Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Bioorg Chem ; 57: 242-250, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25245459

RESUMEN

3-Deoxy-d-arabino-heptulosonate 7-phosphate synthase (DAH7PS) catalyses the first committed step of the shikimate pathway, which produces the aromatic amino acids as well as many other aromatic metabolites. DAH7PS catalyses an aldol-like reaction between phosphoenolpyruvate and erythrose 4-phosphate. Three phosphoenolpyruvate mimics, (R)-phospholactate, (S)-phospholactate and vinyl phosphonate [(E)-2-methyl-3-phosphonoacrylate], were found to competitively inhibit DAH7PS from Neisseria meningitidis, which is the pathogen responsible for bacterial meningitis. The most potent inhibitor was the vinyl phosphonate with a Ki value of 3.9±0.4µM. We report for the first time crystal structures of these compounds bound in the active site of a DAH7PS enzyme which reveals that the inhibitors bind to the active site of the enzyme in binding modes that mimic those of the predicted oxocarbenium and tetrahedral intermediates of the enzyme-catalysed reaction. Furthermore, the inhibitors accommodate the binding of a key active site water molecule. Together, these observations provide strong evidence that this active site water participates directly in the DAH7PS reaction, enabling the facial selectivity of the enzyme-catalysed reaction sequence to be delineated.


Asunto(s)
3-Desoxi-7-Fosfoheptulonato Sintasa/antagonistas & inhibidores , 3-Desoxi-7-Fosfoheptulonato Sintasa/metabolismo , Inhibidores Enzimáticos/química , Meningitis Meningocócica/microbiología , Neisseria meningitidis/enzimología , Fosfoenolpiruvato/análogos & derivados , 3-Desoxi-7-Fosfoheptulonato Sintasa/química , Dominio Catalítico , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Humanos , Meningitis Meningocócica/tratamiento farmacológico , Meningitis Meningocócica/enzimología , Modelos Moleculares , Neisseria meningitidis/química , Neisseria meningitidis/efectos de los fármacos , Fosfoenolpiruvato/farmacología , Agua/química
2.
Biochemistry ; 52(1): 1-3, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23256782

RESUMEN

A series of substrate analogues has been used to determine which chemical moieties of the substrate phosphoenolpyruvate (PEP) contribute to the allosteric inhibition of rabbit muscle pyruvate kinase by phenylalanine. Replacing the carboxyl group of the substrate with a methyl alcohol or removing the phosphate altogether greatly reduces substrate affinity. However, removal of the carboxyl group is the only modification tested that removes the ability to allosterically reduce the level of Phe binding. From this, it can be concluded that the carboxyl group of PEP is responsible for energetic coupling with Phe binding in the allosteric sites.


Asunto(s)
Músculos/enzimología , Fenilalanina/metabolismo , Fosfoenolpiruvato/química , Fosfoenolpiruvato/metabolismo , Piruvato Quinasa/metabolismo , Regulación Alostérica , Sitio Alostérico , Animales , Músculos/química , Fosfoenolpiruvato/análogos & derivados , Unión Proteica , Piruvato Quinasa/química , Conejos , Especificidad por Sustrato
3.
J Am Chem Soc ; 134(31): 12947-57, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22765168

RESUMEN

Proton transfer to carbon represents a significant catalytic challenge because of the large intrinsic energetic barrier and the frequently unfavorable thermodynamics. Multiple kinetic isotope effects (KIEs) were measured for acid-catalyzed hydrolysis of the enol ether functionality of enolpyruvylshikimate 3-phosphate (EPSP) as a nonenzymatic analog of the EPSP synthase (AroA) reaction. The large solvent deuterium KIE demonstrated that protonating C3 was the rate-limiting step, and the lack of solvent hydron exchange into EPSP demonstrated that protonation was irreversible. The reaction mechanism was stepwise, with C3, the methylene carbon, being protonated to form a discrete oxacarbenium ion intermediate before water attack at the cationic center, that is, an AH(‡)*AN (or AH(‡) + AN) mechanism. The calculated 3-(14)C and 3,3-(2)H2 KIEs varied as a function of the extent of proton transfer at the transition state, as reflected in the C3-H(+) bond order, nC3-H+. The calculated 3-(14)C KIE was a function primarily of C3 coupling with the movement of the transferring proton, as reflected in the reaction coordinate contribution ((light)ν(‡)/(heavy)ν(‡)), rather than of changes in bonding. Coupling was strongest in early and late transition states, where the reaction coordinate frequency was lower. The other calculated (14)C and (18)O KIEs were more sensitive to interactions with counterions and solvation in the model structures than nC3-H+. The KIEs revealed a moderately late transition state with significant oxacarbenium ion character and with a C3-H(+) bond order ≈0.6.


Asunto(s)
Éteres/química , Hidrólisis , Fosfoenolpiruvato/análogos & derivados , Fosfoenolpiruvato/química , Protones , Ácido Shikímico/análogos & derivados , Ácido Shikímico/química , 3-Fosfoshikimato 1-Carboxiviniltransferasa/metabolismo , Radioisótopos de Carbono , Deuterio , Cinética , Isótopos de Oxígeno , Teoría Cuántica
4.
Biol Pharm Bull ; 35(4): 606-11, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22466568

RESUMEN

This study examined the cytoprotective and anti-oxidative properties of phosphoenolpyruvic acid (PEP), a glycolysis metabolite with a high-energy phosphate group. PEP (0.1-10 mM) significantly attenuated the decrease in cell viability induced by hydrogen peroxide (H(2)O(2)) in HeLa cells in a dose-dependent manner. PEP also inhibited the decrease in calcein-acetomethoxy-stained cells and the increase in propidium iodide-stained cells that were induced by H(2)O(2). The H(2)O(2)-stimulated increase in intracellular reactive oxygen species was significantly reduced by PEP. PEP also demonstrated scavenging potential against hydroxyl radicals, as assessed by the electron paramagnetic resonance method. In addition, PEP demonstrated scavenging potential against the 1,1-diphenyl-2-picrylhydrazyl radical, a representative artificial radical, although the potential is very weak. PEP (10 mM) slightly inhibited the decrease in cellular ATP content induced by H(2)O(2), but did not show any effects at low doses (0.1, 1 mM). PEP (0.1-10 mM) also attenuated the cell injury but not the decrease in intracellular ATP content, induced by 2-deoxy-D-glucose, a glycolysis inhibitor. These results indicate that PEP exerts cytoprotective effects and has anti-oxidative potential, although the precise cytoprotective mechanisms are not fully elucidated. We suggest that PEP is a functional carbohydrate metabolite with cytoprotective and anti-oxidative activity, and is potentially useful as a therapeutic agent against diseases that involve the oxidative stress.


Asunto(s)
Antioxidantes/farmacología , Citoprotección/efectos de los fármacos , Fosfoenolpiruvato/análogos & derivados , Adenosina Trifosfato/antagonistas & inhibidores , Adenosina Trifosfato/metabolismo , Supervivencia Celular/efectos de los fármacos , Desoxiglucosa/farmacología , Glucólisis , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Oxidantes/farmacología , Fosfoenolpiruvato/farmacología
5.
Bioorg Chem ; 40(1): 48-56, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22204982

RESUMEN

We herein report two innovative methods toward aldehyde enolphosphates and the first saccharidic aldehyde enolphosphates. Aldehyde enolphosphate function is worthwhile to be considered as a good phosphoenolpyruvate analogue.


Asunto(s)
Aldehídos/química , Fosfoenolpiruvato/análogos & derivados , Aluminio/química , Yoduros/química , Monosacáridos/química , Fosfoenolpiruvato/síntesis química , Fosforilación
6.
Biochemistry ; 44(19): 7326-35, 2005 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-15882071

RESUMEN

The enzymes 3-deoxy-d-manno-2-octulosonate-8-phosphate (KDO8P) synthase and 3-deoxy-d-arabino-2-heptulosonate-7-phosphate (DAHP) synthase catalyze a similar aldol-type condensation between phosphoenolpyruvate (PEP) and the corresponding aldose: arabinose 5-phosphate (A5P) and erythrose 4-phosphate (E4P), respectively. While KDO8P synthase is metal-dependent in one class of organisms and metal-independent in another, only a metal-dependent class of DAHP synthases has thus far been identified in nature. We have used catalytically active E and Z isomers of phosphoenol-3-fluoropyruvate [(E)- and (Z)-FPEP, respectively] as mechanistic probes to characterize the differences and/or the similarities between the metal-dependent and metal-independent KDO8P synthases as well as between the metal-dependent KDO8P synthase and DAHP synthase. The direct evidence of the overall stereochemistry of the metal-dependent Aquifex pyrophilus KDO8P synthase (ApKDO8PS) reaction was obtained by using (E)- and (Z)-FPEPs as alternative substrates and by subsequent (19)F NMR analysis of the products. The results reveal the si face addition of the PEP to the re face of the carbonyl of A5P, and establish that the stereochemistry of ApKDO8PS is identical to that of the metal-independent Escherichia coli KDO8P synthase enzyme (EcKDO8PS). In addition, both ApKDO8PS and EcKDO8PS enzymes exhibit high selectivity for (E)-FPEP versus (Z)-FPEP, the relative k(cat)/K(m) ratios being 100 and 33, respectively. In contrast, DAHP synthase does not discriminate between (E)- and (Z)-FPEP (the k(cat)/K(m) being approximately 7 x 10(-)(3) microM(-)(1) s(-)(1) for both compounds). The pre-steady-state burst experiments for EcKDO8PS showed that product release is rate-limiting for the reactions performed with either PEP, (E)-FPEP, or (Z)-FPEP, although the rate constants, for both product formation and product release, were lower for the fluorinated analogues than for PEP [125 and 2.3 s(-)(1) for PEP, 2.5 and 0.2 s(-)(1) for (E)-FPEP, and 9 and 0.1 s(-)(1) for (Z)-FPEP, respectively]. The observed data indicate substantial differences in the PEP subsites and open the opportunity for the design of selective inhibitors against these two families of enzymes.


