) it provides an example of a reverse translational approach where a discovery in humans was brought back, validated, and further investigated at the bench level; and 3) it demonstrates the importance of considering the molybdenum-containing oxidases during the development of new drug entities. SIGNIFICANCE STATEMENT: PF-5190457 is a novel ghrelin receptor inverse agonist that is currently undergoing clinical development for treatment of alcohol use disorder. PF-6870961, a major hydroxyl metabolite of the compound, was observed in human plasma, but was absent in human liver microsomal incubations. PF-6870961 was biosynthesized using liver cytosol, and the site of hydroxylation on the pyrimidine ring was characterized. Inhibitors of aldehyde oxidase and xanthine oxidase inhibited the formation of PF-6870961 in human liver cytosol, suggesting both enzymes were involved in the metabolism of the drug. This information is important for patient selection in subsequent clinical studies.


Asunto(s)
Aldehído Oxidasa/metabolismo , Azetidinas/farmacocinética , Hígado/metabolismo , Receptores de Ghrelina/antagonistas & inhibidores , Compuestos de Espiro/farmacocinética , Xantina Oxidasa/metabolismo , Administración Oral , Alcoholismo/tratamiento farmacológico , Aldehído Oxidasa/antagonistas & inhibidores , Aldehído Oxidasa/química , Animales , Azetidinas/administración & dosificación , Biotransformación/efectos de los fármacos , Citosol/metabolismo , Febuxostat/farmacología , Femenino , Ghrelina/antagonistas & inhibidores , Hepatocitos/metabolismo , Humanos , Hígado/citología , Ratones , Microsomas Hepáticos , Molibdeno/química , Clorhidrato de Raloxifeno/farmacología , Compuestos de Espiro/administración & dosificación , Xantina Oxidasa/antagonistas & inhibidores , Xantina Oxidasa/química
14.
Phys Chem Chem Phys ; 21(25): 13545-13554, 2019 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-31172995

RESUMEN

Human aldehyde oxidase (hAOX1) is a molybdenum dependent enzyme that plays an important role in the metabolism of various compounds either endogenous or xenobiotics. Due to its promiscuity, hAOX1 plays a major role in the pharmacokinetics of many drugs and therefore has gathered a lot of attention from the scientific community and, particularly, from the pharmaceutical industry. In this work, homology modelling, molecular docking and molecular dynamics simulations were used to study the structure of the monomer and dimer of human AOX. The results with the monomer of hAOX1 allowed to shed some light on the role played by thioridazine and two malonate ions that are co-crystalized in the recent X-ray structure of hAOX1. The results show that these molecules endorse several conformational rearrangements in the binding pocket of the enzyme and these changes have an impact in the active site topology as well as in the stability of the substrate (phthalazine). The results show that the presence of both molecules open two gates located at the entrance of the binding pocket, from which results the flooding of the active site. They also endorse several modifications in the shape of the binding pocket (namely the position of Lys893) that, together with the presence of the solvent molecules, favour the release of the substrate to the solvent. Further insights were also obtained with the assembled homodimer of hAOX1. The allosteric inhibitor (THI) binds closely to the region where the dimerization of both monomers occur. These findings suggest that THI can interfere with protein dimerization.


Asunto(s)
Aldehído Oxidasa/química , Dominio Catalítico , Cristalización , Humanos , Cinética , Malonatos/química , Modelos Moleculares , Ftalazinas/química , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Solventes , Tioridazina/química
15.
FEBS Open Bio ; 9(5): 925-934, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30985987

RESUMEN

Human aldehyde oxidase (hAOX1) is a molybdenum enzyme with high toxicological importance, but its physiological role is still unknown. hAOX1 metabolizes different classes of xenobiotics and is one of the main drug-metabolizing enzymes in the liver, along with cytochrome P450. hAOX1 oxidizes and inactivates a large number of drug molecules and has been responsible for the failure of several phase I clinical trials. The interindividual variability of drug-metabolizing enzymes caused by single nucleotide polymorphisms (SNPs) is highly relevant in pharmaceutical treatments. In this study, we present the crystal structure of the inactive variant G1269R, revealing the first structure of a molybdenum cofactor (Moco)-free form of hAOX1. These data allowed to model, for the first time, the flexible Gate 1 that controls access to the active site. Furthermore, we inspected the thermostability of wild-type hAOX1 and hAOX1 with various SNPs (L438V, R1231H, G1269R or S1271L) by CD spectroscopy and ThermoFAD, revealing that amino acid exchanges close to the Moco site can impact protein stability up to 10 °C. These results correlated with biochemical and structural data and enhance our understanding of hAOX1 and the effect of SNPs in the gene encoding this enzyme in the human population. ENZYMES: Aldehyde oxidase (EC1.2.3.1); xanthine dehydrogenase (EC1.17.1.4); xanthine oxidase (EC1.1.3.2). DATABASES: Structural data are available in the Protein Data Bank under the accession number 6Q6Q.


