Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Molecules ; 28(17)2023 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-37687214

RESUMEN

The mitochondrial amidoxime reducing component (mARC) is a human molybdoenzyme known to catalyze the reduction of various N-oxygenated substrates. The physiological function of mARC enzymes, however, remains unknown. In this study, we examine the reduction of hydrogen peroxide (H2O2) by the human mARC1 and mARC2 enzymes. Furthermore, we demonstrate an increased sensitivity toward H2O2 for HEK-293T cells with an MTARC1 knockout, which implies a role of mARC enzymes in the cellular response to oxidative stress. H2O2 is a reactive oxygen species (ROS) formed in all living cells involved in many physiological processes. Furthermore, H2O2 constitutes the first mARC substrate without a nitrogen-oxygen bond, implying that mARC enzymes may have a substrate spectrum going beyond the previously examined N-oxygenated compounds.


Asunto(s)
Peróxido de Hidrógeno , Oximas , Humanos , Oximas/farmacología , Mitocondrias , Catálisis
2.
Proc Natl Acad Sci U S A ; 115(47): 11958-11963, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30397129

RESUMEN

Biotransformation enzymes ensure a viable homeostasis by regulating reversible cycles of oxidative and reductive reactions. The metabolism of nitrogen-containing compounds is of high pharmaceutical and toxicological relevance because N-oxygenated metabolites derived from reactions mediated by cytochrome P450 enzymes or flavin-dependent monooxygenases are in some cases highly toxic or mutagenic. The molybdenum-dependent mitochondrial amidoxime-reducing component (mARC) was found to be an extremely efficient counterpart, which is able to reduce the full range of N-oxygenated compounds and thereby mediates detoxification reactions. However, the 3D structure of this enzyme was unknown. Here we present the high-resolution crystal structure of human mARC. We give detailed insight into the coordination of its molybdenum cofactor (Moco), the catalytic mechanism, and its ability to reduce a wide range of N-oxygenated compounds. The identification of two key residues will allow future discrimination between mARC paralogs and ensure correct annotation. Since our structural findings contradict in silico predictions that are currently made by online databases, we propose domain definitions for members of the superfamily of Moco sulfurase C-terminal (MOSC) domain-containing proteins. Furthermore, we present evidence for an evolutionary role of mARC for the emergence of the xanthine oxidase protein superfamily. We anticipate the hereby presented crystal structure to be a starting point for future descriptions of MOSC proteins, which are currently poorly structurally characterized.


Asunto(s)
Proteínas Mitocondriales/química , Proteínas Mitocondriales/ultraestructura , Oxidorreductasas/química , Oxidorreductasas/ultraestructura , Catálisis , Coenzimas , Cristalografía por Rayos X/métodos , Células Eucariotas/metabolismo , Humanos , Metaloproteínas , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Molibdeno/metabolismo , Cofactores de Molibdeno , Oxidación-Reducción , Oxidorreductasas/metabolismo , Estructura Terciaria de Proteína , Pteridinas
3.
Drug Metab Dispos ; 46(10): 1396-1402, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30045842

RESUMEN

The mitochondrial amidoxime reducing component is a recently discovered molybdenum enzyme in mammals which, in concert with the electron transport proteins cytochrome b5 and NADH cytochrome b5 reductase, catalyzes the reduction of N-oxygenated structures. This three component enzyme system plays a major role in N-reductive drug metabolism. Belonging to the group of N-hydroxylated structures, hydroxamic acids are also potential substrates of the mARC-system. Hydroxamic acids show a variety of pharmacological activities and are therefore often found in drug candidates. They can also exhibit toxic properties as is the case for many aryl hydroxamic acids formed during the metabolism of arylamides. Biotransformation assays using recombinant human proteins, subcellular porcine tissue fractions as well as human cell culture were performed. Here the mARC-dependent reduction of the model compound benzhydroxamic acid is reported in addition to the reduction of three drugs. In comparison with other known substrates of the molybdenum depending enzyme system (e.g., amidoxime prodrugs) the conversion rates measured here are slower, thereby reflecting the mediocre metabolic stability and oral bioavailability of distinct hydroxamic acids. Moreover, the toxic N-hydroxylated metabolite of the analgesic phenacetin, N-hydroxyphenacetin, is not reduced by the mARC-system under the chosen conditions. This confirms the high toxicity of this component, as it needs to be detoxified by other pathways. This work highlights the need to monitor the N-reductive metabolism of new drug candidates by the mARC-system when evaluating the metabolic stability of hydroxamic acid-containing structures or the potential risks of toxic metabolites.


Asunto(s)
Ácidos Hidroxámicos/metabolismo , Proteínas Mitocondriales/metabolismo , Oxidorreductasas/metabolismo , Animales , Biotransformación , Citocromo-B(5) Reductasa/metabolismo , Citocromos b5/metabolismo , Humanos , Oxidación-Reducción , Receptor EphB3 , Porcinos
4.
J Med Chem ; 63(12): 6538-6546, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31790578

RESUMEN

For the development of new drugs, the investigation of their metabolism is of central importance. In the past, the focus was mostly on the consideration of established enzymes leading to oxidations such as cytochrome P450. However, reductive metabolism by the mARC enzyme system can play an important role in particular for nitrogen containing functional groups. A rapid test was established to give developers of new drugs in the preclinical stage the opportunity to test the metabolism by mARC. To demonstrate the relevance and validity of the new test system, known and potential substrates were applied to this new assay. All known substrates could be detected by the system. Furthermore, several new substrates were found including long-established drugs such as hydroxyurea and new compounds in development such as epacdadostat.


Asunto(s)
Bioensayo/métodos , Inactivación Metabólica , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Oxidorreductasas/metabolismo , Oximas/metabolismo , Humanos , Tasa de Depuración Metabólica , Oxidación-Reducción , Especificidad por Sustrato
5.
Acta Crystallogr D Struct Biol ; 74(Pt 5): 422-432, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29717713

RESUMEN

The high-resolution crystal structure of the flavin-dependent monooxygenase (FMO) from the African locust Zonocerus variegatus is presented and the kinetics of structure-based protein variants are discussed. Z. variegatus expresses three flavin-dependent monooxygenase (ZvFMO) isoforms which contribute to a counterstrategy against pyrrolizidine alkaloids (PAs). PAs are protoxic compounds produced by some angiosperm lineages as a chemical defence against herbivores. N-Oxygenation of PAs and the accumulation of PA N-oxides within their haemolymph result in two evolutionary advantages for these insects: (i) they circumvent the defence mechanism of their food plants and (ii) they can use PA N-oxides to protect themselves against predators, which cannot cope with the toxic PAs. Despite a high degree of sequence identity and a similar substrate spectrum, the three ZvFMO isoforms differ greatly in enzyme activity. Here, the crystal structure of the Z. variegatus PA N-oxygenase (ZvPNO), the most active ZvFMO isoform, is reported at 1.6 Šresolution together with kinetic studies of a second isoform, ZvFMOa. This is the first available crystal structure of an FMO from class B (of six different FMO subclasses, A-F) within the family of flavin-dependent monooxygenases that originates from a more highly developed organism than yeast. Despite the differences in sequence between family members, their overall structure is very similar. This indicates the need for high conservation of the three-dimensional structure for this type of reaction throughout all kingdoms of life. Nevertheless, this structure provides the closest relative to the human enzyme that is currently available for modelling studies. Of note, the crystal structure of ZvPNO reveals a unique dimeric arrangement as well as small conformational changes within the active site that have not been observed before. A newly observed kink within helix α8 close to the substrate-binding path might indicate a potential mechanism for product release. The data show that even single amino-acid exchanges in the substrate-entry path, rather than the binding site, have a significant impact on the specific enzyme activity of the isoforms.


Asunto(s)
Saltamontes/enzimología , Oxigenasas de Función Mixta/química , Alcaloides de Pirrolicidina/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Cinética , Isoformas de Proteínas/química
6.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 6): 337-344, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29870017

RESUMEN

The human mitochondrial amidoxime reducing component (hmARC) is a molybdenum cofactor-dependent enzyme that is involved in the reduction of a diverse range of N-hydroxylated compounds of either physiological or xenobiotic origin. In this study, the use of a fusion-protein approach with T4 lysozyme (T4L) to determine the structure of this hitherto noncrystallizable enzyme by X-ray crystallography is described. A set of four different hmARC-T4L fusion proteins were designed. Two of them contained either an N-terminal or a C-terminal T4L moiety fused to hmARC, while the other two contained T4L as an internal fusion partner tethered to the hmARC enzyme between two predicted secondary-structure elements. One of these internal fusion constructs could be expressed and crystallized successfully. The hmARC-T4L crystals diffracted to 1.7 Šresolution using synchrotron radiation and belonged to space group P212121 with one molecule in the asymmetric unit. Initial attempts to solve the structure by molecular replacement using T4L did not result in electron-density distributions that were sufficient for model building and interpretation of the hmARC moiety. However, this study emphasizes the utility of the T4L fusion-protein approach, which can be used for the crystallization and structure determination of membrane-bound proteins as well as soluble proteins.


Asunto(s)
Coenzimas/química , Metaloproteínas/química , Proteínas Mitocondriales/química , Muramidasa/química , Oxidorreductasas/química , Fragmentos de Péptidos/química , Pteridinas/química , Secuencia de Aminoácidos , Coenzimas/genética , Cristalización/métodos , Humanos , Metaloproteínas/genética , Proteínas Mitocondriales/genética , Cofactores de Molibdeno , Muramidasa/genética , Oxidorreductasas/genética , Fragmentos de Péptidos/genética , Difracción de Rayos X/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA