Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Biol Chem ; 300(3): 105728, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38325740

RESUMEN

Serine palmitoyltransferase (SPT) catalyzes the pyridoxal-5'-phosphate (PLP)-dependent decarboxylative condensation of l-serine and palmitoyl-CoA to form 3-ketodihydrosphingosine (KDS). Although SPT was shown to synthesize corresponding products from amino acids other than l-serine, it is still arguable whether SPT catalyzes the reaction with d-serine, which is a question of biological importance. Using high substrate and enzyme concentrations, KDS was detected after the incubation of SPT from Sphingobacterium multivorum with d-serine and palmitoyl-CoA. Furthermore, the KDS comprised equal amounts of 2S and 2R isomers. 1H-NMR study showed a slow hydrogen-deuterium exchange at Cα of serine mediated by SPT. We further confirmed that SPT catalyzed the racemization of serine. The rate of the KDS formation from d-serine was comparable to those for the α-hydrogen exchange and the racemization reaction. The structure of the d-serine-soaked crystal (1.65 Å resolution) showed a distinct electron density of the PLP-l-serine aldimine, interpreted as the racemized product trapped in the active site. The structure of the α-methyl-d-serine-soaked crystal (1.70 Å resolution) showed the PLP-α-methyl-d-serine aldimine, mimicking the d-serine-SPT complex prior to racemization. Based on these enzymological and structural analyses, the synthesis of KDS from d-serine was explained as the result of the slow racemization to l-serine, followed by the reaction with palmitoyl-CoA, and SPT would not catalyze the direct condensation between d-serine and palmitoyl-CoA. It was also shown that the S. multivorum SPT catalyzed the racemization of the product KDS, which would explain the presence of (2R)-KDS in the reaction products.


Asunto(s)
Serina C-Palmitoiltransferasa , Serina , Sphingobacterium , Dominio Catalítico , Cristalización , Medición de Intercambio de Deuterio , Electrones , Hidrógeno/metabolismo , Palmitoil Coenzima A/metabolismo , Serina/análogos & derivados , Serina/metabolismo , Serina C-Palmitoiltransferasa/química , Serina C-Palmitoiltransferasa/metabolismo , Sphingobacterium/enzimología , Sphingobacterium/metabolismo , Esfingosina/análogos & derivados , Esfingosina/biosíntesis , Esfingosina/metabolismo , Estereoisomerismo , Especificidad por Sustrato
2.
J Biol Chem ; 299(5): 104684, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37030501

RESUMEN

Serine palmitoyltransferase (SPT) is a key enzyme of sphingolipid biosynthesis, which catalyzes the pyridoxal-5'-phosphate-dependent decarboxylative condensation reaction of l-serine (l-Ser) and palmitoyl-CoA (PalCoA) to form 3-ketodihydrosphingosine called long chain base (LCB). SPT is also able to metabolize l-alanine (l-Ala) and glycine (Gly), albeit with much lower efficiency. Human SPT is a membrane-bound large protein complex containing SPTLC1/SPTLC2 heterodimer as the core subunits, and it is known that mutations of the SPTLC1/SPTLC2 genes increase the formation of deoxy-type of LCBs derived from l-Ala and Gly to cause some neurodegenerative diseases. In order to study the substrate recognition of SPT, we examined the reactivity of Sphingobacterium multivorum SPT on various amino acids in the presence of PalCoA. The S. multivorum SPT could convert not only l-Ala and Gly but also l-homoserine, in addition to l-Ser, into the corresponding LCBs. Furthermore, we obtained high-quality crystals of the ligand-free form and the binary complexes with a series of amino acids, including a nonproductive amino acid, l-threonine, and determined the structures at 1.40 to 1.55 Å resolutions. The S. multivorum SPT accommodated various amino acid substrates through subtle rearrangements of the active-site amino acid residues and water molecules. It was also suggested that non-active-site residues mutated in the human SPT genes might indirectly influence the substrate specificity by affecting the hydrogen-bonding networks involving the bound substrate, water molecules, and amino acid residues in the active site of this enzyme. Collectively, our results highlight SPT structural features affecting substrate specificity for this stage of sphingolipid biosynthesis.


Asunto(s)
Serina C-Palmitoiltransferasa , Sphingobacterium , Humanos , Palmitoil Coenzima A/química , Palmitoil Coenzima A/metabolismo , Serina/química , Serina C-Palmitoiltransferasa/genética , Serina C-Palmitoiltransferasa/metabolismo , Sphingobacterium/enzimología , Esfingolípidos/metabolismo , Especificidad por Sustrato
3.
Biochemistry ; 55(12): 1801-12, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26979298

RESUMEN

Adenosine diphosphate ribose pyrophosphatase (ADPRase), a member of the Nudix family proteins, catalyzes the metal-induced and concerted general acid-base hydrolysis of ADP ribose (ADPR) into AMP and ribose-5'-phosphate (R5P). The ADPR-hydrolysis reaction of ADPRase from Thermus thermophilus HB8 (TtADPRase) requires divalent metal cations such as Mn(2+), Zn(2+), or Mg(2+) as cofactors. Here, we report the reaction pathway observed in the catalytic center of TtADPRase, based on cryo-trapping X-ray crystallography at atomic resolutions around 1.0 Å using Mn(2+) as the reaction trigger, which was soaked into TtADPRase-ADPR binary complex crystals. Integrating 11 structures along the reaction timeline, five reaction states of TtADPRase were assigned, which were ADPRase alone (E), the ADPRase-ADPR binary complex (ES), two ADPRase-ADPR-Mn(2+) reaction intermediates (ESM, ESMM), and the postreaction state (E'). Two Mn(2+) ions were inserted consecutively into the catalytic center of the ES-state and ligated by Glu86 and Glu82, which are highly conserved among the Nudix family, in the ESM- and ESMM-states. The ADPR-hydrolysis reaction was characterized by electrostatic, proximity, and orientation effects, and by preferential binding for the transition state. A new reaction mechanism is proposed, which differs from previous ones suggested from structure analyses with nonhydrolyzable substrate analogues or point-mutated ADPRases.


Asunto(s)
Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/metabolismo , Coenzimas/química , Coenzimas/metabolismo , Manganeso/química , Manganeso/metabolismo , Sitios de Unión/fisiología , Cristalografía por Rayos X , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
4.
J Biochem ; 171(1): 27-29, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34750609

RESUMEN

Since the discovery of D-amino acid oxidase (DAO) in 1935, many studies have been conducted without clarifying its 3D structure for a long time. In 1996, the crystal structure of DAO was determined, and it was shown that the catalytic bases required for the two catalytic mechanisms were not present in the active site. The crystal structure of DAO in complex with o-aminobenzoate was solved and is used for modeling Michaelis complex. The Michaelis complex model provided structural information leading to a new mechanism for reductive half-reaction of DAO. Currently, DAO is being researched for medical and applied purposes.


Asunto(s)
D-Aminoácido Oxidasa , ortoaminobenzoatos , Aminoácidos , Catálisis , Dominio Catalítico , D-Aminoácido Oxidasa/metabolismo , Modelos Moleculares
5.
Acta Crystallogr F Struct Biol Commun ; 78(Pt 12): 408-415, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36458620

RESUMEN

Serine palmitoyltransferase (SPT) catalyses the first reaction in sphingolipid biosynthesis: the decarboxylative condensation of L-serine (L-Ser) and palmitoyl-CoA to form 3-ketodihydrosphingosine. SPT from Sphingobacterium multivorum has been isolated and its crystal structure in complex with L-Ser has been determined at 2.3 Šresolution (PDB entry 3a2b). However, the quality of the crystal was not good enough to judge the conformation of the cofactor molecule and the orientations of the side chains of the amino-acid residues in the enzyme active site. The crystal quality was improved by revision of the purification procedure and by optimization of both the crystallization procedure and the post-crystallization treatment conditions. Here, the crystal structure of SPT complexed with tris(hydroxymethyl)aminomethane (Tris), a buffer component, was determined at 1.65 Šresolution. The protein crystallized at 20°C and diffraction data were collected from the crystals to a resolution of 1.65 Å. The crystal belonged to the tetragonal space group P41212, with unit-cell parameters a = b = 61.32, c = 208.57 Å. Analysis of the crystal structure revealed C4-C5-C5A-O4P (77°) and C5-C5A-O4P-P (-143°) torsion angles in the phosphate-group moiety of the cofactor pyridoxal 5'-phosphate (PLP) that are more reasonable than those observed in the previously reported crystal structure (14° and 151°, respectively). Furthermore, the clear electron density showing a Schiff-base linkage between PLP and the bulky artificial ligand Tris indicated exceptional flexibility of the active-site cavity of this enzyme. These findings open up the possibility for further study of the detailed mechanisms of substrate recognition and catalysis by this enzyme.


Asunto(s)
Serina C-Palmitoiltransferasa , Trometamina , Cristalografía por Rayos X , Fosfato de Piridoxal , Serina
6.
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
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 ; 165(2): 185-195, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30423116

RESUMEN

Homoserine dehydrogenase from Thermus thermophilus (TtHSD) is a key enzyme in the aspartate pathway that catalyses the reversible conversion of l-aspartate-ß-semialdehyde to l-homoserine (l-Hse) with NAD(P)H. We determined the crystal structures of unliganded TtHSD, TtHSD complexed with l-Hse and NADPH, and Lys99Ala and Lys195Ala mutant TtHSDs, which have no enzymatic activity, complexed with l-Hse and NADP+ at 1.83, 2.00, 1.87 and 1.93 Å resolutions, respectively. Binding of l-Hse and NADPH induced the conformational changes of TtHSD from an open to a closed form: the mobile loop containing Glu180 approached to fix l-Hse and NADPH, and both Lys99 and Lys195 could make hydrogen bonds with the hydroxy group of l-Hse. The ternary complex of TtHSDs in the closed form mimicked a Michaelis complex better than the previously reported open form structures from other species. In the crystal structure of Lys99Ala TtHSD, the productive geometry of the ternary complex was almost preserved with one new water molecule taking over the hydrogen bonds associated with Lys99, while the positions of Lys195 and l-Hse were significantly retained with those of the wild-type enzyme. These results propose new possibilities that Lys99 is the acid-base catalytic residue of HSDs.


Asunto(s)
Homoserina Deshidrogenasa/química , Homoserina/química , NADP/química , Cristalografía por Rayos X , Homoserina/metabolismo , Homoserina Deshidrogenasa/metabolismo , Modelos Moleculares , NADP/metabolismo , Conformación Proteica , Thermus thermophilus/enzimología
10.
J Am Chem Soc ; 130(3): 792-3, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18161973

RESUMEN

Dynamic and efficient inversion of peptide helices by an achiral NO3- anion was programmed in terms of time scale from milliseconds to hours.


Asunto(s)
Nitratos/química , Compuestos Organometálicos/química , Péptidos/química , Acetonitrilos/química , Aniones/química , Cristalografía por Rayos X , Níquel/química , Estructura Secundaria de Proteína , Factores de Tiempo , Zinc/química
11.
Sci Rep ; 8(1): 14228, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30242198

RESUMEN

The biosynthesis of heme is strictly regulated, probably because of the toxic effects of excess heme and its biosynthetic precursors. In many organisms, heme biosynthesis starts with the production of 5-aminolevulinic acid (ALA) from glycine and succinyl-coenzyme A, a process catalyzed by a homodimeric enzyme, pyridoxal 5'-phosphate (PLP)-dependent 5-aminolevulinate synthase (ALAS). ALAS activity is negatively regulated by heme in various ways, such as the repression of ALAS gene expression, degradation of ALAS mRNA, and inhibition of mitochondrial translocation of the mammalian precursor protein. There has been no clear evidence, however, that heme directly binds to ALAS to negatively regulate its activity. We found that recombinant ALAS from Caulobacter crescentus was inactivated via a heme-mediated feedback manner, in which the essential coenzyme PLP was rel eased to form the inactive heme-bound enzyme. The spectroscopic properties of the heme-bound ALAS showed that a histidine-thiolate hexa-coordinated ferric heme bound to each subunit with a one-to-one stoichiometry. His340 and Cys398 were identified as the axial ligands of heme, and mutant ALASs lacking either of these ligands became resistant to heme-mediated inhibition. ALAS expressed in C. crescentus was also found to bind heme, suggesting that heme-mediated feedback inhibition of ALAS is physiologically relevant in C. crescentus.


Asunto(s)
5-Aminolevulinato Sintetasa/metabolismo , Caulobacter crescentus/metabolismo , Hemo/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Coenzimas/metabolismo , Histidina/metabolismo , Humanos , Ligandos , Fosfato de Piridoxal/metabolismo , ARN Mensajero/metabolismo
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 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
14.
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
15.
Structure ; 12(8): 1413-23, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15296735

RESUMEN

CTP synthetase (CTPs) catalyzes the last step in CTP biosynthesis, in which ammonia generated at the glutaminase domain reacts with the ATP-phosphorylated UTP at the synthetase domain to give CTP. Glutamine hydrolysis is active in the presence of ATP and UTP and is stimulated by the addition of GTP. We report the crystal structures of Thermus thermophilus HB8 CTPs alone, CTPs with 3SO4(2-), and CTPs with glutamine. The enzyme is folded into a homotetramer with a cross-shaped structure. Based on the binding mode of sulfate anions to the synthetase site, ATP and UTP are computer modeled into CTPs with a geometry favorable for the reaction. Glutamine bound to the glutaminase domain is situated next to the triad of Glu-His-Cys as a catalyst and a water molecule. Structural information provides an insight into the conformational changes associated with the binding of ATP and UTP and the formation of the GTP binding site.


Asunto(s)
Ligasas de Carbono-Nitrógeno/química , Citidina Trifosfato/química , Guanosina Trifosfato/química , Modelos Moleculares , Uridina Trifosfato/química , Sitios de Unión , Cristalografía por Rayos X , Ácido Glutámico/química , Glutaminasa/química , Thermus thermophilus/enzimología
16.
Biochim Biophys Acta ; 1647(1-2): 321-4, 2003 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-12686152

RESUMEN

Histidinol phosphate aminotransferase (HPAT) is a pyridoxal 5'-phosphate (PLP)-dependent aminotransferase classified into Subgroup I aminotransferase, in which aspartate aminotransferase (AspAT) is the prototype. In order to expand our knowledge on the reaction mechanism of Subgroup I aminotransferases, HPAT is an enzyme suitable for detailed mechanistic studies because of having low sequence identity with AspAT and a unique substrate recognition mode. Here we investigated the spectroscopic properties of HPAT and the effect of the C4-C4' strain of the PLP-Lys(214) Schiff base on regulating the Schiff base pK(a) in HPAT. Similar to AspAT, the PLP-form HPAT showed pH-dependent absorption spectral change with maxima at 340 nm at high pH and 420 nm at low pH, having a low pK(a) of 6.6. The pK(a) value of the methylamine-reconstituted K214A mutant enzyme was increased from 6.6 to 10.6. Mutation of Asn(157) to Ala increased the pK(a) to 9.2. Replacement of Arg(335) by Leu increased the pK(a) to 8.6. On the other hand, the pK(a) value of the N157A/R335L double mutant enzyme was 10.6. These data indicate that the strain of the Schiff base is the principal factor to decrease the pK(a) in HPAT and is crucial for the subsequent increase in the Schiff base pK(a) during catalysis, although the electrostatic effect of the arginine residue that binds the negatively charged group of the substrate is larger in HPAT than that in AspAT. Our findings also support the idea that the strain mechanism is common to Subgroup I aminotransferases.


Asunto(s)
Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Transaminasas/química , Cinética , Bases de Schiff , Electricidad Estática , Transaminasas/metabolismo
17.
Biochim Biophys Acta ; 1647(1-2): 103-9, 2003 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-12686117

RESUMEN

The notion of "ground-state destabilization" has been well documented in enzymology. It is the unfavourable interaction (strain) in the enzyme-substrate complex, and increases the k(cat) value without changing the k(cat)/K(m) value. During the course of the investigation on the reaction mechanism of aspartate aminotransferase (AAT), we found another type of strain that is crucial for catalysis: the strain of the distorted internal aldimine in the unliganded enzyme. This strain raises the energy level of the starting state (E+S), thereby reducing the energy gap between E+S and ES(++) and increasing the k(cat)/K(m) value. Further analysis on the reaction intermediates showed that the Michaelis complex of AAT with aspartate contains strain energy due to an unfavourable interaction between the main chain carbonyl oxygen and the Tyr225-aldimine hydrogen-bonding network. This belongs to the classical type of strain. In each case, the strain is reflected in the pK(a) value of the internal aldimine. In the historical explanation of the reaction mechanism of AAT, the shifts in the aldimine pK(a) have been considered to be the driving forces for the proton transfer during catalysis. However, the above findings indicate that the true driving forces are the strain energy inherent to the respective intermediates. We describe here how these strain energies are generated and are used for catalysis, and show that variations in the aldimine pK(a) during catalysis are no more than phenomenological results of adjusting the energy levels of the reaction intermediates for efficient catalysis.


Asunto(s)
Aspartato Aminotransferasas/química , Aspartato Aminotransferasas/metabolismo , Catálisis , Enlace de Hidrógeno , Termodinámica
18.
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
19.
FEBS J ; 282(21): 4201-17, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26279274

RESUMEN

The Brevibacillus brevis BBR47_28440 gene (referred to as ddlR) encodes an MocR/GabR family transcriptional regulator consisting of an N-terminal helix-turn-helix DNA binding domain and a C-terminal aminotransferase-like domain. The ddlR gene is located just upstream of the d-alanyl-d-alanine ligase gene (ddl) in the B. brevis genome, and these two genes form an operon. Gel-shift assays indicated that purified DdlR binds specifically to the DNA region that includes putative -35 and -10 regions of the ddlR promoter. A 6-bp inverted repeat that overlaps the -10 region of the ddlR promoter was found to be important for the binding. In vivo reporter assays confirmed that DdlR is an activator of the ddlR-ddl operon. Spectroscopic analyses indicated that purified DdlR is a pyridoxal 5'-phosphate binding transcriptional regulator that has dipeptide binding ability for d-alanyl-d-alanine, the enzymatic product of Ddl, and glycylglycine. DdlR is capable of forming a dipeptide-pyridoxal 5'-phosphate external aldimine, but it lacks aminotransferase activity. Bioinformatic analyses suggest that DdlR-mediated transcriptional regulation of ddlR and ddl may occur in multiple bacterial systems such as Actinobacteria and Bacillus species.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Brevibacillus/genética , Brevibacillus/metabolismo , Péptido Sintasas/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Secuencia de Bases , Sitios de Unión/genética , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Dipéptidos/metabolismo , Genes Bacterianos , Secuencias Invertidas Repetidas , Datos de Secuencia Molecular , Operón , Filogenia , Regiones Promotoras Genéticas , Unión Proteica , Homología de Secuencia de Aminoácido , Transactivadores/química , Activación Transcripcional
20.
J Biochem ; 132(5): 759-65, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12417026

RESUMEN

Imidazole glycerol phosphate synthase (IGPs) catalyzes the fifth step in the histidine biosynthetic pathway located at the branch point to de novo purine biosynthesis. IGPs is a multienzyme comprising glutaminase and synthase subunits. The glutaminase activity, which hydrolyzes glutamine to give ammonia, is coupled with substrate binding to the synthase subunit. The three-dimensional structure of the IGPs from Thermus thermophilus HB8 has been determined at 2.3 A resolution, and compared with the previously determined structures for the yeast and Thermotoga maritima enzymes. The structure of each subunit is similar to that of the corresponding domain in the yeast enzyme or subunit in the T. maritima enzyme. However, the overall structure is significantly different from the yeast and T. maritima enzymes, indicating that IGPs may change the relative orientation between the two subunits and close the glutaminase site upon glutamine binding. The putative ammonia tunnel, which carries nascent ammonia from glutaminase to the synthase site, has a closed gate comprising a cyclic salt bridge formed by four charged residues of the synthase subunit. The side chain of Lys100 in the cyclic salt bridge might change its side chain direction to form new interactions with the main chain carbonyl group of glutamine from the synthase subunit and the hydoxyl group of tyrosine from the glutaminase subunit, resulting in the opening of the gate for ammonia transfer.


Asunto(s)
Aminohidrolasas/química , Amoníaco/metabolismo , Thermus thermophilus/enzimología , Aminohidrolasas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Glutaminasa/metabolismo , Glutamina/metabolismo , Conformación Proteica , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA