Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Arch Biochem Biophys ; 540(1-2): 26-32, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24071515

RESUMEN

Several pathways of biotic dechlorination can be found in enzymes, each characterized by different chlorine isotopic fractionation, which can thus serve as a signature of a particular mechanism. Unlike other dehalogenases, DL-2-haloacid dehalogenase, DL-DEX, converts both enantiomers of the substrate. Chlorine isotope effects for this enzyme are larger than in the case of other dehalogenases. Recently, the 3D structure of this enzyme became available and enabled us to model these isotope effects and seek their origin. We show that the elevated values of the chlorine kinetic isotope effects originate in part in the processes of binding and migration within the enzyme active site that precede the dehalogenation step.


Asunto(s)
Hidrolasas/química , Hidrolasas/metabolismo , Simulación del Acoplamiento Molecular , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Hidrolasas/antagonistas & inhibidores , Hidrolasas/genética , Isótopos , Mutagénesis Sitio-Dirigida , Mutación , Propionatos/metabolismo , Propionatos/farmacología , Unión Proteica , Estereoisomerismo
2.
Chemistry ; 18(27): 8392-402, 2012 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-22674735

RESUMEN

The high substrate specificity of fluoroacetate dehalogenase was explored by using crystallographic analysis, fluorescence spectroscopy, and theoretical computations. A crystal structure for the Asp104Ala mutant of the enzyme from Burkholderia sp. FA1 complexed with fluoroacetate was determined at 1.2 Å resolution. The orientation and conformation of bound fluoroacetate is different from those in the crystal structure of the corresponding Asp110Asn mutant of the enzyme from Rhodopseudomonas palustris CGA009 reported recently (J. Am. Chem. Soc. 2011, 133, 7461). The fluorescence of the tryptophan residues of the wild-type and Trp150Phe mutant enzymes from Burkholderia sp. FA1 incubated with fluoroacetate and chloroacetate was measured to gain information on the environment of the tryptophan residues. The environments of the tryptophan residues were found to be different between the fluoroacetate- and chloroacetate-bound enzymes; this would come from different binding modes of these two substrates in the active site. Docking simulations and QM/MM optimizations were performed to predict favorable conformations and orientations of the substrates. The F atom of the substrate is oriented toward Arg108 in the most stable enzyme-fluoroacetate complex. This is a stable but unreactive conformation, in which the small O-C-F angle is not suitable for the S(N)2 displacement of the F(-) ion. The cleavage of the C-F bond is initiated by the conformational change of the substrate to a near attack conformation (NAC) in the active site. The second lowest energy conformation is appropriate for NAC; the C-O distance and the O-C-F angle are reasonable for the S(N) 2 reaction. The activation energy is greatly reduced in this conformation because of three hydrogen bonds between the leaving F atom and surrounding amino acid residues. Chloroacetate cannot reach the reactive conformation, due to the longer C-Cl bond; this results in an increase of the activation energy despite the weaker C-Cl bond.


Asunto(s)
Burkholderia/enzimología , Hidrolasas/metabolismo , Rhodopseudomonas/enzimología , Espectrometría de Fluorescencia/métodos , Sitios de Unión , Catálisis , Simulación por Computador , Fluoroacetatos/química , Fluoroacetatos/metabolismo , Histidina/química , Hidrolasas/química , Modelos Teóricos , Conformación Molecular , Especificidad por Sustrato , Triptófano/química , Tirosina/química
3.
J Biochem ; 150(3): 295-302, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21593057

RESUMEN

The genome sequence of an extremely thermophilic bacterium, Thermus thermophilus HB8, revealed that TTHA0350 is a tandem-type universal stress protein (Usp) consisting of two Usp domains. Usp proteins, which are characterized by a conserved domain consisting of 130-160 amino acids, are inducibly expressed under a large number of stress conditions. The N-terminal domain of TTHA0350 contains a motif similar to the consensus ATP-binding one (G-2 x-G-9x-G-(S/T)), but the C-terminal one seems to lack the consensus motif. In order to determine its structural properties, we determined the crystal structures of TTHA0350 in the unliganded form and TTHA0350•2ATP at 2.50 and 1.70 Šresolution, respectively. This is the first structure determination of a Usp family protein in both unliganded and ATP-liganded forms. TTHA0350 is folded into a fan-shaped structure which is similar to that of tandem-type Usp protein Rv2623 from Mycobacterium tuberculosis. However, the dimer assembly with C2-symmetry in TTHA0350 is quite different from that with D2-symmetry in Rv2623. The X-ray structure showed that not only the N-terminal but also the C-terminal domain binds one ATP, although the ATP-binding motif could not be detected in the C-terminal domain. The loop interacting with ATP in the C-terminal domain is in a conformation quite different from that in the N-terminal domain.


Asunto(s)
Adenosina Trifosfato/química , Proteínas Bacterianas/química , Proteínas Portadoras/química , Proteínas de Choque Térmico/química , Thermus thermophilus/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Sitios de Unión , Cristalografía por Rayos X , Proteínas de Choque Térmico/genética , Datos de Secuencia Molecular , Proteínas de Unión a Fosfato , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína
4.
Artículo en Inglés | MEDLINE | ID: mdl-20693661

RESUMEN

Hypoxanthine-guanine phosphoribosyltransferase (HGPRTase), which is a key enzyme in the purine-salvage pathway, catalyzes the synthesis of IMP or GMP from alpha-D-phosphoribosyl-1-pyrophosphate and hypoxanthine or guanine, respectively. Structures of HGPRTase from Thermus thermophilus HB8 in the unliganded form, in complex with IMP and in complex with GMP have been determined at 2.1, 1.9 and 2.2 A resolution, respectively. The overall fold of the IMP complex was similar to that of the unliganded form, but the main-chain and side-chain atoms of the active site moved to accommodate IMP. The overall folds of the IMP and GMP complexes were almost identical to each other. Structural comparison of the T. thermophilus HB8 enzyme with 6-oxopurine PRTases for which structures have been determined showed that these enzymes can be tentatively divided into groups I and II and that the T. thermophilus HB8 enzyme belongs to group I. The group II enzymes are characterized by an N-terminal extension with additional secondary elements and a long loop connecting the second alpha-helix and beta-strand compared with the group I enzymes.


Asunto(s)
Hipoxantina Fosforribosiltransferasa/química , Thermus thermophilus/enzimología , Cristalografía por Rayos X , Guanosina Monofosfato/química , Guanosina Monofosfato/metabolismo , Hipoxantina Fosforribosiltransferasa/metabolismo , Inosina Monofosfato/química , Inosina Monofosfato/metabolismo , Modelos Moleculares , Pliegue de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Especificidad por Sustrato
5.
J Biol Chem ; 285(16): 12133-9, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20164179

RESUMEN

Selenocysteine lyase (SCL) catalyzes the pyridoxal 5'-phosphate-dependent removal of selenium from l-selenocysteine to yield l-alanine. The enzyme is proposed to function in the recycling of the micronutrient selenium from degraded selenoproteins containing selenocysteine residue as an essential component. The enzyme exhibits strict substrate specificity toward l-selenocysteine and no activity to its cognate l-cysteine. However, it remains unclear how the enzyme distinguishes between selenocysteine and cysteine. Here, we present mechanistic studies of selenocysteine lyase from rat. ESI-MS analysis of wild-type and C375A mutant SCL revealed that the catalytic reaction proceeds via the formation of an enzyme-bound selenopersulfide intermediate on the catalytically essential Cys-375 residue. UV-visible spectrum analysis and the crystal structure of SCL complexed with l-cysteine demonstrated that the enzyme reversibly forms a nonproductive adduct with l-cysteine. Cys-375 on the flexible loop directed l-selenocysteine, but not l-cysteine, to the correct position and orientation in the active site to initiate the catalytic reaction. These findings provide, for the first time, the basis for understanding how trace amounts of a selenium-containing substrate is distinguished from excessive amounts of its cognate sulfur-containing compound in a biological system.


Asunto(s)
Liasas/química , Liasas/metabolismo , Selenio/metabolismo , Azufre/metabolismo , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Dominio Catalítico/genética , Secuencia Conservada , Cristalografía por Rayos X , Cisteína/química , Cartilla de ADN/genética , Técnicas In Vitro , Liasas/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Conformación Proteica , Multimerización de Proteína , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Especificidad por Sustrato
6.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 10): 1098-106, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19770507

RESUMEN

D-Alanine-D-alanine ligase (Ddl) is one of the key enzymes in peptidoglycan biosynthesis and is an important target for drug discovery. The enzyme catalyzes the condensation of two D-Ala molecules using ATP to produce D-Ala-D-Ala, which is the terminal peptide of a peptidoglycan monomer. The structures of five forms of the enzyme from Thermus thermophilus HB8 (TtDdl) were determined: unliganded TtDdl (2.3 A resolution), TtDdl-adenylyl imidodiphosphate (2.6 A), TtDdl-ADP (2.2 A), TtDdl-ADP-D-Ala (1.9 A) and TtDdl-ATP-D-Ala-D-Ala (2.3 A). The central domain rotates as a rigid body towards the active site in a cumulative manner in concert with the local conformational change of three flexible loops depending upon substrate or product binding, resulting in an overall structural change from the open to the closed form through semi-open and semi-closed forms. Reaction-intermediate models were simulated using TtDdl-complex structures and other Ddl structures previously determined by X-ray methods. The catalytic process accompanied by the cumulative conformational change has been elucidated based on the intermediate models in order to provide new insights regarding the details of the catalytic mechanism.


Asunto(s)
Péptido Sintasas/química , Thermus thermophilus/enzimología , Dominio Catalítico , Cristalografía por Rayos X , Ligandos , Modelos Moleculares , Conformación Proteica
7.
J Biol Chem ; 284(38): 25944-52, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19640845

RESUMEN

D-serine is an endogenous coagonist for the N-methyl-D-aspartate receptor and is involved in excitatory neurotransmission in the brain. Mammalian pyridoxal 5'-phosphate-dependent serine racemase, which is localized in the mammalian brain, catalyzes the racemization of L-serine to yield D-serine and vice versa. The enzyme also catalyzes the dehydration of D- and L-serine. Both reactions are enhanced by Mg.ATP in vivo. We have determined the structures of the following three forms of the mammalian enzyme homolog from Schizosaccharomyces pombe: the wild-type enzyme, the wild-type enzyme in the complex with an ATP analog, and the modified enzyme in the complex with serine at 1.7, 1.9, and 2.2 A resolution, respectively. On binding of the substrate, the small domain rotates toward the large domain to close the active site. The ATP binding site was identified at the domain and the subunit interface. Computer graphics models of the wild-type enzyme complexed with L-serine and D-serine provided an insight into the catalytic mechanisms of both reactions. Lys-57 and Ser-82 located on the protein and solvent sides, respectively, with respect to the cofactor plane, are acid-base catalysts that shuttle protons to the substrate. The modified enzyme, which has a unique "lysino-D-alanyl" residue at the active site, also exhibits catalytic activities. The crystal-soaking experiment showed that the substrate serine was actually trapped in the active site of the modified enzyme, suggesting that the lysino-D-alanyl residue acts as a catalytic base in the same manner as inherent Lys-57 of the wild-type enzyme.


Asunto(s)
Adenosina Trifosfato/química , Racemasas y Epimerasas/química , Proteínas de Schizosaccharomyces pombe/química , Schizosaccharomyces/enzimología , Serina/química , Adenosina Trifosfato/metabolismo , Animales , Catálisis , Dominio Catalítico/fisiología , Mamíferos , Estructura Terciaria de Proteína/fisiología , Racemasas y Epimerasas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Serina/metabolismo , Homología Estructural de Proteína
8.
J Bacteriol ; 191(8): 2630-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19218394

RESUMEN

Fluoroacetate dehalogenase catalyzes the hydrolytic defluorination of fluoroacetate to produce glycolate. The enzyme is unique in that it catalyzes the cleavage of a carbon-fluorine bond of an aliphatic compound: the bond energy of the carbon-fluorine bond is among the highest found in natural products. The enzyme also acts on chloroacetate, although much less efficiently. We here determined the X-ray crystal structure of the enzyme from Burkholderia sp. strain FA1 as the first experimentally determined three-dimensional structure of fluoroacetate dehalogenase. The enzyme belongs to the alpha/beta hydrolase superfamily and exists as a homodimer. Each subunit consists of core and cap domains. The catalytic triad, Asp104-His271-Asp128, of which Asp104 serves as the catalytic nucleophile, was found in the core domain at the domain interface. The active site was composed of Phe34, Asp104, Arg105, Arg108, Asp128, His271, and Phe272 of the core domain and Tyr147, His149, Trp150, and Tyr212 of the cap domain. An electron density peak corresponding to a chloride ion was found in the vicinity of the N(epsilon1) atom of Trp150 and the N(epsilon2) atom of His149, suggesting that these are the halide ion acceptors. Site-directed replacement of each of the active-site residues, except for Trp150, by Ala caused the total loss of the activity toward fluoroacetate and chloroacetate, whereas the replacement of Trp150 caused the loss of the activity only toward fluoroacetate. An interaction between Trp150 and the fluorine atom is probably an absolute requirement for the reduction of the activation energy for the cleavage of the carbon-fluorine bond.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Burkholderia/química , Burkholderia/enzimología , Hidrolasas/química , Hidrolasas/metabolismo , Acetatos/metabolismo , Sustitución de Aminoácidos/genética , Proteínas Bacterianas/genética , Dominio Catalítico , Cristalografía por Rayos X , Análisis Mutacional de ADN , Fluoroacetatos/metabolismo , Hidrolasas/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
9.
J Biochem ; 145(4): 421-4, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19155267

