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











Base de dados
Intervalo de ano de publicação
1.
J Org Chem ; 79(13): 5939-47, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24611689

RESUMO

The high diastereoselectivity of the hydrogenation of artemisinate by diazene to form dihydroartemisinate (diastereoselective ratio, dr, 97:3) necessary for efficient production of artemisin has been rationalized by state-of-the-art DFT calculations and identification of the noncovalent interactions by coupled ELF/NCI analysis. Remarkably, a single conformer of artemisinate is responsible for the high diastereoselectivity of the reaction. NMR studies confirm the preference for a single conformation that is found to be identical to that predicted by the calculations. The calculations and ELF/NCI analyses show that the hydrogenation of the exocyclic activated C═C double bond has a low energy barrier and that the lowest transition state and the preferred conformation of free artemisinate develop the same network of weak noncovalent interactions between the electron donor groups (oxygen and exocyclic C═C double bond) and CH bonds of the cis-decalene group of the artemisinate, which rationalize the high diastereoselectivity unusual for a strongly exothermic reaction.


Assuntos
Artemisininas/química , Imidas/química , Oxigênio/química , Ligação de Hidrogênio , Espectroscopia de Ressonância Magnética , Conformação Molecular , Estereoisomerismo
2.
PLoS One ; 6(5): e19013, 2011 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-21589929

RESUMO

BACKGROUND: Rasburicase (Fasturtec® or Elitek®, Sanofi-Aventis), the recombinant form of urate oxidase from Aspergillus flavus, is a therapeutic enzyme used to prevent or decrease the high levels of uric acid in blood that can occur as a result of chemotherapy. It is produced by Sanofi-Aventis and currently purified via several standard steps of chromatography. This work explores the feasibility of replacing one or more chromatography steps in the downstream process by a crystallization step. It compares the efficacy of two crystallization techniques that have proven successful on pure urate oxidase, testing them on impure urate oxidase solutions. METHODOLOGY/PRINCIPAL FINDINGS: Here we investigate the possibility of purifying urate oxidase directly by crystallization from the fermentation broth. Based on attractive interaction potentials which are known to drive urate oxidase crystallization, two crystallization routes are compared: a) by increased polymer concentration, which induces a depletion attraction and b) by decreased salt concentration, which induces attractive interactions via a salting-in effect. We observe that adding polymer, a very efficient way to crystallize pure urate oxidase through the depletion effect, is not an efficient way to grow crystals from impure solution. On the other hand, we show that dialysis, which decreases salt concentration through its strong salting-in effect, makes purification of urate oxidase from the fermentation broth possible. CONCLUSIONS: The aim of this study is to compare purification efficacy of two crystallization methods. Our findings show that crystallization of urate oxidase from the fermentation broth provides purity comparable to what can be achieved with one chromatography step. This suggests that, in the case of urate oxidase, crystallization could be implemented not only for polishing or concentration during the last steps of purification, but also as an initial capture step, with minimal changes to the current process.


Assuntos
Cristalização , Urato Oxidase/isolamento & purificação , Cromatografia em Gel , Fermentação , Focalização Isoelétrica , Sais , Urato Oxidase/química
3.
Proteins ; 79(6): 1964-76, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21491497

RESUMO

Urate oxidase (EC 1.7.3.3 or UOX) catalyzes the conversion of uric acid using gaseous molecular oxygen to 5-hydroxyisourate and hydrogen peroxide in absence of any cofactor or transition metal. The catalytic mechanism was investigated using X-ray diffraction, electron spin resonance spectroscopy (ESR), and quantum mechanics calculations. The X-ray structure of the anaerobic enzyme-substrate complex gives credit to substrate activation before the dioxygen fixation in the peroxo hole, where incoming and outgoing reagents (dioxygen, water, and hydrogen peroxide molecules) are handled. ESR spectroscopy establishes the initial monoelectron activation of the substrate without the participation of dioxygen. In addition, both X-ray structure and quantum mechanic calculations promote a conserved base oxidative system as the main structural features in UOX that protonates/deprotonates and activate the substrate into the doublet state now able to satisfy the Wigner's spin selection rule for reaction with molecular oxygen in its triplet ground state.


Assuntos
Aspergillus flavus/enzimologia , Urato Oxidase/química , Urato Oxidase/metabolismo , Aspergillus flavus/química , Cristalografia por Raios X , Espectroscopia de Ressonância de Spin Eletrônica , Modelos Moleculares , Ligação Proteica , Teoria Quântica , Ácido Úrico/química , Ácido Úrico/metabolismo
4.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 6): 714-24, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20516624

RESUMO

Urate oxidase (uricase; EC 1.7.3.3; UOX) from Aspergillus flavus catalyzes the oxidation of uric acid in the presence of molecular oxygen to 5-hydroxyisourate in the degradation cascade of purines; intriguingly, catalysis proceeds using neither a metal ion (Fe, Cu etc.) nor a redox cofactor. UOX is a tetrameric enzyme with four active sites located at the interface of two subunits; its structure was refined at atomic resolution (1 A) using new crystal data in the presence of xanthine and at near-atomic resolution (1.3-1.7 A) in complexes with the natural substrate (urate) and two inhibitors: 8-nitroxanthine and 8-thiouric acid. Three new features of the structural and mechanistic behaviour of the enzyme were addressed. Firstly, the high resolution of the UOX-xanthine structure allowed the solution of an old structural problem at a contact zone within the tetramer; secondly, the protonation state of the substrate was determined from both a halochromic inhibitor complex (UOX-8-nitroxanthine) and from the H-atom distribution in the active site, using the structures of the UOX-xanthine and the UOX-uric acid complexes; and thirdly, it was possible to extend the general base system, characterized by the conserved catalytic triad Thr-Lys-His, to a large water network that is able to buffer and shuttle protons back and forth between the substrate and the peroxo hole along the reaction pathway.


Assuntos
Aspergillus flavus/enzimologia , Prótons , Urato Oxidase/química , Ácido Úrico/química , Cristalografia por Raios X , Ligantes , Modelos Moleculares , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Especificidade por Substrato , Urato Oxidase/metabolismo , Ácido Úrico/análogos & derivados , Ácido Úrico/metabolismo
5.
Bioorg Chem ; 37(4): 111-25, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19539344

RESUMO

Urate oxidase catalyzes the transformation of uric acid in 5-hydroxyisourate, an unstable compound which is latter decomposed into allantoïn. Crystallographic data have shown that urate oxidase binds a dianion urate species deprotonated in N3 and N7, while kinetics experiments have highlighted the existence of several intermediates during catalysis. We have employed a quantum mechanical approach to analyze why urate oxidase is selective for one particular dianion and to explore all possible reaction pathways for the oxidation of one uric acid species by molecular dioxygen in presence of water. Our results indicate the urate dianion deprotonated in N3 and N7 is among all urate species that can coexist in solution it is the compound which will lose the most easiestly one electron in presence of molecular dioxygen. In addition, the transformation of this dianion in 5-hydroxyisourate is thermodynamically the most favorable reaction. Finally, several reaction pathways can be drawn, each starting with the spontaneous transfer of one electron from the urate dianion to molecular dioxygen. During that period, the existence of a 5-hydroperoxyisourate intermediate, which has been proposed elsewhere, does not seem mandatory.


Assuntos
Oxigênio/química , Urato Oxidase/metabolismo , Ácido Úrico/análogos & derivados , Alantoína/biossíntese , Biocatálise , Cristalografia por Raios X , Oxirredução , Teoria Quântica , Termodinâmica , Urato Oxidase/química , Ácido Úrico/química
6.
BMC Struct Biol ; 8: 32, 2008 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-18638417

RESUMO

BACKGROUND: Urate oxidase (EC 1.7.3.3 or UOX) catalyzes the conversion of uric acid and gaseous molecular oxygen to 5-hydroxyisourate and hydrogen peroxide, in the absence of cofactor or particular metal cation. The functional enzyme is a homo-tetramer with four active sites located at dimeric interfaces. RESULTS: The catalytic mechanism was investigated through a ternary complex formed between the enzyme, uric acid, and cyanide that stabilizes an intermediate state of the reaction. When uric acid is replaced by a competitive inhibitor, no complex with cyanide is formed. CONCLUSION: The X-ray structure of this compulsory ternary complex led to a number of mechanistic evidences that support a sequential mechanism in which the two reagents, dioxygen and a water molecule, process through a common site located 3.3 A above the mean plane of the ligand. This site is built by the side chains of Asn 254, and Thr 57, two conserved residues belonging to two different subunits of the homo-tetramer. The absence of a ternary complex between the enzyme, a competitive inhibitor, and cyanide suggests that cyanide inhibits the hydroxylation step of the reaction, after the initial formation of a hydroperoxyde type intermediate.