Asunto(s)
Aldehído Oxidasa/química , Polimorfismo de Nucleótido Simple , Coenzimas , Cristalografía por Rayos X , Humanos , Metaloproteínas , Modelos Moleculares , Cofactores de Molibdeno , Pteridinas
16.
Biosci Biotechnol Biochem ; 83(1): 129-136, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30286706

RESUMEN

Auxin is thought to be an important factor in the induction of galls by galling insects. We have previously shown that both galling and nongalling insects synthesize indole-3-acetic acid (IAA) from tryptophan (Trp) via two intermediates, indole-3-acetaldoxime (IAOx) and indole-3-acetaldehyde (IAAld). In this study, we isolated an enzyme that catalyzes the last step "IAAld → IAA" from a silk-gland extract of Bombyx mori. The enzyme, designated "BmIAO1", contains two 2Fe-2S iron-sulfur-cluster-binding domains, an FAD-binding domain, and a molybdopterin-binding domain, which are conserved in aldehyde oxidases. BmIAO1 causes the nonenzymatic conversion of Trp to IAAld and the enzymatic conversion of IAOx to IAA, suggesting that BmIAO1 alone is responsible for IAA production in B. mori. However, a detailed comparison of pure BmIAO1 and the crude silk-gland extract suggested the presence of other enzymes involved in IAA production from Trp. Abbreviations: BA: benzoic acid; CE: collision energy; CXP: collision cell exit potential; DP: declustering potential; IAA: indole-3-acetic acid; IBI1: IAA biosynthetic inhibitor-1; IAAld: indole-3-acetaldehyde; ICA: indole-3-carboxylic acid; IAOx: indole-3-acetaldoxime; IEtOH: indole-3-ethanol; LC-MS/MS: liquid chromatography-tandem mass spectrometry; Trp: tryptophan.


Asunto(s)
Aldehído Oxidasa/metabolismo , Bombyx/fisiología , Ácidos Indolacéticos/metabolismo , Reguladores del Crecimiento de las Plantas/metabolismo , Aldehído Oxidasa/química , Aldehído Oxidasa/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Bombyx/anatomía & histología , Bombyx/enzimología , Catálisis , Cromatografía/métodos , Electroforesis en Gel de Poliacrilamida , Homología de Secuencia de Aminoácido , Espectrofotometría Ultravioleta/métodos , Especificidad por Sustrato , Espectrometría de Masas en Tándem/métodos
17.
Eur J Drug Metab Pharmacokinet ; 44(2): 275-286, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30382490

RESUMEN

BACKGROUND: Aldehyde oxidase (AOX) is an important molybdenum-containing enzyme with high similarity with xanthine oxidase (XO). AOX involved in the metabolism of a large array of aldehydes and N-heterocyclic compounds and its activity is highly substrate-dependent. OBJECTIVES: The aim of this work was to study the effect of five important phenothiazine drugs on AOX activity using benzaldehyde and phenanthridine as aldehyde and N-heterocyclic substrates, respectively. METHODS: The effect of trifluperazine, chlorpromazine, perphenazine, thioridazine and promethazine on rat liver AOX was measured spectrophotometrically. To predict the mode of interactions between the studied compounds and AOX, a combination of homology modeling and a molecular docking study was performed. RESULTS: All phenothiazines could inhibit AOX activity measured either by phenanthridine or benzaldehyde with almost no effect on XO activity. In the case of benzaldehyde oxidation, the lowest and highest half-maximal inhibitory concentration (IC50) values were obtained for promethazine (IC50 = 0.9 µM), and trifluoperazine (IC50 = 3.9 µM), respectively; whereas perphenazine (IC50 = 4.3 µM), and trifluoperazine (IC50 = 49.6 µM) showed the strongest and weakest inhibitory activity against AOX-catalyzed phenanthridine oxidation, respectively. The in silico findings revealed that the binding site of thioridazine is near the dimer interference, and that hydrophobic interactions are of great importance in all the tested phenothiazines. CONCLUSION: The five studied phenothiazine drugs showed dual inhibitory effects on AOX activity towards aldehydes and N-heterocycles as two major classes of enzyme substrates. Most of the interactions between the phenothiazine-related drugs and AOX in the binding pocket showed a hydrophobic nature.


Asunto(s)
Aldehído Oxidasa/antagonistas & inhibidores , Aldehído Oxidasa/metabolismo , Aldehídos/metabolismo , Simulación por Computador , Compuestos Heterocíclicos/metabolismo , Fenotiazinas/metabolismo , Aldehído Oxidasa/química , Secuencia de Aminoácidos , Animales , Sitios de Unión/fisiología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Masculino , Simulación del Acoplamiento Molecular/métodos , Fenotiazinas/química , Fenotiazinas/farmacología , Estructura Secundaria de Proteína , Ratas , Ratas Wistar
18.
FEBS Lett ; 592(12): 2126-2139, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29749013

RESUMEN

Nitric oxide (NO) is an important gasotransmitter involved in numerous intra- and intercellular signaling events. In addition to the oxidative pathway of NO generation, which includes three NO synthase (NOS) isoforms in mammals, a reductive pathway contributes to NO generation. In this pathway, nitrite is reduced to NO by various metal-containing proteins. Among these, all members of the eukaryotic molybdenum (Mo)-dependent enzyme family were found to be able to reduce nitrite to NO. This Review focuses on the current state of research in the field of Mo-dependent nitrite reduction in eukaryotes. An overview on the five eukaryotic Mo-enzymes is given, and similarities as well as differences in their nitrite reduction mechanisms are presented and discussed in the context of physiological relevance.


Asunto(s)
Vías Biosintéticas , Molibdeno/química , Óxido Nítrico/biosíntesis , Nitritos/química , Aldehído Oxidasa/química , Aldehído Oxidasa/metabolismo , Animales , Humanos , Nitrato-Reductasa/química , Nitrato-Reductasa/metabolismo , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Conformación Proteica , Sulfito-Oxidasa/química , Sulfito-Oxidasa/metabolismo , Xantina Deshidrogenasa/metabolismo
19.
J Biochem ; 163(4): 321-328, 2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29319807

RESUMEN

Hetero-trimeric aldehyde oxidases of bacterial origin, which use O2 to catalyse the oxidation of various aldehydes but not those of aromatic N-heterocycles, belong to the xanthine oxidase family. In the present study, the crystal structure of a recombinant aldehyde oxidase from Methylobacillus sp. KY4400 (Mb-AOX) was determined at 2.5 Å resolution. The structures of its subunits resemble those of the corresponding subunits or domains of other structurally characterised enzymes belonging to the family, and include a [4Fe-4 S] cluster in the medium subunit like that found in Escherichia coli periplasmic aldehyde oxidoreductase (EP-AOR). A funnel leading to the si-face of the isoalloxazine ring of FAD, which is narrower than those in mouse liver AOX3 and human AOX1, is also present and it is even narrower than that in EP-AOR. The environment surrounding the ring in Mb-AOX and EP-AOR is subtly different, which might account for their different abilities to use O2. A remarkable characteristic of the Mo catalytic centre in Mb-AOX is a tryptophan situated near the centre instead of the alanine present in other xanthine oxidase family members. The tryptophan residue together with other residue differences might play an important role in binding to aldehydes such as n-heptylaldehyde in Mb-AOX.


Asunto(s)
Aldehído Oxidasa/química , Methylobacillus/enzimología , Aldehído Oxidasa/aislamiento & purificación , Secuencia de Aminoácidos , Cristalografía por Rayos X , Modelos Moleculares , Conformación Proteica , Alineación de Secuencia
20.
PLoS One ; 13(1): e0191819, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29370288

RESUMEN

Mammalian aldehyde oxidases (AOXs) are molybdo-flavoenzymes which are present in many tissues in various mammalian species, including humans and rodents. Different species contain a different number of AOX isoforms. In particular, the reasons why mammals other than humans express a multiplicity of tissue-specific AOX enzymes is unknown. In mouse, the isoforms mAOX1, mAOX3, mAOX4 and mAOX2 are present. We previously established a codon-optimized heterologous expression systems for the mAOX1-4 isoforms in Escherichia coli that gives yield to sufficient amounts of active protein for kinetic characterizations and sets the basis in this study for site-directed mutagenesis and structure-function studies. A direct and simultaneous comparison of the enzymatic properties and characteristics of the four enzymes on a larger number of substrates has never been performed. Here, thirty different structurally related aromatic, aliphatic and N-heterocyclic compounds were used as substrates, and the kinetic parameters of all four mAOX enzymes were directly compared. The results show that especially mAOX4 displays a higher substrate selectivity, while no major differences between mAOX1, mAOX2 and mAOX3 were identified. Generally, mAOX1 was the enzyme with the highest catalytic turnover for most substrates. To understand the factors that contribute to the substrate specificity of mAOX4, site-directed mutagenesis was applied to substitute amino acids in the substrate-binding funnel by the ones present in mAOX1, mAOX3, and mAOX2. An increase in activity was obtained by the amino acid exchange M1088V in the active site identified to be specific for mAOX4, to the amino acid identified in mAOX3.


Asunto(s)
Aldehído Oxidasa/genética , Aldehído Oxidasa/metabolismo , Aldehído Oxidasa/química , Aldehído Oxidorreductasas/química , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Aldehídos/química , Aldehídos/metabolismo , Sustitución de Aminoácidos , Animales , Benzaldehídos/química , Benzaldehídos/metabolismo , Dominio Catalítico/genética , Flavoproteínas/química , Flavoproteínas/genética , Flavoproteínas/metabolismo , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/metabolismo , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...