RESUMEN

Serine racemase synthesizes d-serine, a physiological agonist of the NMDA receptor in mammalian brains. Schizosaccharomyces pombe produces serine racemase (spSR) that is highly similar to the brain enzyme. Our mass-spectrometric and X-ray studies revealed that spSR is modified with its natural substrate serine. spSR remains partially active even though its essential Lys57 inherently forming a Schiff base with the coenzyme pyridoxal 5'-phosphate is converted to N(6)-(R-2-amino-2-carboxyethyl)-l-lysyl (lysino-d-alanyl) residue. This indicates that the alpha-amino group of the d-alanyl moiety of the lysino-d-alanyl residue serves as a catalytic base in the same manner as the epsilon-amino group of Lys57 of the original spSR.


Asunto(s)
Biocatálisis , Lisinoalanina/metabolismo , Racemasas y Epimerasas/metabolismo , Schizosaccharomyces/enzimología , Alanina Racemasa/metabolismo , Dominio Catalítico , Activación Enzimática , Serina/metabolismo , Electricidad Estática
10.
Artículo en Inglés | MEDLINE | ID: mdl-18540056

RESUMEN

It was essential for the structural genomics of Thermus thermophilus HB8 to efficiently crystallize a number of proteins. To this end, three conventional robots, an HTS-80 (sitting-drop vapour diffusion), a Crystal Finder (hanging-drop vapour diffusion) and a TERA (modified microbatch) robot, were subjected to a crystallization condition screening test involving 18 proteins from T. thermophilus HB8. In addition, a TOPAZ (microfluidic free-interface diffusion) designed specifically for initial screening was also briefly examined. The number of diffraction-quality crystals and the time of appearance of crystals increased in the order HTS-80, Crystal Finder, TERA. With the HTS-80 and Crystal Finder, the time of appearance was short and the rate of salt crystallization was low. With the TERA, the number of diffraction-quality crystals was high, while the time of appearance was long and the rate of salt crystallization was relatively high. For the protein samples exhibiting low crystallization success rates, there were few crystallization conditions that were common to the robots used. In some cases, the success rate depended greatly on the robot used. The TOPAZ showed the shortest time of appearance and the highest success rate, although the crystals obtained were too small for diffraction studies. These results showed that the combined use of different robots significantly increases the chance of obtaining crystals, especially for proteins exhibiting low crystallization success rates. The structures of 360 of 944 purified proteins have been successfully determined through the combined use of an HTS-80 and a TERA.


Asunto(s)
Proteínas Bacterianas/química , Thermus thermophilus/química , Proteínas Bacterianas/aislamiento & purificación , Cristalización , Cristalografía por Rayos X , Escherichia coli/genética , Microfluídica , Conformación Proteica , Factores de Tiempo , Difracción de Rayos X
11.
Biochemistry ; 46(44): 12618-27, 2007 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-17929834

RESUMEN

Monofunctional histidinol phosphate phosphatase from Thermus thermophilus HB8, which catalyzes the dephosphorylation of l-histidinol phosphate, belongs to the PHP family, together with the PHP domain of bacterial DNA polymerase III and family X DNA polymerase. We have determined the structures of the complex with a sulfate ion, the complex with a phosphate ion, and the unliganded form at 1.6, 2.1, and 1.8 A resolution, respectively. The enzyme exists as a tetramer, and the subunit consists of a distorted (betaalpha)7 barrel with one linker and one C-terminal tail. Three metal sites located on the C-terminal side of the barrel are occupied by Fe1, Fe2, and Zn ions, respectively, forming a trinuclear metal center liganded by seven histidines, one aspartate, one glutamate, and one hydroxide with two Fe ions bridged by the hydroxide. In the complexes, the sulfate or phosphate ion is coordinated to three metal ions, resulting in octahedral, trigonal bipyramidal, and tetrahedral geometries around the Fe1, Fe2, and Zn ions, respectively. The ligand residues are derived from the four motifs that characterize the PHP family and from two motifs conserved in histidinol phosphate phosphatases. The (betaalpha)7 barrel and the metal cluster are closely related in nature and architecture to the (betaalpha)8 barrel and the mononuclear or dinuclear metal center in the amidohydrolase superfamily, respectively. The coordination behavior of the phosphate ion toward the metal center supports the mechanism in which the bridging hydroxide makes a direct attack on the substrate phosphate tridentately bound to the two Fe ions and Zn ion to hydrolyze the phosphoester bond.


Asunto(s)
Histidinol-Fosfatasa/química , Thermus thermophilus/enzimología , Amidohidrolasas/química , Sitios de Unión , Cristalografía por Rayos X , Dimerización , Histidinol-Fosfatasa/metabolismo , Metales/metabolismo , Modelos Biológicos , Modelos Moleculares , Unión Proteica
12.
Artículo en Inglés | MEDLINE | ID: mdl-17620717

RESUMEN

DL-2-Haloacid dehalogenase from Methylobacterium sp. CPA1 (DL-DEX Mb) is a unique enzyme that catalyzes the dehalogenation reaction without the formation of an ester intermediate. A recombinant form of DL-DEX Mb has been expressed in Escherichia coli, purified and crystallized using the hanging-drop vapour-diffusion method. The crystal belongs to the hexagonal space group P6(3), with unit-cell parameters a = b = 186.2, c = 114.4 A. The crystals are likely to contain between four and eight monomers in the asymmetric unit, with a V(M) value of 4.20-2.10 A3 Da(-1). A self-rotation function revealed peaks on the chi = 180 degrees section. X-ray data have been collected to 1.75 A resolution.


Asunto(s)
Hidrolasas/química , Hidrolasas/genética , Methylobacterium/enzimología , Cristalografía por Rayos X/métodos , Regulación Enzimológica de la Expresión Génica , Hidrolasas/biosíntesis , Hidrolasas/aislamiento & purificación
13.
J Biol Chem ; 281(45): 34365-73, 2006 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-16963440

RESUMEN

Reversible 2,6-dihydroxybenzoate decarboxylase from Rhizobium sp. strain MTP-10005 belongs to a nonoxidative decarboxylase family. We have determined the structures of the following three forms of the enzyme: the native form, the complex with the true substrate (2,6-dihydroxybenzoate), and the complex with 2,3-dihydroxybenzaldehyde at 1.7-, 1.9-, and 1.7-A resolution, respectively. The enzyme exists as a tetramer, and the subunit consists of one (alphabeta)8 triose-phosphate isomerase-barrel domain with three functional linkers and one C-terminal tail. The native enzyme possesses one Zn2+ ion liganded by Glu8, His10, His164, Asp287, and a water molecule at the active site center, although the enzyme has been reported to require no cofactor for its catalysis. The substrate carboxylate takes the place of the water molecule and is coordinated to the Zn2+ ion. The 2-hydroxy group of the substrate is hydrogen-bonded to Asp287, which forms a triad together with His218 and Glu221 and is assumed to be the catalytic base. On the basis of the geometrical consideration, substrate specificity is uncovered, and the catalytic mechanism is proposed for the novel Zn2+-dependent decarboxylation.


Asunto(s)
Carboxiliasas/química , Rhizobium/enzimología , Zinc/metabolismo , Benzaldehídos/metabolismo , Sitios de Unión , Carboxiliasas/metabolismo , Catecoles/metabolismo , Cristalización , Cristalografía por Rayos X , Hidroxibenzoatos/metabolismo , Modelos Moleculares , Oxidación-Reducción , Conformación Proteica , Especificidad por Sustrato
14.
J Biochem ; 139(5): 873-9, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16751595

RESUMEN

Comparison of the primary structures of pig kidney D-amino acid oxidase (DAO) and human brain D-aspartate oxidase (DDO) revealed a notable difference at I215-N225 of DAO and the corresponding region, R216-G220, of DDO. A DAO mutant, in which I215-N225 is substituted by R216-G220 of DDO, showed D-aspartate-oxidizing activity that wild-type DAO does not exhibit, together with a considerable decrease in activity toward D-alanine. These findings indicate that I215-N225 of DAO contributes profoundly to its substrate specificity. Based on these results and the crystal structure of DAO, we systematically mutated the E220-Y224 region within the short stretch in question and obtained five mutants (220D224G, 221D224G, 222D224G, 223D224G, and 224D), in each of which an aspartate residue is mutated to E220-Y224. All of the mutants exhibited decreased apparent K(m) values toward D-arginine, i.e., to one-seventh to one-half that of wild type DAO. The specificity constant, k(cat app)/K(m app), for D-arginine increased by one order of magnitude for the 221D224G or 222D224G mutant, whereas that for D-alanine or D-serine decreased to marginal or nil.


Asunto(s)
D-Aminoácido Oxidasa/metabolismo , Riñón/metabolismo , Animales , Sitios de Unión/genética , Encéfalo/metabolismo , Clonación Molecular , D-Aminoácido Oxidasa/genética , D-Aminoácido Oxidasa/aislamiento & purificación , D-Aspartato Oxidasa/genética , D-Aspartato Oxidasa/aislamiento & purificación , D-Aspartato Oxidasa/metabolismo , Humanos , Cinética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Ingeniería de Proteínas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Especificidad por Sustrato , Porcinos
15.
J Biochem ; 139(4): 789-95, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16672280

RESUMEN

The three-dimensional structure of rat-liver acyl-CoA oxidase-II (ACO-II) in a complex with a C12-fatty acid was solved by the molecular replacement method based on the uncomplexed ACO-II structure. The crystalline form of the complex was obtained by cocrystallization of ACO-II with dodecanoyl-CoA. The crystalline complex possessed, in the active-site crevice, only the fatty acid moiety that had been formed through hydrolysis of the thioester bond. The overall dimeric structure and the folding pattern of each subunit are essentially superimposable on those of uncomplexed ACO-II. The active site including the flavin ring of FAD, the crevice embracing the fatty acyl moiety, and adjacent amino acid side chains are superimposably conserved with the exception of Glu421, whose carboxylate group is tilted away to accommodate the fatty acid. One of the carboxyl oxygens of the bound fatty acid is hydrogen-bonded to the amide hydrogen of Glu421, the presumed catalytic base, and to the ribityl 2'-hydroxyl group of FAD. This hydrogen-bonding network correlates well with the substrate recognition/activation in acyl-CoA dehydrogenase. The binding mode of C12-fatty acid suggests that the active site does not close upon substrate binding, but remains spacious during the entire catalytic process, the oxygen accessibility in the oxidative half-reaction thereby being maintained.


Asunto(s)
Acil-CoA Deshidrogenasas/química , Acil-CoA Oxidasa/química , Ácidos Grasos/química , Hígado/enzimología , Acil-CoA Deshidrogenasas/metabolismo , Acil-CoA Oxidasa/metabolismo , Animales , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X/métodos , Ácidos Grasos/metabolismo , Enlace de Hidrógeno , Modelos Químicos , Oxidación-Reducción , Oxígeno/química , Oxígeno/metabolismo , Conformación Proteica , Estructura Secundaria de Proteína , Ratas , Especificidad por Sustrato
16.
J Biosci Bioeng ; 99(6): 541-7, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16233829

RESUMEN

A new family of NAD(P)H-dependent oxidoreductases is now recognized as a protein family distinct from conventional Rossmann-fold proteins. Numerous putative proteins belonging to the family have been annotated as malate dehydrogenase (MDH) or lactate dehydrogenase (LDH) according to the previous classification as type-2 malate/L-lactate dehydrogenases. However, recent biochemical and genetic studies have revealed that the protein family consists of a wide variety of enzymes with unique catalytic activities other than MDH or LDH activity. Based on their sequence homologies and plausible functions, the family proteins can be grouped into eight clades. This classification would be useful for reliable functional annotation of the new family of NAD(P)H-dependent oxidoreductases.


Asunto(s)
Archaea/enzimología , Modelos Moleculares , NADH NADPH Oxidorreductasas/química , Secuencia de Aminoácidos , Activación Enzimática , Datos de Secuencia Molecular , NADH NADPH Oxidorreductasas/clasificación , Conformación Proteica , Pliegue de Proteína , Homología de Secuencia de Aminoácido
17.
J Biol Chem ; 280(49): 40875-84, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16192274

RESUMEN

Delta(1)-Piperideine-2-carboxylate/Delta(1)-pyrroline-2-carboxylate reductase from Pseudomonas syringae pv. tomato belongs to a novel sub-class in a large family of NAD(P)H-dependent oxidoreductases distinct from the conventional MDH/LDH superfamily characterized by the Rossmann fold. We have determined the structures of the following three forms of the enzyme: the unliganded form, the complex with NADPH, and the complex with NADPH and pyrrole-2-carboxylate at 1.55-, 1.8-, and 1.7-A resolutions, respectively. The enzyme exists as a dimer, and the subunit consists of three domains; domain I, domain II (NADPH binding domain), and domain III. The core of the NADPH binding domain consists of a seven-stranded predominantly antiparallel beta-sheet fold (which we named SESAS) that is characteristic of the new oxidoreductase family. The enzyme preference for NADPH over NADH is explained by the cofactor binding site architecture. A comparison of the overall structures revealed that the mobile domains I and III change their conformations to produce the catalytic form. This conformational change plays important roles in substrate recognition and the catalytic process. The active site structure of the catalytic form made it possible to identify the catalytic Asp:Ser:His triad and investigate the catalytic mechanism from a stereochemical point of view.


Asunto(s)
Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/química , Conformación Proteica , Pirrolina Carboxilato Reductasas/química , Sitios de Unión , Catálisis , Cristalización , Cristalografía por Rayos X , Dimerización , Escherichia coli/genética , Modelos Moleculares , NAD/metabolismo , NADP/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Prolina/análogos & derivados , Prolina/metabolismo , Estructura Secundaria de Proteína , Pseudomonas syringae/enzimología , Pirrolina Carboxilato Reductasas/genética , Pirrolina Carboxilato Reductasas/metabolismo , Proteínas Recombinantes , Especificidad por Sustrato
18.
J Biol Chem ; 280(44): 37246-56, 2005 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-16141215

RESUMEN

This study presents the first three-dimensional structures of human cytosolic branched-chain aminotransferase (hBCATc) isozyme complexed with the neuroactive drug gabapentin, the hBCATc Michaelis complex with the substrate analog, 4-methylvalerate, and the mitochondrial isozyme (hBCATm) complexed with gabapentin. The branched-chain aminotransferases (BCAT) reversibly catalyze transamination of the essential branched-chain amino acids (leucine, isoleucine, valine) to alpha-ketoglutarate to form the respective branched-chain alpha-keto acids and glutamate. The cytosolic isozyme is the predominant BCAT found in the nervous system, and only hBCATc is inhibited by gabapentin. Pre-steady state kinetics show that 1.3 mm gabapentin can completely inhibit the binding of leucine to reduced hBCATc, whereas 65.4 mm gabapentin is required to inhibit leucine binding to hBCATm. Structural analysis shows that the bulky gabapentin is enclosed in the active-site cavity by the shift of a flexible loop that enlarges the active-site cavity. The specificity of gabapentin for the cytosolic isozyme is ascribed at least in part to the location of the interdomain loop and the relative orientation between the small and large domain which is different from these relationships in the mitochondrial isozyme. Both isozymes contain a CXXC center and form a disulfide bond under oxidizing conditions. The structure of reduced hBCATc was obtained by soaking the oxidized hBCATc crystals with dithiothreitol. The close similarity in active-site structures between cytosolic enzyme complexes in the oxidized and reduced states is consistent with the small effect of oxidation on pre-steady state kinetics of the hBCATc first half-reaction. However, these kinetic data do not explain the inactivation of hBCATm by oxidation of the CXXC center. The structural data suggest that there is a larger effect of oxidation on the interdomain loop and residues surrounding the CXXC center in hBCATm than in hBCATc.


Asunto(s)
Aminas/metabolismo , Anticonvulsivantes/metabolismo , Ácidos Ciclohexanocarboxílicos/metabolismo , Citosol/enzimología , Mitocondrias/enzimología , Transaminasas/química , Transaminasas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Cristalización , Cristalografía por Rayos X , Ditiotreitol/metabolismo , Gabapentina , Isoenzimas/química , Isoleucina/química , Lisina/química , Modelos Moleculares , Datos de Secuencia Molecular , Oxidación-Reducción , Conformación Proteica , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Valina/química
19.
Biochemistry ; 44(23): 8218-29, 2005 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-15938611

RESUMEN

The mechanism for the reaction of aspartate aminotransferase with the C4 substrate, l-aspartate, has been well established. The binding of the C4 substrate induces conformational change in the enzyme from the open to the closed form, and the entire reaction proceeds in the closed form of the enzyme. On the contrary, little is known about the reaction with the C5 substrate, l-glutamate. In this study, we analyzed the pH-dependent binding of 2-methyl-l-glutamate to the enzyme and showed that the interaction between the amino group of 2-methyl-l-glutamate and the pyridoxal 5'-phosphate aldimine is weak compared to that between 2-methyl-l-aspartate and the aldimine. The structures of the Michaelis complexes of the enzyme with l-aspartate and l-glutamate were modeled on the basis of the maleate and glutarate complex structures of the enzyme. The result showed that l-glutamate binds to the open form of the enzyme in an extended conformation, and its alpha-amino group points in the opposite direction of the aldimine, while that of l-aspartate is close to the aldimine. These models explain the observations for 2-methyl-l-glutamate and 2-methyl-l-aspartate. The crystal structures of the complexes of aspartate aminotransferase with phosphopyridoxyl derivatives of l-glutamate, d-glutamate, and 2-methyl-l-glutamate were solved as the models for the external aldimine and ketimine complexes of l-glutamate. All the structures were in the closed form, and the two carboxylate groups and the arginine residues binding them are superimposable on the external aldimine complex with 2-methyl-l-aspartate. Taking these facts altogether, it was strongly suggested that the binding of l-glutamate to aspartate aminotransferase to form the Michaelis complex does not induce a conformational change in the enzyme, and that the conformational change to the closed form occurs during the transaldimination step. The hydrophobic residues of the entrance of the active site, including Tyr70, are considered to be important for promoting the transaldimination process and hence the recognition of the C5 substrate.


Asunto(s)
Aminoácidos Dicarboxílicos/química , Aspartato Aminotransferasas/química , Proteínas de Escherichia coli/química , Iminas/química , Aminas/química , Aminoácidos Dicarboxílicos/metabolismo , Aspartato Aminotransferasas/metabolismo , Ácido Aspártico/química , Sitios de Unión , Cristalografía por Rayos X , Proteínas de Escherichia coli/metabolismo , Glutamatos/química , Ácido Glutámico/química , Concentración de Iones de Hidrógeno , Ligandos , Modelos Químicos , N-Metilaspartato/análogos & derivados , N-Metilaspartato/química , Conformación Proteica , Especificidad por Sustrato
20.
Chem Rec ; 5(3): 160-72, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15889412

RESUMEN

Pyridoxal 5'-phosphate-dependent aminotransferases reversibly catalyzes the transamination reaction in which the alpha-amino group of amino acid 1 is transferred to the 2-oxo acid of amino acid 2 (usually 2-oxoglutarate) to produce the 2-oxo acid of amino acid 1 and amino acid 2 (glutamate). An aminotransferase must thus be able to recognize and bind two kinds of amino acids (amino acids 1 and 2), the side chains of which are different in shape and properties, from among many other small molecules. The dual substrate recognition mechanism has been discovered based on three-dimensional structures of aromatic amino acids, histidinol phosphate, glutamine:phenylpyruvate, acetylornithine, and branched-chain amino acid aminotransferases. There are two representative strategies for dual substrate recognition. An aromatic amino acid aminotransferase prepares charged and neutral pockets for acidic and aromatic side chains, respectively, at the same place by a large-scale rearrangement of the hydrogen-bond network caused by the induced fit. In a branched-chain aminotransferase, the same hydrophobic cavity implanted with hydrophilic sites accommodates both hydrophobic and acidic side chains without side-chain rearrangements of the active-site residues, which is reminiscent of the lock and key mechanism. Dual substrate recognition in other aminotransferases is attained by combining the two representative methods.


Asunto(s)
Aminoácidos/química , Aminoácidos/metabolismo , Transaminasas/metabolismo , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Ácidos Cetoglutáricos/química , Ácidos Cetoglutáricos/metabolismo , Conformación Molecular , Estereoisomerismo , Especificidad por Sustrato , Transaminasas/química , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...