Assuntos
Aspergillus flavus/enzimologia , Cianetos/farmacologia , Urato Oxidase/antagonistas & inibidores , Urato Oxidase/química , Catálise/efeitos dos fármacos , Cristalografia por Raios X , Cianetos/química , Eletricidade Estática , Especificidade por Substrato/efeitos dos fármacos , Urato Oxidase/metabolismo , Ácido Úrico/química
7.
Biophys J ; 95(5): 2415-22, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18375516

RESUMO

The localization of dioxygen sites in oxygen-binding proteins is a nontrivial experimental task and is often suggested through indirect methods such as using xenon or halide anions as oxygen probes. In this study, a straightforward method based on x-ray crystallography under high pressure of pure oxygen has been developed. An application is given on urate oxidase (UOX), a cofactorless enzyme that catalyzes the oxidation of uric acid to 5-hydroxyisourate in the presence of dioxygen. UOX crystals in complex with a competitive inhibitor of its natural substrate are submitted to an increasing pressure of 1.0, 2.5, or 4.0 MPa of gaseous oxygen. The results clearly show that dioxygen binds within the active site at a location where a water molecule is usually observed but does not bind in the already characterized specific hydrophobic pocket of xenon. Moreover, crystallizing UOX in the presence of a large excess of chloride (NaCl) shows that one chloride ion goes at the same location as the oxygen. The dioxygen hydrophilic environment (an asparagine, a histidine, and a threonine residues), its absence within the xenon binding site, and its location identical to a water molecule or a chloride ion suggest that the dioxygen site is mainly polar. The implication of the dioxygen location on the mechanism is discussed with respect to the experimentally suggested transient intermediates during the reaction cascade.


Assuntos
Cristalografia por Raios X/métodos , Oxigênio/metabolismo , Urato Oxidase/química , Urato Oxidase/metabolismo , Aspergillus flavus/enzimologia , Sítios de Ligação , Catálise , Cristalização , Modelos Moleculares , Pressão , Cloreto de Sódio/metabolismo , Água/metabolismo , Xenônio/metabolismo
8.
FEBS Lett ; 580(8): 2087-91, 2006 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-16545381

RESUMO

Urate oxidase from Aspergillus flavus catalyzes the degradation of uric acid to [S]-allantoin through 5-hydroxyisourate as a metastable intermediate. The second degradation step is thought either catalyzed by another specific enzyme, or spontaneous. The structure of the enzyme was known at high resolution by X-ray diffraction of I222 crystals complexed with a purine-type inhibitor (8-azaxanthin). Analyzing the X-ray structure of urate oxidase treated with an excess of urate, the natural substrate, shows unexpectedly that the active site recaptures [S]-allantoin from the racemic end product of a second degradation step.


Assuntos
Alantoína/metabolismo , Urato Oxidase/metabolismo , Ácido Úrico/metabolismo , Aspergillus flavus/enzimologia , Sítios de Ligação , Cristalografia por Raios X , Modelos Moleculares , Conformação Molecular , Proteínas Recombinantes/metabolismo , Ácido Úrico/química
9.
Artigo em Inglês | MEDLINE | ID: mdl-16511330

RESUMO

Crystallization and preliminary neutron diffraction measurements of rasburicase, a recombinant urate oxidase enzyme expressed by a genetically modified Saccharomyces cerevisiae strain, complexed with a purine-type inhibitor (8-azaxanthin) are reported. Neutron Laue diffraction data were collected to 2.1 A resolution using the LADI instrument from a crystal (grown in D2O) with volume 1.8 mm3. The aim of this neutron diffraction study is to determine the protonation states of the inhibitor and residues within the active site. This will lead to improved comprehension of the enzymatic mechanism of this important enzyme, which is used as a protein drug to reduce toxic uric acid accumulation during chemotherapy. This paper illustrates the high quality of the neutron diffraction data collected, which are suitable for high-resolution structural analysis. In comparison with other neutron protein crystallography studies to date in which a hydrogenated protein has been used, the volume of the crystal was relatively small and yet the data still extend to high resolution. Furthermore, urate oxidase has one of the largest primitive unit-cell volumes (space group I222, unit-cell parameters a = 80, b = 96, c = 106 A) and molecular weights (135 kDa for the homotetramer) so far successfully studied with neutrons.


Assuntos
Urato Oxidase/química , Xantinas/química , Sítios de Ligação , Cristalização/métodos , Óxido de Deutério , Difração de Nêutrons/métodos , Proteínas Recombinantes/química , Saccharomyces cerevisiae/metabolismo
10.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 3): 218-29, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15735331

RESUMO

Urate oxidase from Aspergillus flavus (uricase or Uox; EC 1.7.3.3) is a 135 kDa homotetramer with a subunit consisting of 301 amino acids. It catalyses the first step of the degradation of uric acid into allantoin. The structure of the extracted enzyme complexed with a purine-type inhibitor (8-azaxanthin) had been solved from high-resolution X-ray diffraction of I222 crystals. Expression of the recombinant enzyme in Saccharomyces cerevisiae followed by a new purification procedure allowed the crystallization of both unliganded and liganded enzymes utilizing the same conditions but in various crystal forms. Here, four different crystal forms of Uox are analyzed. The diversity of the Uox crystal forms appears to depend strongly on the chemicals used as inhibitors. In the presence of uracil and 5,6-diaminouracil crystals usually belong to the trigonal space group P3(1)21, the asymmetric unit (AU) of which contains one tetramer of Uox (four subunits). Chemical oxidation of 5,6-diaminouracil within the protein may occur, leading to the canonical (I222) packing with one subunit per AU. Coexistence of two crystal forms, P2(1) with two tetramers per AU and I222, was found in the same crystallization drop containing another inhibitor, guanine. Finally, a fourth form, P2(1)2(1)2 with one tetramer per AU, resulted fortuitously in the presence of cymelarsan, an additive. Of all the reported forms, the I222 crystal forms present by far the best X-ray diffraction resolution (approximately 1.6 angstroms resolution compared with 2.3-3.2 angstroms for the other forms). The various structures and contacts in all crystalline lattices are compared. The backbones are essentially conserved except for the region near the active site. Its location at the dimer interface is thus likely to be at the origin of the crystal contact changes as a response to the various bound inhibitors.


Assuntos
Aspergillus flavus/enzimologia , Urato Oxidase/química , Cristalografia por Raios X , Inibidores Enzimáticos/metabolismo , Modelos Moleculares , Conformação Proteica , Urato Oxidase/antagonistas & inibidores , Urato Oxidase/metabolismo
11.
Acta Crystallogr D Biol Crystallogr ; 60(Pt 3): 453-62, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14993669

RESUMO

High-resolution X-ray structures of the complexes of Aspergillus flavus urate oxidase (Uox) with three inhibitors, 8-azaxanthin (AZA), 9-methyl uric acid (MUA) and oxonic acid (OXC), were determined in an orthorhombic space group (I222). In addition, the ligand-free enzyme was also crystallized in a monoclinic form (P2(1)) and its structure determined. Higher accuracy in the three new enzyme-inhibitor complex structures (Uox-AZA, Uox-MUA and Uox-OXC) with respect to the previously determined structure of Uox-AZA (PDB code 1uox) leads to a reversed position of the inhibitor in the active site of the enzyme. The corrected anchoring of the substrate (uric acid) allows an improvement in the understanding of the enzymatic mechanism of urate oxidase.


Assuntos
Aspergillus flavus/enzimologia , Domínio Catalítico , Proteínas Fúngicas , Ácido Oxônico , Urato Oxidase , Ácido Úrico , Xantinas , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/química , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Proteínas Fúngicas/antagonistas & inibidores , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Humanos , Camundongos , Conformação Molecular , Dados de Sequência Molecular , Ácido Oxônico/química , Ácido Oxônico/metabolismo , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Alinhamento de Sequência , Urato Oxidase/antagonistas & inibidores , Urato Oxidase/química , Urato Oxidase/metabolismo , Ácido Úrico/análogos & derivados , Ácido Úrico/química , Ácido Úrico/metabolismo , Xantinas/química , Xantinas/metabolismo
12.
J Biol Chem ; 278(38): 36887-96, 2003 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-12847093

RESUMO

The three iodothyronine selenodeiodinases catalyze the initiation and termination of thyroid hormone effects in vertebrates. Structural analyses of these proteins have been hindered by their integral membrane nature and the inefficient eukaryotic-specific pathway for selenoprotein synthesis. Hydrophobic cluster analysis used in combination with Position-specific Iterated BLAST reveals that their extramembrane portion belongs to the thioredoxin-fold superfamily for which experimental structure information exists. Moreover, a large deiodinase region imbedded in the thioredoxin fold shares strong similarities with the active site of iduronidase, a member of the clan GH-A-fold of glycoside hydrolases. This model can explain a number of results from previous mutagenesis analyses and permits new verifiable insights into the structural and functional properties of these enzymes.


Assuntos
Iodeto Peroxidase/química , Iodeto Peroxidase/fisiologia , Sequência de Aminoácidos , Proteínas de Bactérias/química , Sítios de Ligação , Linhagem Celular , Bases de Dados como Assunto , Relação Dose-Resposta a Droga , Glicosídeo Hidrolases/química , Humanos , Cinética , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Proteínas de Plantas/química , Reação em Cadeia da Polimerase , Dobramento de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Tiorredoxinas/química , Hormônios Tireóideos/metabolismo , Iodotironina Desiodinase Tipo II
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA