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 Bacteriol ; 183(7): 2234-40, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11244062

RESUMO

In Salmonella enterica serovar Typhimurium, purine nucleotides and thiamine are synthesized by a branched pathway. The last known common intermediate, aminoimidazole ribonucleotide (AIR), is formed from formylglycinamidine ribonucleotide (FGAM) and ATP by AIR synthetase, encoded by the purI gene in S. enterica. Reduced flux through the first five steps of de novo purine synthesis results in a requirement for purines but not necessarily thiamine. To examine the relationship between the purine and thiamine biosynthetic pathways, purI mutants were made (J. L. Zilles and D. M. Downs, Genetics 143:37-44, 1996). Unexpectedly, some mutant purI alleles (R35C/E57G and K31N/A50G/L218R) allowed growth on minimal medium but resulted in thiamine auxotrophy when exogenous purines were supplied. To explain the biochemical basis for this phenotype, the R35C/E57G mutant PurI protein was purified and characterized kinetically. The K(m) of the mutant enzyme for FGAM was unchanged relative to the wild-type enzyme, but the V(max) was decreased 2.5-fold. The K(m) for ATP of the mutant enzyme was 13-fold increased. Genetic analysis determined that reduced flux through the purine pathway prevented PurI activity in the mutant strain, and purR null mutations suppressed this defect. The data are consistent with the hypothesis that an increased FGAM concentration has the ability to compensate for the lower affinity of the mutant PurI protein for ATP.


Assuntos
Carbono-Nitrogênio Ligases/genética , Mutação , Salmonella typhimurium/enzimologia , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/fisiologia , Carbono-Nitrogênio Ligases/metabolismo , Cinética , Proteínas Repressoras/fisiologia
2.
Curr Opin Chem Biol ; 4(5): 567-72, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11006546

RESUMO

Structural studies, sequence alignments, and biochemistry have provided new insights into the evolution of the purine biosynthetic pathway. The importance of chemistry, the binding of ribose 5-phosphate (common to all purine biosynthetic intermediates), and transient protein-protein interactions in channeling of chemically unstable intermediates have all been examined in the past few years.


Assuntos
Evolução Molecular , Purinas/biossíntese , Modelos Moleculares , Proteínas/química , Purinas/química
3.
Biochemistry ; 39(14): 3927-36, 2000 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-10747780

RESUMO

Polyhydroxybutyrate (PHB) synthases catalyze the conversion of beta-hydroxybutyryl coenzyme A (HBCoA) to PHB. These enzymes require an active site cysteine nucleophile for covalent catalysis. A protein BLASTp search using the Class III Chromatium vinosum synthase sequence reveals high homology to prokaryotic lipases whose crystal structures are known. The homology is very convincing in the alpha-beta-elbow (with the active site nucleophile)-alpha-beta structure, residues 131-175 of the synthase. A conserved histidine of the Class III PHB synthases aligns with the active site histidine of the lipases using the ClustalW algorithm. This is intriguing as this histidine is approximately 200 amino acids removed in sequence space from the catalytic nucleophile. Different threading algorithms suggest that the Class III synthases belong to the alpha/beta hydrolase superfamily which includes prokaryotic lipases. Mutagenesis studies were carried out on C. vinosum synthase C149, H331, H303, D302, and C130 residues. These studies reveal that H331 is the general base catalyst that activates the nucleophile, C149, for covalent catalysis. The model indicates that C130 is not involved in catalysis as previously proposed [Müh, U., Sinskey, A. J., Kirby, D. P., Lane, W. S., and Stubbe, J. (1999) Biochemistry 38, 826-837]. Studies with D302 mutants suggest D302 functions as a general base catalyst in activation of the 3-hydroxyl of HBCoA (or a hydroxybutyrate acyl enzyme) for nucleophilic attack on the covalently linked thiol ester intermediate. The relationship of the lipase model to previous models based on fatty acid synthases is discussed.


Assuntos
Aciltransferases/química , Lipase/química , Modelos Moleculares , Aciltransferases/genética , Sequência de Aminoácidos , Chromatium , Lipase/genética , Mecânica , Dados de Sequência Molecular , Conformação Proteica , Alinhamento de Sequência
4.
Structure ; 7(11): 1395-406, 1999 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-10574791

RESUMO

BACKGROUND: Conversion of 5-aminoimidazole ribonucleotide (AIR) to 4-carboxyaminoimidazole ribonucleotide (CAIR) in Escherichia coli requires two proteins - PurK and PurE. PurE has recently been shown to be a mutase that catalyzes the unusual rearrangement of N(5)-carboxyaminoimidazole ribonucleotide (N(5)-CAIR), the PurK reaction product, to CAIR. PurEs from higher eukaryotes are homologous to E. coli PurE, but use AIR and CO(2) as substrates to produce CAIR directly. RESULTS: The 1.50 A crystal structure of PurE reveals an octameric structure with 422 symmetry. A central three-layer (alphabetaalpha) sandwich domain and a kinked C-terminal helix form the folded structure of the monomeric unit. The structure reveals a cleft at the interface of two subunits and near the C-terminal helix of a third subunit. Co-crystallization experiments with CAIR confirm this to be the mononucleotide-binding site. The nucleotide is bound predominantly to one subunit, with conserved residues from a second subunit making up one wall of the cleft. CONCLUSIONS: The crystal structure of PurE reveals a unique quaternary structure that confirms the octameric nature of the enzyme. An analysis of the native crystal structure, in conjunction with sequence alignments and studies of co-crystals of PurE with CAIR, reveals the location of the active site. The environment of the active site and the analysis of conserved residues between the two classes of PurEs suggests a model for the differences in their substrate specificities and the relationship between their mechanisms.


Assuntos
Carboxiliases/química , Escherichia coli/química , Purinas/biossíntese , Sequência de Aminoácidos , Sítios de Ligação , Carboxiliases/metabolismo , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Homologia de Sequência de Aminoácidos
5.
Biochemistry ; 38(47): 15480-92, 1999 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-10569930

RESUMO

Escherichia coli PurK, a dimeric N5-carboxyaminoimidazole ribonucleotide (N5-CAIR) synthetase, catalyzes the conversion of 5-aminoimidazole ribonucleotide (AIR), ATP, and bicarbonate to N5-CAIR, ADP, and Pi. Crystallization of both a sulfate-liganded and the MgADP-liganded E. coli PurK has resulted in structures at 2.1 and 2.5 A resolution, respectively. PurK belongs to the ATP grasp superfamily of C-N ligase enzymes. Each subunit of PurK is composed of three domains (A, B, and C). The B domain contains a flexible, glycine-rich loop (B loop, T123-G130) that is disordered in the sulfate-PurK structure and becomes ordered in the MgADP-PurK structure. MgADP is wedged between the B and C domains, as with all members of the ATP grasp superfamily. Other enzymes in this superfamily contain a conserved Omega loop proposed to interact with the B loop, define the specificity of their nonnucleotide substrate, and protect the acyl phosphate intermediate formed from this substrate. PurK contains a minimal Omega loop without conserved residues. In the reaction catalyzed by PurK, carboxyphosphate is the putative acyl phosphate intermediate. The sulfate of the sulfate ion-liganded PurK interacts electrostatically with Arg 242 and the backbone amide group of Asn 245, components of the J loop of the C domain. This sulfate may reveal the location of the carboxyphosphate binding site. Conserved residues within the C-terminus of the C domain define a pocket that is proposed to bind AIR in collaboration with an N-terminal strand loop helix motif in the A domain (P loop, G8-L1). The P loop is proposed to bind the phosphate of AIR on the basis of similar binding sites observed in PurN and PurE and proposed in PurD and PurT, four other enzymes in the purine pathway.


Assuntos
Adenosina Trifosfatases/química , Proteínas de Bactérias/química , Carboxiliases , Proteínas de Escherichia coli , Complexo de Golgi/química , Proteínas de Membrana/química , Fosfatos , Ribonucleotídeos/química , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Simulação por Computador , Cristalografia por Raios X , Dimerização , Escherichia coli/enzimologia , Complexo de Golgi/metabolismo , Ligantes , Proteínas de Membrana/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Compostos Organofosforados/química , Compostos Organofosforados/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Ribonucleotídeos/metabolismo , Homologia de Sequência de Aminoácidos
6.
Structure ; 7(9): 1155-66, 1999 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10508786

RESUMO

BACKGROUND: The purine biosynthetic pathway in procaryotes enlists eleven enzymes, six of which use ATP. Enzymes 5 and 6 of this pathway, formylglycinamide ribonucleotide (FGAR) amidotransferase (PurL) and aminoimidazole ribonucleotide (AIR) synthetase (PurM) utilize ATP to activate the oxygen of an amide within their substrate toward nucleophilic attack by a nitrogen. AIR synthetase uses the product of PurL, formylglycinamidine ribonucleotide (FGAM) and ATP to make AIR, ADP and P(i). RESULTS: The structure of a hexahistidine-tagged PurM has been solved by multiwavelength anomalous diffraction phasing techniques using protein containing 28 selenomethionines per asymmetric unit. The final model of PurM consists of two crystallographically independent dimers and four sulfates. The overall R factor at 2.5 A resolution is 19.2%, with an R(free) of 26.4%. The active site, identified in part by conserved residues, is proposed to be a long groove generated by the interaction of two monomers. A search of the sequence databases suggests that the ATP-binding sites between PurM and PurL may be structurally conserved. CONCLUSIONS: The first structure of a new class of ATP-binding enzyme, PurM, has been solved and a model for the active site has been proposed. The structure is unprecedented, with an extensive and unusual sheet-mediated intersubunit interaction defining the active-site grooves. Sequence searches suggest that two successive enzymes in the purine biosynthetic pathway, proposed to use similar chemistries, will have similar ATP-binding domains.


Assuntos
Carbono-Nitrogênio Ligases com Glutamina como Doadora de N-Amida , Carbono-Nitrogênio Ligases/química , Carbono-Nitrogênio Ligases/metabolismo , Proteínas de Escherichia coli , Escherichia coli/enzimologia , Ligases , Purinas/biossíntese , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Carbono-Nitrogênio Ligases/isolamento & purificação , Cristalização , Cristalografia por Raios X , Dimerização , Escherichia coli/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Dobramento de Proteína , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Sulfatos/metabolismo
7.
Biochemistry ; 38(31): 9831-9, 1999 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-10433689

RESUMO

Aminoimidazole ribonucleotide (AIR) synthetase (PurM) catalyzes the conversion of formylglycinamide ribonucleotide (FGAM) and ATP to AIR, ADP, and P(i), the fifth step in de novo purine biosynthesis. The ATP binding domain of the E. coli enzyme has been investigated using the affinity label [(14)C]-p-fluorosulfonylbenzoyl adenosine (FSBA). This compound results in time-dependent inactivation of the enzyme which is accelerated by the presence of FGAM, and gives a K(i) = 25 microM and a k(inact) = 5.6 x 10(-)(2) min(-)(1). The inactivation is inhibited by ADP and is stoichiometric with respect to AIR synthetase. After trypsin digestion of the labeled enzyme, a single labeled peptide has been isolated, I-X-G-V-V-K, where X is Lys27 modified by FSBA. Site-directed mutants of AIR synthetase were prepared in which this Lys27 was replaced with a Gln, a Leu, and an Arg and the kinetic parameters of the mutant proteins were measured. All three mutants gave k(cat)s similar to the wild-type enzyme and K(m)s for ATP less than that determined for the wild-type enzyme. Efforts to inactivate the chicken liver trifunctional AIR synthetase with FSBA were unsuccessful, despite the presence of a Lys27 equivalent. The role of Lys27 in ATP binding appears to be associated with the methylene linker rather than its epsilon-amino group. The specific labeling of the active site by FSBA has helped to define the active site in the recently determined structure of AIR synthetase [Li, C., Kappock, T. J., Stubbe, J., Weaver, T. M., and Ealick, S. E. (1999) Structure (in press)], and suggests additional flexibility in the ATP binding region.


Assuntos
Trifosfato de Adenosina/metabolismo , Carbono-Nitrogênio Ligases/genética , Carbono-Nitrogênio Ligases/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Adenosina/análogos & derivados , Adenosina/química , Adenosina/metabolismo , Trifosfato de Adenosina/química , Marcadores de Afinidade/química , Marcadores de Afinidade/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Carbono-Nitrogênio Ligases/antagonistas & inibidores , Carbono-Nitrogênio Ligases/química , Galinhas , Estabilidade de Medicamentos , Ativação Enzimática/genética , Estabilidade Enzimática/genética , Escherichia coli/crescimento & desenvolvimento , Cinética , Fígado/enzimologia , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/isolamento & purificação , Fragmentos de Peptídeos/metabolismo
8.
Biochemistry ; 38(10): 3012-8, 1999 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-10074353

RESUMO

Formation of 4-carboxy-5-aminoimidazole ribonucleotide (CAIR) in the purine pathway in most prokaryotes requires ATP, HCO3-, aminoimidazole ribonucleotide (AIR), and the gene products PurK and PurE. PurK catalyzes the conversion of AIR to N5-carboxyaminoimidazole ribonucleotide (N5-CAIR) in a reaction that requires both ATP and HCO3-. PurE catalyzes the unusual rearrangement of N5-CAIR to CAIR. To investigate the mechanism of this rearrangement, [4,7-13C]-N5-CAIR and [7-14C]-N5-CAIR were synthesized and separately incubated with PurE in the presence of ATP, aspartate, and 4-(N-succinocarboxamide)-5-aminoimidazole ribonucleotide (SAICAR) synthetase (PurC). The SAICAR produced was isolated and analyzed by NMR spectroscopy or scintillation counting, respectively. The PurC trapping of CAIR as SAICAR was required because of the reversibility of the PurE reaction. Results from both experiments reveal that the carboxylate group of the carbamate of N5-CAIR is transferred directly to generate CAIR without equilibration with CO2/HCO3- in solution. The mechanistic implications of these results relative to the PurE-only (CO2- and AIR-requiring) AIR carboxylases are discussed.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Carboxiliases/química , Escherichia coli/enzimologia , Ribonucleotídeos/química , Aminoimidazol Carboxamida/síntese química , Aminoimidazol Carboxamida/química , Isótopos de Carbono , Catálise , Sequência Conservada , Transporte de Elétrons , Lisina/química , Lisina/genética , Espectroscopia de Ressonância Magnética , Mutagênese Sítio-Dirigida , Ribonucleotídeos/síntese química
9.
Biochemistry ; 37(45): 15647-62, 1998 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-9843369

RESUMO

Glycinamide ribonucleotide synthetase (GAR-syn) catalyzes the second step of the de novo purine biosynthetic pathway; the conversion of phosphoribosylamine, glycine, and ATP to glycinamide ribonucleotide (GAR), ADP, and Pi. GAR-syn containing an N-terminal polyhistidine tag was expressed as the SeMet incorporated protein for crystallographic studies. In addition, the protein as isolated contains a Pro294Leu mutation. This protein was crystallized, and the structure solved using multiple-wavelength anomalous diffraction (MAD) phase determination and refined to 1.6 A resolution. GAR-syn adopts an alpha/beta structure that consists of four domains labeled N, A, B, and C. The N, A, and C domains are clustered to form a large central core structure whereas the smaller B domain is extended outward. Two hinge regions, which might readily facilitate interdomain movement, connect the B domain and the main core. A search of structural databases showed that the structure of GAR-syn is similar to D-alanine:D-alanine ligase, biotin carboxylase, and glutathione synthetase, despite low sequence similarity. These four enzymes all utilize similar ATP-dependent catalytic mechanisms even though they catalyze different chemical reactions. Another ATP-binding enzyme with low sequence similarity but unknown function, synapsin Ia, was also found to share high structural similarity with GAR-syn. Interestingly, the GAR-syn N domain shows similarity to the N-terminal region of glycinamide ribonucleotide transformylase and several dinucleotide-dependent dehydrogenases. Models of ADP and GAR binding were generated based on structure and sequence homology. On the basis of these models, the active site lies in a cleft between the large domain and the extended B domain. Most of the residues that facilitate ATP binding belong to the A or B domains. The N and C domains appear to be largely responsible for substrate specificity. The structure of GAR-syn allows modeling studies of possible channeling complexes with PPRP amidotransferase.


Assuntos
Escherichia coli/enzimologia , Hidroximetil e Formil Transferases/química , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Simulação por Computador , Cristalografia por Raios X , Hidroximetil e Formil Transferases/biossíntese , Hidroximetil e Formil Transferases/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Fosforribosilglicinamido Formiltransferase , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
10.
J Biol Chem ; 270(51): 30532-44, 1995 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-8530485

RESUMO

The non-heme iron-dependent metalloenzyme, rat hepatic phenylalanine hydroxylase (EC 1.14.16.1; phenylalanine 4-monooxygenase (PAH) was overexpressed in Escherichia coli and purified to homogeneity, allowing a detailed comparison of the kinetic, hydrodynamic, and spectroscopic properties of its allosteric states. The homotetrameric recombinant enzyme, which is highly active and contains 0.7-0.8 iron atoms per subunit, is identical to the native enzyme in several properties: Km, 6-methyltetrahydropterin = 61 microM and L-Phe = 170 microM; Vmax = 9 s-1 (compared to 45 microM, 180 microM, and 13 s-1 for the rat hepatic enzyme). L-Phe and lysolecithin treatment induce the rPAHT-->rPAHR (where r is recombinant) allosteric transformation necessary for rPAH activity. Characteristic changes in the fluorescence spectra, increased hydrophobicity, a large activation energy barrier, and a 10% volume increase of the tetrameric structure are consistent with a significant reorganization of the protein following allosteric activation. However, optical and EPR spectroscopic data suggest that only minor changes occur in the primary coordination sphere (carboxylate/histidine/water) of the catalytic iron center. Detailed steady state kinetic investigations, using 6-methyltetrahydropterin as cofactor and lysolecithin as activator, indicate rPAH follows a sequential mechanism. A catalytic Arrhenius Eact of 14.6 +/- 0.3 kcal/mol subunit was determined from temperature-dependent stopped-flow kinetics data. rPAH inactivates during L-Phe hydroxylation with a half-life of 4.3 min at 25 degrees C, corresponding to an Arrhenius Eact of 10 +/- 1 kcal/mol subunit for the inactivation process. Catechol binding (2.4 x 10(6) M-1) is shown to occur only at catalytically competent iron sites. Ferrous rPAH binds NO, giving rise to an ST = 3/2 spin system.


Assuntos
Fígado/enzimologia , Fenilalanina Hidroxilase/química , Fenilalanina Hidroxilase/metabolismo , Conformação Proteica , Regulação Alostérica , Animais , Western Blotting , Cromatografia em Gel , Cromatografia por Troca Iônica , Clonagem Molecular , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Escherichia coli , Expressão Gênica , Cinética , Lisofosfatidilcolinas/farmacologia , Substâncias Macromoleculares , Peso Molecular , Fenilalanina Hidroxilase/isolamento & purificação , Fosforilação , Plasmídeos , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Espectrometria de Fluorescência , Espectrofotometria , Termodinâmica
12.
Carcinogenesis ; 12(4): 671-8, 1991 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2013131

RESUMO

The ability of diverse carotenoid to inhibit methylcholanthrene-induced transformation of 10T1/2 cells has been investigated. When delivered using tetrahydrofuran as a novel solvent, all carotenoids were absorbed by cultured cells. When continuously administered to methylcholanthrene-treated cultures 7 days after removal of the carcinogen, canthaxanthin, beta-carotene, alpha-carotene and lycopene inhibited the production of transformed foci in a dose-dependent manner in the above order of potency. This activity was not associated with drug toxicity or antiproliferative effects. Renierapurpurin and bixin did not inhibit transformation at concentrations less than or equal to 10(-5) M. Lutein was inhibitory at 10(-5) M, but was inactive at lower concentrations. Because of differences in stability in culture medium (alpha-carotene less than beta-carotene less than canthaxanthin less than lycopene less than lutein) and structure, cellular levels of drug differed up to 8-fold after administration of identical concentrations of compounds. Carotenoids with polar groups achieved highest cellular levels, however cellular uptake did not correlate with activity. For example, lutein, the most polar and most stable, reached the highest concentration in cells yet required a concentration of 10(-5) M for activity in the transformation assay, while alpha-carotene, the least stable and least concentrated by cells, was comparably active at 3 X 10(6) M. alpha-Tocopherol, a potent lipid-phase antioxidant, was as active as lycopene in the transformation assay but at a 10-fold higher concentration did not approach the activity of beta-carotene or canthaxanthin. Because the most potent of the carotenoids tested (i.e. beta-carotene, alpha-carotene, canthaxanthin) all have the potential for conversion to retinoids (though this has never been demonstrated in mammals for canthaxanthin), it is suggested that these compounds have two components to their action; one related to their antioxidant properties, the other to their pro-vitamin A activities.


Assuntos
Carotenoides/farmacologia , Transformação Celular Neoplásica/induzido quimicamente , Animais , Carotenoides/farmacocinética , Carotenoides/toxicidade , Células Cultivadas , Furanos/farmacologia , Metilcolantreno , Camundongos , Veículos Farmacêuticos/farmacologia , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...