Asunto(s)
3-Desoxi-7-Fosfoheptulonato Sintasa/metabolismo , Aldehído-Liasas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Fosfoenolpiruvato/análogos & derivados , Fosfoenolpiruvato/metabolismo , 3-Desoxi-7-Fosfoheptulonato Sintasa/química , Aldehído-Liasas/química , Proteínas Bacterianas/química , Sitios de Unión , Radioisótopos de Carbono/metabolismo , Catálisis , Cromatografía Líquida de Alta Presión , Proteínas de Escherichia coli/química , Cinética , Pentosafosfatos/química , Pentosafosfatos/metabolismo , Fosfoenolpiruvato/química , Estereoisomerismo , Especificidad por Sustrato , Fosfatos de Azúcar/química , Fosfatos de Azúcar/metabolismo
7.
Drug Des Discov ; 18(2-3): 91-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14675946

RESUMEN

3-Deoxy-D-manno-octulosonate 8-phosphate (KDO8P) is the phosphorylated precursor of KDO, an essential sugar of the lipopolysaccharide of Gram negative bacteria. KDO8P is produced by a specific synthase (KDO8PS) by condensing arabinose 5-phosphate (A5P) and phosphoenolpyruvate (PEP), with release of inorganic phosphate. As KDO8PS is present in bacteria and plants, but not in mammalian cells, and mutations that inactivate KDO8PS also block cell replication, KDO8PS is a promising target for the design of new antimicrobials that act by blocking lipopolysaccharide biosynthesis. Previous studies have shown that a compound mimicking an intermediate of the condensation reaction is a good ligand and a powerful inhibitor. Here we report on the crystallographic investigation of the binding to KDO8PS of new derivatives of this original inhibitor. The structures of the enzyme in complex with these compounds, and also with the PEP analogs, 2-phosphoglyceric acid (2-PGA) and Z-methyl-PEP, point to future strategies for the design of novel inhibitors of KDO8PS.


Asunto(s)
Aldehído-Liasas/antagonistas & inhibidores , Aldehído-Liasas/química , Sitios de Unión , Cristalografía por Rayos X , Ácidos Glicéricos/química , Bacterias Gramnegativas/química , Modelos Moleculares , Fosfoenolpiruvato/análogos & derivados , Fosfoenolpiruvato/química , Unión Proteica , Estereoisomerismo , Relación Estructura-Actividad , Azúcares Ácidos/química
8.
Biochemistry ; 42(23): 6986-95, 2003 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-12795593

RESUMEN

AroA (EPSP synthase) catalyzes carboxyvinyl transfer through addition of shikimate 3-phosphate (S3P) to phosphoenolpyruvate (PEP) to form a tetrahedral intermediate (THI), followed by phosphate elimination to give enolpyruvylshikimate 3-phosphate (EPSP). A novel approach, partitioning analysis, was used to elucidate the roles of catalytic residues in each step of the reaction. Partitioning analysis involved trapping and purifying [1-(14)C]THI, degrading it with AroA, and quantitating the products. Wild-type AroA gave a partitioning factor, f(PEP) = 0.25 +/- 0.02 at pH 7.5, where f(PEP) = [[1-(14)C]PEP]/([[1-(14)C]PEP] + [[1-(14)C]EPSP]). Eighteen mutations were made to 14 amino acids to discover which residues preferentially catalyzed either the addition or the elimination step. Mutating a residue catalyzing one step (e.g., addition) should change f(PEP) to favor the opposite step (e.g., elimination). No mutants caused large changes in f(PEP), with experimental values from 0.07 to 0.41. This implied that there are no side chains that catalyze only addition or elimination, which further implied that the same residues are general acid/base catalysts in both forward and reverse THI breakdown. Only Lys22 (protonating S3P hydroxyl or phosphate) and Glu341 (deprotonating C3 of PEP) are correctly situated in the active site. In the overall reaction, Lys22 would act as a general base during addition, while Glu341 would act as a general acid. Almost half of the mutations (eight of 18) caused a >1000-fold decrease in specific activity, demonstrating that a large number of residues are important for transition state stabilization, "ensemble catalysis", in contrast to some enzymes where a single amino acid can be responsible for up to 10(8)-fold catalytic enhancement.


Asunto(s)
Transferasas Alquil y Aril/metabolismo , 3-Fosfoshikimato 1-Carboxiviniltransferasa , Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/genética , Sustitución de Aminoácidos , Sitios de Unión , Isótopos de Carbono , Catálisis , Simulación por Computador , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Lisina/genética , Lisina/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Análisis Numérico Asistido por Computador , Fosfoenolpiruvato/análogos & derivados , Fosfoenolpiruvato/química , Fosfoenolpiruvato/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Ácido Shikímico/análogos & derivados , Ácido Shikímico/metabolismo
9.
Biochemistry ; 42(16): 4744-50, 2003 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-12705838

RESUMEN

Enzyme I (EI), the first component of the phosphoenolpyruvate (PEP):sugar phosphotransferase system (PTS), consists of an N-terminal domain with the phosphorylation site (His-189) and a C-terminal domain with the PEP binding site. Here we use C3-substituted PEP analogues as substrates and inhibitors and the EI(C502A) mutant to characterize structure-activity relationships of the PEP binding site. EI(C502A) is 10 000 times less active than wild-type EI [EI(wt)] with PEP as the substrate, whereas the two forms are equally active with ZClPEP. Cys-502 acts as an acid-base catalyst which stereospecifically protonates the pyruvoyl enolate at C3. The electron-withdrawing chlorine of ZClPEP can compensate for the lack of Cys-502, and in this case, the released 3-Cl-enolate is protonated nonstereospecifically. Several PEP analogues were assayed as inhibitors and as substrates. The respective K(I)/K(m) ratios vary between 3 and 40 for EI(wt), but they are constant and around unity for EI(C502A). EI(wt) with PEP as the substrate is inhibited by oxalate, whereas EI(C502A) with ZClPEP is not. The different behavior of EI(wt) and EI(C502A) toward the PEP analogues and oxalate suggests that the PEP binding site of EI(wt) exists in a "closed" and an "open" form. The open to closed transition is triggered by the interaction of the substrate with Cys-502. The closed conformation is sterically disfavored by C3-modified substrate analogues such as ZClPEP and ZMePEP. If site closure does not occur as with EI(C502A) and bulky substrates, the transition state is stabilized by electron dispersion to the electron-withdrawing substituent at C3.


Asunto(s)
Cisteína/química , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/química , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Fosfotransferasas (Aceptor del Grupo Nitrogenado)/química , Fosfotransferasas (Aceptor del Grupo Nitrogenado)/metabolismo , Sitios de Unión , Catálisis , Cisteína/fisiología , Dimerización , Inhibidores Enzimáticos/metabolismo , Isomerismo , Cinética , Mutación , Oxalatos/metabolismo , Fosfoenolpiruvato/análogos & derivados , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Fosfotransferasas (Aceptor del Grupo Nitrogenado)/genética , Conformación Proteica , Protones , Relación Estructura-Actividad
10.
J Am Chem Soc ; 125(15): 4662-9, 2003 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-12683839

RESUMEN

In this report the mode of inhibition of mechanism-based inhibitor (2, K(i) = 0.4 microM) of 3-deoxy-d-manno-2-octulosonate-8-phosphate synthase (KDO8PS), which was designed to mimic the combined key features of its natural substrates arabinose-5-phosphate (A5P) and phoshoenolpyruvate (PEP) into a single molecule, was investigated. Our earlier solid-state NMR observations identified the inhibitor to bind in a way that partly mimics A5P, while the phosphonate moiety of its PEP-mimicking part exhibits no interactions with enzyme residues. This result was apparently in disagreement with the competitive inhibition of 2 against PEP and with the later solved crystal structure of KDO8PS-2 binary complex identifying the interactions of its PEP-mimicking part with the enzyme residues that were not detected by solid-state NMR. To solve this discrepancy, further solid-state REDOR NMR and (31)P solution NMR experiments were applied to a variety of enzyme complexes with the substrates and inhibitor. In particular, a novel frequency-selective REDOR experiment was developed and applied. Integration of the solution and solid-state NMR data clearly demonstrates that under conditions of stoichiometric enzyme-ligand ratio at thermodynamic equilibrium (a) PEP binding is unperturbed by the presence of 2 and (b) both PEP and 2 can bind simultaneously to the synthase, i.e., form a ternary complex with PEP occupying its own subsite and 2 occupying A5P's subsite. The latter observation suggests that under the conditions used in our NMR measurements, the inhibition pattern of 2 against PEP should have a mixed type character. Furthermore, the NMR data directly demonstrate the distinction between the relative binding strength of the two moieties of 2: enzyme interactions with PEP-mimicking moiety are much weaker than those with the A5P moiety. This observation is in agreement with KDO8PS-2 crystal structure showing only remote contacts of the phosphonate due to large structural changes of binding site residues. It is concluded that these phosphonate-enzyme interactions evidenced by both (31)P solution NMR and X-ray are too weak to be preserved under the lyophilization of KDO8PS-2 binary complex and therefore are not evidenced by the solid-state REDOR spectra.


Asunto(s)
Aldehído-Liasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Resonancia Magnética Nuclear Biomolecular/métodos , Pentosafosfatos/química , Fosfoenolpiruvato/análogos & derivados , Aldehído-Liasas/metabolismo , Sitios de Unión , Unión Competitiva , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Pentosafosfatos/metabolismo , Fosfoenolpiruvato/química , Fosfoenolpiruvato/metabolismo , Fósforo/química
11.
Eur J Biochem ; 269(13): 3226-36, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12084063

RESUMEN

The synthesis of 10 new phosphoenolpyruvate (PEP) analogues with modifications in the phosphate and the carboxylate function is described. Included are two potential irreversible inhibitors of PEP-utilizing enzymes. One incorporates a reactive chloromethylphosphonate function replacing the phosphate group of PEP. The second contains a chloromethyl group substituting for the carboxylate function of PEP. An improved procedure for the preparation of the known (Z)- and (E)-3-chloro-PEP is also given. The isomers were obtained as a 4 : 1 mixture, resolved by anion-exchange chromatography after the last reaction step. The stereochemistry of the two isomers was unequivocally assigned from the (3)J(H-C) coupling constants between the carboxylate carbons and the vinyl protons. All of these and other known PEP-analogues were tested as reversible and irreversible inhibitors of Mg2+- and Mn2+- activated PEP-utilizing enzymes: enzyme I of the phosphoenolpyruvate:sugar phosphotransferase system (PTS), pyruvate kinase, PEP carboxylase and enolase. Without exception, the most potent inhibitors were those with substitution of a vinyl proton. Modification of the phosphate and the carboxylate groups resulted in less effective compounds. Enzyme I was the least tolerant to such modifications. Among the carboxylate-modified analogues, only those replaced by a negatively charged group inhibited pyruvate kinase and enolase. Remarkably, the activity of PEP carboxylase was stimulated by derivatives with neutral groups at this position in the presence of Mg2+, but not with Mn2+. For the irreversible inhibition of these enzymes, (Z)-3-Cl-PEP was found to be a very fast-acting and efficient suicide inhibitor of enzyme I (t(1/2) = 0.7 min).


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/antagonistas & inhibidores , Fosfoenolpiruvato/química , Fosfoenolpiruvato/farmacología , Bioquímica/métodos , Evaluación Preclínica de Medicamentos , Activación Enzimática , Inhibidores Enzimáticos/metabolismo , Isomerismo , Fosfoenolpiruvato/análogos & derivados , Fosfoenolpiruvato/metabolismo , Fosfoenolpiruvato Carboxilasa/antagonistas & inhibidores , Fosfoenolpiruvato Carboxilasa/metabolismo , Fosfopiruvato Hidratasa/antagonistas & inhibidores , Fosfopiruvato Hidratasa/metabolismo , Fosfotransferasas (Aceptor del Grupo Nitrogenado)/antagonistas & inhibidores , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/metabolismo , Relación Estructura-Actividad
12.
Biochim Biophys Acta ; 1386(1): 132-44, 1998 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-9675261

RESUMEN

The antibiotic phosphomycin (1,2-epoxypropylphosphonic acid), an analog of phosphoenolpyruvate (PEP), behaved not as an inhibitor, but as an activator, of the enzyme phosphoenolpyruvate carboxylase (PEPC) from maize leaves. Multiple activation studies indicated that the analog binds to the Glc6P-allosteric site producing a more activated enzyme than Glc6P itself. Because of this, we used phosphomycin as a tool to further extend our understanding of the mechanisms of allosteric regulation of C4-PEPC. Initial velocity data from detailed kinetic studies, in which the concentrations of free and Mg-complexed PEP and phosphomycin were controlled, are consistent with: (1) the true activator is free phosphomycin, which competes with free PEP for the Glc6P-allosteric site; and (2) the Mg-phosphomycin complex caused inhibition by binding to the active site in competition with MgPEP. Therefore, although the Glc6P-allosteric site and the active site are able to bind the same ligands, they differ in the form of substrate and activator they bind. This important difference allows the full expression of the potential of activation and prevents inhibition by the activators, including the physiological ones, which are mostly uncomplexed at physiological free Mg2+ concentrations. At fixed low substrate concentrations, the saturation kinetics of the enzyme by phosphomycin showed positive cooperativity at pH 7.3 and 8.3, although at the latter pH, the kinetics of saturation by the substrate was hyperbolic. The cosolute glycerol greatly increased the affinity of the enzyme for phosphomycin and abolished the cooperativity in its binding, but did not eliminate the heterotropic effects of the activator. Therefore, the heterotropic and homotropic effects of the activator are not always coupled to the homotropic effects of the substrate, which argues against the two-state model previously proposed to explain the allosteric properties of maize-leaf PEPC.


Asunto(s)
Fosfomicina/metabolismo , Fosfoenolpiruvato Carboxilasa/metabolismo , Fosfoenolpiruvato/análogos & derivados , Hojas de la Planta/enzimología , Zea mays/enzimología , Regulación Alostérica , Interacciones Farmacológicas , Activación Enzimática , Fosfomicina/farmacología , Glicerol/farmacología , Cinética , Magnesio/metabolismo , Modelos Químicos
13.
Acta Crystallogr B ; 52 ( Pt 5): 865-75, 1996 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-8900032

RESUMEN

The crystal structure of (C6H11NH3+)3. Pep3-.H2O, where Pep3- = (O-)2P(O)-O-C(CH2)-CO2-, is reported and the systematic structural variations among 19 crystallographic occurrences of H3Pep, H2Pep-, HPep2- and Pep3- species, which are important phosphate donors in the ATP cycle of bioenergetics, are reviewed. Tris(cyclohexylammonium) phosphoenolpyruvate monohydrate, (C6H11NH3+)3.-[O3POC(CH2)CO2]3-.H2O, M(r) = 483.6, m.p. 418-420K; T = 296(1)K; orthorhombic, P2(1)2(1)2(1); a = 16.7042(5), b = 24.4881 (6), c = 6.38910 (10) A; V = 2613.49(11) A3, Z = 4, Dx = 1.23, Dm = 1.22 mg mm-3, mu = 0.14 mm-1 for lambda(MoK alpha) = 0.7107 A; F(000) = 1056 e; R(magnitude of F) = 0.0608 for 6056 hkl and hkl data with (sin theta)/lambda < or = 0.65 A-1.


Asunto(s)
Fosfoenolpiruvato/análogos & derivados , Cristalografía por Rayos X , Enlace de Hidrógeno , Estructura Molecular
14.
Biochemistry ; 35(5): 1692-9, 1996 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-8634301

RESUMEN

High-resolution crystallographic data show that Glu 168 and Glu 211 lie on opposite surfaces of the active site from Lys 345. Two different proposals for general base catalysis have emerged from these structural studies. In one scheme, the carboxylate side chains of Glu 168 and Glu 211 are proposed to ionize a trapped water molecule and the OH- serves as the base [Lebioda, L., & Stec, B. (1991) Biochemistry 30, 2817-2822]. In the other proposal, the epsilon-amino group of Lys 345 functions in general base catalysis [Wedekind, J. E., Poyner, R. R., Reed, G. H., & Rayment, I. (1994) Biochemistry 33, 9333-9342]. Genes encoding site specific mutations of these active site residues of yeast enolase, K345A, E168Q, and E211Q, have been prepared. The respective protein products of the wild type and mutant genes were expressed in Escherichia coli and isolated in homogeneous form. All three mutant proteins possess severely depressed activities in the overall reaction- < 1 part in 10(5) of wild type activity. Properties of the three mutant proteins in partial reactions were examined to define more clearly the roles of these residues in the catalytic cycle. The K345A variant fails to catalyze the exchange of the C-2 proton of 2-phospho-D-glycerate with deuterium in D2O, whereas both the E211Q and E168Q mutant proteins are functional in this partial reaction. For E211Q and E168Q enolases, exchange is essentially complete prior to appearance of product, and this observation provides further support for an intermediate in the normal reaction. K345A enolase is inactive in the ionization of tartronate semialdehyde phosphate (TSP), whereas both E168Q and E211Q proteins alter the tautomeric state or catalyze ionization of bound TSP. Wild type enolase catalyzes hydrolysis of (Z)-3-chloro-2-phosphoenolpyruvate by addition of OH- and elimination of Cl- at C-3. This reaction mimics the addition of OH- to C-3 of phosphoenolpyruvate in the reverse reaction with the normal product. All three mutant proteins are depressed in their abilities to carry out this reaction. In single-turnover assays, the activities vary in the order K345A > E168Q >> E211Q. These results suggest that Lys 345 functions as the base in the ionization of 2-PGA and that Glu 211 participates in the second step of the reaction.


Asunto(s)
Fosfopiruvato Hidratasa/metabolismo , Secuencia de Bases , Sitios de Unión , Catálisis , Ácidos Glicéricos/metabolismo , Hidrólisis , Modelos Químicos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosfoenolpiruvato/análogos & derivados , Fosfoenolpiruvato/metabolismo , Fosfopiruvato Hidratasa/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Tartronatos/metabolismo , Levaduras/enzimología , Levaduras/genética
15.
Nat Struct Biol ; 2(8): 644-53, 1995 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7552726

RESUMEN

The crystal structure of UDP-N-acetylenolpyruvylglucosamine reductase in the presence of its substrate, enolpyruvyl-UDP-N-acetylglucosamine, has been solved to 2.7 A resolution. This enzyme is responsible for the synthesis of UDP-N-acetylmuramic acid in bacterial cell wall biosynthesis and consequently provides an attractive target for the design of antibacterial agents. The structure reveals a novel flavin binding motif, shows a striking alignment of the flavin with the substrate, and suggests a catalytic mechanism for the reduction of this unusual enol ether.


Asunto(s)
Deshidrogenasas de Carbohidratos/química , Deshidrogenasas de Carbohidratos/metabolismo , Fosfoenolpiruvato/análogos & derivados , Estructura Secundaria de Proteína , Uridina Difosfato N-Acetilglucosamina/análogos & derivados , Secuencia de Aminoácidos , Antiinfecciosos/química , Sitios de Unión , Pared Celular/metabolismo , Gráficos por Computador , Simulación por Computador , Cristalografía por Rayos X , Diseño de Fármacos , Flavina-Adenina Dinucleótido/metabolismo , Enlace de Hidrógeno , Ligandos , Modelos Moleculares , Modelos Estructurales , Datos de Secuencia Molecular , Fosfoenolpiruvato/química , Fosfoenolpiruvato/metabolismo , Uridina Difosfato N-Acetilglucosamina/química , Uridina Difosfato N-Acetilglucosamina/metabolismo , Uridina Difosfato Ácido N-Acetilmurámico/química , Uridina Difosfato Ácido N-Acetilmurámico/metabolismo
16.
Bioorg Med Chem ; 3(3): 217-25, 1995 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7606383

RESUMEN

The quantitative structure-activity relationships (QSAR) of all known competitive inhibitors of the enzyme phosphoenolpyruvate carboxylase from C4 plants were investigated by means of molecular mechanics, the semiempirical quantum chemical methods MNDO and AM1, and the Hansch approach. In the case of phosphoenolpyruvate analogues, the hydrophobicity and steric impediment of the combined cis and trans substituents, the bond distance to the cis substituent along with its volume, dipole moment, the distance between the phosphorus and the carbonyl carbon, and the net electric charges on the phosphate and substituent groups are the main factors that govern their binding to the active site. For the phosphoglycolate analogues, the difference in the HOMO-LUMO energies, the magnitudes of their dipole moments and their non-polar surfaces, and the distance between the phosphorus and the carbonyl carbon are the variables that control their binding to the active site. These results, in conjunction with a discriminant analysis, also suggest that these inhibitors can actually be divided into two groups, according to the way they presumably interact with the active site.


Asunto(s)
Inhibidores Enzimáticos/química , Fosfoenolpiruvato Carboxilasa/antagonistas & inhibidores , Fosfoenolpiruvato/análogos & derivados , Sitios de Unión , Unión Competitiva , Inhibidores Enzimáticos/farmacología , Glicolatos/farmacología , Fosfoenolpiruvato/metabolismo , Fosfoenolpiruvato/farmacología , Fosfoenolpiruvato Carboxilasa/metabolismo , Ácido Fosfonoacético/farmacología , Relación Estructura-Actividad
17.
Eur J Biochem ; 209(2): 735-43, 1992 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-1330557

RESUMEN

The enzyme carboxyphosphonoenolpyruvate mutase catalyses the formation of one of the two C-P bonds in bialaphos, a potent herbicide isolated from Streptomyces hygroscopicus. The gene encoding the enzyme has been cloned from a subgenomic library from S. hygroscopicus by colony hybridisation using an exact nucleotide probe. An open reading frame has been identified that encodes a protein of molecular mass 32700 Da, in good agreement with the subunit molecular mass of the carboxyphosphonoenolpyruvate mutase recently isolated from this source [Hidaka, T., Imai, S., Hara, O., Anzai, H., Murakami, T., Nagaoka, K. & Seto, H. (1990) J. Bacteriol. 172, 3066-3072]. The gene shares significant sequence similarity with that of phosphoenolpyruvate mutase, an enzyme that catalyses the related interconversion of phosphoenolpyruvate and phosphonopyruvate. When the carboxyphosphonoenolpyruvate-mutase gene was subcloned into the vector pET11a, the mutase was expressed as about 20% of the total soluble cellular protein in Escherichia coli. The mutase has been purified to homogeneity in three steps in 40% yield. With malate dehydrogenase/NADH, (hydroxyphosphinyl)pyruvate gives (hydroxyphosphinyl)lactate (kcat 164 s-1 and Km 680 microM) and this spectrophotometric assay for the product of the mutase reaction has been employed in the mechanistic studies. The kinetics for the mutase reaction have been evaluated for the substrate, carboxyphosphonoenolpyruvate, and for the putative reaction intermediate carboxyphosphinopyruvate, both of which have been prepared by chemical synthesis. Carboxyphosphonoenolpyruvate is converted to (hydroxyphosphinyl)pyruvate with a kcat of 0.020 s-1 and a Km of 270 microM, and carboxyphosphinopyruvate is converted to (hydroxyphosphinyl)pyruvate with a kcat of 7.6 x 10(-4) s-1 and a Km of 2.2 microM. Although the exogenously added intermediate is not kinetically competent, these results suggest that the mechanism for the mutase reaction involves an initial rearrangement to the intermediate carboxyphosphinopyruvate, followed by decarboxylation to yield the product (hydroxyphosphinyl)pyruvate.


Asunto(s)
Genes Bacterianos , Lactatos , Fosfotransferasas (Fosfomutasas) , Fosfotransferasas/metabolismo , Piruvatos , Streptomyces/enzimología , Secuencia de Aminoácidos , Secuencia de Bases , Cromatografía , Clonación Molecular , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Escherichia coli/genética , Expresión Génica , Biblioteca Genómica , Cinética , Datos de Secuencia Molecular , Peso Molecular , Oligodesoxirribonucleótidos , Sistemas de Lectura Abierta , Compuestos Organofosforados/síntesis química , Compuestos Organofosforados/metabolismo , Fosfoenolpiruvato/análogos & derivados , Fosfoenolpiruvato/síntesis química , Fosfoenolpiruvato/metabolismo , Fosfotransferasas/genética , Fosfotransferasas/aislamiento & purificación , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Mapeo Restrictivo , Streptomyces/genética , Especificidad por Sustrato
18.
Biochemistry ; 31(28): 6432-40, 1992 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-1633157

RESUMEN

The catalytic mechanism of phosphoenolpyruvate (PEP) carboxylase from Zea mays has been studied using (Z)- and (E)-3-fluorophosphoenolpyruvate (F-PEP) as substrates. Both (Z)- and (E)-F-PEP partition between carboxylation to produce 3-fluorooxalacetate and hydrolysis to produce 3-fluoropyruvate. Carboxylation accounts for 3% of the reaction observed with (Z)-F-PEP, resulting in the formation of (R)-3-fluorooxalacetate, and for 86% of the reaction of (E)-F-PEP forming (S)-3-fluorooxalacetate. Carboxylation of F-PEP occurs on the 2-re face, which corresponds to the 2-si face of PEP. The partitioning of F-PEP between carboxylation and hydrolysis is insensitive to pH but varies with metal ion. Use of 18O-labeled bicarbonate produces phosphate that is multiply labeled with 18O; in addition, 18O is also incorporated into residual (Z)- and (E)-F-PEP. The 13(V/K) isotope effect on the carboxylation of F-PEP catalyzed by PEP carboxylase at pH 8.0, 25 degrees C, is 1.049 +/- 0.003 for (Z)-F-PEP and 1.009 +/- 0.006 for (E)-F-PEP. These results are consistent with a mechanism in which carboxylation of PEP occurs via attack of the enolate of pyruvate on CO2 rather than carboxy phosphate. In this mechanism phosphorylation of bicarbonate to give carboxy phosphate and decarboxylation of the latter are reversible steps. An irreversible step, however, precedes partitioning between carboxylation to give oxalacetate and release of CO2, which results in hydrolysis of PEP.


Asunto(s)
Fosfoenolpiruvato Carboxilasa/metabolismo , Fosfoenolpiruvato/análogos & derivados , Fosfoenolpiruvato/metabolismo , Zea mays/enzimología , Bicarbonatos/metabolismo , Dióxido de Carbono/metabolismo , Cationes Bivalentes , Concentración de Iones de Hidrógeno , Hidrólisis , Isomerismo , Cinética , Espectroscopía de Resonancia Magnética , Malato Deshidrogenasa/metabolismo , Relación Estructura-Actividad
19.
Biochemistry ; 31(24): 5534-44, 1992 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-1344882

RESUMEN

Previous studies of Escherichia coli 5-enolpyruvoylshikimate-3-phosphate synthase (EPSPS, EC 2.5.1.19) have suggested that the kinetic reaction mechanism for this enzyme in the forward direction is equilibrium ordered with shikimate 3-phosphate (S3P) binding first followed by phosphoenolpyruvate (PEP). Recent results from this laboratory, however, measuring direct binding of PEP and PEP analogues to free EPSPS suggest more random character to the enzyme. Steady-state kinetic and spectroscopic studies presented here indicate that E. coli EPSPS does indeed follow a random kinetic mechanism. Initial velocity studies with S3P and PEP show competitive substrate inhibition by PEP added to a normal intersecting pattern. Substrate inhibition is proposed to occur by competitive binding of PEP at the S3P site [Ki(PEP) = 6-8 mM]. To test for a productive EPSPS.PEP binary complex, the reaction order of EPSPS was evaluated with shikimic acid and PEP as substrates. The mechanism for this reaction is equilibrium ordered with PEP binding first giving a Kia value for PEP in agreement with the independently measured Kd of 0.39 mM (shikimate Km = 25 mM). Results from this study also show that the 3-phosphate moiety of S3P offers 8.7 kcal/mol in binding energy versus a hydroxyl in this position. Over 60% of this binding energy is expressed in binding of substrate to enzyme rather than toward increasing kcat. Glyphosate inhibition of shikimate turnover was poor with approximately 8 x 10(4) loss in binding capacity compared to the normal reaction, consistent with the independently measured Kd of 12 mM for the EPSPS.glyphosate binary complex. The EPSPS.glyphosate complex induces shikimate binding, however, by a factor of 7 greater than EPSPS.PEP. Carboxyallenyl phosphate and (Z)-3-fluoro-PEP were found to be strong inhibitors of the enzyme that have surprising affinity for the S3P binding domain in addition to the PEP site as measured both kinetically and by direct observation with 31P NMR. The collective data indicate that the true kinetic mechanism for EPSPS in the forward direction is random with synergistic binding occurring between substrates and inhibitors. The synergism explains how the mechanism can be random with S3P and PEP, but yet equilibrium ordered with PEP binding first for shikimate turnover. Synergism also accounts for how glyphosate can be a strong inhibitor of the normal reaction, but poor versus shikimate turnover.


Asunto(s)
Transferasas Alquil y Aril , Escherichia coli/enzimología , Transferasas/metabolismo , 3-Fosfoshikimato 1-Carboxiviniltransferasa , Glicina/análogos & derivados , Glicina/farmacología , Cinética , Espectroscopía de Resonancia Magnética , Fosfoenolpiruvato/análogos & derivados , Fosfoenolpiruvato/metabolismo , Ácido Shikímico/análogos & derivados , Ácido Shikímico/metabolismo , Transferasas/antagonistas & inhibidores , Transferasas/química , Glifosato
20.
Arch Biochem Biophys ; 277(1): 143-8, 1990 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-2306115

RESUMEN

(Z)-3-Chlorophosphoenolpyruvate has been synthesized by the reaction of 3,3-dichloropyruvic acid with trimethylphosphite, followed by deesterification. This compound is a competitive inhibitor of pyruvate kinase and phosphoenolpyruvate carboxylase. Pyruvate kinase is not inactivated upon prolonged incubation with the compound, but phosphoenolpyruvate carboxylase is slowly inactivated (t1/2 = 5 h). The compound is a substrate for both enzymes, being acted upon by pyruvate kinase approximately 0.1% as rapidly as phosphoenolpyruvate itself. In the case of phosphoenolpyruvate carboxylase, the compound is converted into a 3:1 mixture of chloropyruvate and chlorooxalacetate, at an overall rate that is about 25% the carboxylation rate for phosphoenolpyruvate.


Asunto(s)
Carboxiliasas/antagonistas & inhibidores , Fosfoenolpiruvato Carboxilasa/antagonistas & inhibidores , Fosfoenolpiruvato/análogos & derivados , Piruvato Quinasa/antagonistas & inhibidores , Unión Competitiva , Indicadores y Reactivos , Isomerismo , Fosfoenolpiruvato/síntesis química , Fosfoenolpiruvato/farmacología , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA