Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Open Biol ; 12(9): 220179, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36128717

RESUMEN

In humans, a single enzyme 2-aminoadipic semialdehyde synthase (AASS) catalyses the initial two critical reactions in the lysine degradation pathway. This enzyme evolved to be a bifunctional enzyme with both lysine-2-oxoglutarate reductase (LOR) and saccharopine dehydrogenase domains (SDH). Moreover, AASS is a unique drug target for inborn errors of metabolism such as glutaric aciduria type 1 that arise from deficiencies downstream in the lysine degradation pathway. While work has been done to elucidate the SDH domain structurally and to develop inhibitors, neither has been done for the LOR domain. Here, we purify and characterize LOR and show that it is activated by alkylation of cysteine 414 by N-ethylmaleimide. We also provide evidence that AASS is rate-limiting upon high lysine exposure of mice. Finally, we present the crystal structure of the human LOR domain. Our combined work should enable future efforts to identify inhibitors of this novel drug target.


Asunto(s)
Lisina , Sacaropina Deshidrogenasas , Errores Innatos del Metabolismo de los Aminoácidos , Animales , Encefalopatías Metabólicas , Cisteína , Etilmaleimida , Glutaril-CoA Deshidrogenasa/deficiencia , Humanos , Lisina/metabolismo , Ratones , Sacaropina Deshidrogenasas/química , Sacaropina Deshidrogenasas/metabolismo
2.
Arch Biochem Biophys ; 584: 98-106, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26342457

RESUMEN

Saccharopine reductase catalyzes the reductive amination of l-α-aminoadipate-δ-semialdehyde with l-glutamate to give saccharopine. Two mechanisms have been proposed for the reductase, one that makes use of enzyme side chains as acid-base catalytic groups, and a second, in which the reaction is catalyzed by enzyme-bound reactants. Site-directed mutagenesis was used to change acid-base candidates in the active site of the reductase to eliminate their ionizable side chain. Thus, the D126A, C154S and Y99F and several double mutant enzymes were prepared. Kinetic parameters in the direction of glutamate formation exhibited modest decreases, inconsistent with the loss of an acid-base catalyst. The pH-rate profiles obtained with all mutant enzymes decrease at low and high pH, suggesting acid and base catalytic groups are still present in all enzymes. Solvent kinetic deuterium isotope effects are all larger than those observed for wild type enzyme, and approximately equal to one another, suggesting the slow step is the same as that of wild type enzyme, a conformational change to open the site and release products (in the direction of saccharopine formation). Overall, the acid-base chemistry is likely catalyzed by bound reactants, with the exception of deprotonation of the α-amine of glutamate, which likely requires an enzyme residue.


Asunto(s)
Mutagénesis Sitio-Dirigida , Mutación Missense , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Sacaropina Deshidrogenasas/química , Sustitución de Aminoácidos , Medición de Intercambio de Deuterio , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Sacaropina Deshidrogenasas/genética , Sacaropina Deshidrogenasas/metabolismo
3.
Adv Protein Chem Struct Biol ; 100: 153-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26415844

RESUMEN

In this chapter, we discuss the influence of an anisotropic protein environment on the reaction mechanisms of saccharopine reductase and uroporphyrinogen decarboxylase, respectively, via the use of a quantum mechanical and molecular mechanical (QM/MM) approach. In addition, we discuss the importance of selecting a suitable DFT functional to be used in a QM/MM study of a key intermediate in the mechanism of 8R-lipoxygenase, a nonheme iron enzyme. In the case of saccharopine reductase, while the enzyme utilizes a substrate-assisted catalytic pathway, it was found that only through treating the polarizing effect of the active site, via the use of an electronic embedding formalism, was agreement with experimental kinetic data obtained. Similarly, in the case of uroporphyrinogen decarboxylase, the effect of the protein environment on the catalytic mechanism was found to be such that the calculated rate-limiting barrier is in good agreement with related experimentally determined values for the first decarboxylation of the substrate. For 8R-lipoxygenase, it was found that the geometries and energies of the multicentered open-shell intermediate complexes formed during the mechanism are quite sensitive to the choice of the density functional theory method. Thus, while density functional theory has become the method of choice in QM/MM studies, care must be taken in the selection of a particular high-level method.


Asunto(s)
Araquidonato Lipooxigenasas/química , Simulación de Dinámica Molecular , Sacaropina Deshidrogenasas/química , Uroporfirinógeno Descarboxilasa/química , Animales , Anisotropía , Antozoos/química , Antozoos/enzimología , Dominio Catalítico , Humanos , Cinética , Teoría Cuántica , Especificidad por Sustrato , Termodinámica
4.
J Biol Chem ; 290(32): 19756-69, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26088140

RESUMEN

Combinatorial gene regulation through feed-forward loops (FFLs) can bestow specificity and temporal control to client gene expression; however, characteristics of binding sites that mediate these effects are not established. We previously showed that the glucocorticoid receptor (GR) and KLF15 form coherent FFLs that cooperatively induce targets such as the amino acid-metabolizing enzymes AASS and PRODH and incoherent FFLs exemplified by repression of MT2A by KLF15. Here, we demonstrate that GR and KLF15 physically interact and identify low affinity GR binding sites within glucocorticoid response elements (GREs) for PRODH and AASS that contribute to combinatorial regulation with KLF15. We used deep sequencing and electrophoretic mobility shift assays to derive in vitro GR binding affinities across sequence space. We applied these data to show that AASS GRE activity correlated (r(2) = 0.73) with predicted GR binding affinities across a 50-fold affinity range in transfection assays; however, the slope of the linear relationship more than doubled when KLF15 was expressed. Whereas activity of the MT2A GRE was even more strongly (r(2) = 0.89) correlated with GR binding site affinity, the slope of the linear relationship was sharply reduced by KLF15, consistent with incoherent FFL logic. Thus, GRE architecture and co-regulator expression together determine the functional parameters that relate GR binding site affinity to hormone-induced transcriptional responses. Utilization of specific affinity response functions and GR binding sites by FFLs may contribute to the diversity of gene expression patterns within GR-regulated transcriptomes.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Nucleares/metabolismo , Prolina Oxidasa/metabolismo , Receptores de Glucocorticoides/metabolismo , Elementos de Respuesta , Sacaropina Deshidrogenasas/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Sitios de Unión , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Línea Celular , Dexametasona/farmacología , Ensayo de Cambio de Movilidad Electroforética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Factores de Transcripción de Tipo Kruppel/química , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Prolina Oxidasa/química , Prolina Oxidasa/genética , Regiones Promotoras Genéticas , Unión Proteica , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Sacaropina Deshidrogenasas/química , Sacaropina Deshidrogenasas/genética , Transducción de Señal
5.
J Mol Graph Model ; 44: 17-25, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23732302

RESUMEN

Saccharopine dehydrogenase (SDH) is the last enzyme in the AAA pathway of l-lysine biosynthesis. On the basis of crystal structures of SDH, the whole catalytic cycle of SDH has been studied by using density functional theory (DFT) method. Calculation results indicate that hydride transfer is the rate-limiting step with an energy barrier of 25.02kcal/mol, and the overall catalytic reaction is calculated to be endothermic by 9.63kcal/mol. Residue Lys77 is proved to be functional only in the process of saccharopine deprotonation until the formation of product l-lysine, and residue His96 is confirmed to take part in multiple proton transfer processes and can be described as a proton transfer station. From the point of view of energy, the SDH catalytic reaction for the synthesis of l-lysine is unfavorable compared with its reverse reaction for the synthesis of saccharopine. These results are essentially consistent with the experimental observations from pH dependence of kinetic parameters and isotope effects.


Asunto(s)
Modelos Teóricos , Protones , Sacaropina Deshidrogenasas/química , Modelos Moleculares , Conformación Molecular , Piridinas/química , Bases de Schiff/química , Termodinámica
6.
Biochemistry ; 51(4): 857-66, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22243403

RESUMEN

Saccharopine dehydrogenase (SDH) catalyzes the final reaction in the α-aminoadipate pathway, the conversion of l-saccharopine to l-lysine (Lys) and α-ketoglutarate (α-kg) using NAD⁺ as an oxidant. The enzyme utilizes a general acid-base mechanism to conduct its reaction with a base proposed to accept a proton from the secondary amine of saccharopine in the oxidation step and a group proposed to activate water to hydrolyze the resulting imine. Crystal structures of an open apo form and a closed form of the enzyme with saccharopine and NADH bound have been determined at 2.0 and 2.2 Å resolution, respectively. In the ternary complex, a significant movement of domain I relative to domain II that closes the active site cleft between the two domains and brings H96 and K77 into the proximity of the substrate binding site is observed. The hydride transfer distance is 3.6 Å, and the side chains of H96 and K77 are properly positioned to act as acid-base catalysts. Preparation of the K77M and H96Q single-mutant and K77M/H96Q double-mutant enzymes provides data consistent with their role as the general acid-base catalysts in the SDH reaction. The side chain of K77 initially accepts a proton from the ε-amine of the substrate Lys and eventually donates it to the imino nitrogen as it is reduced to a secondary amine in the hydride transfer step, and H96 protonates the carbonyl oxygen as the carbinolamine is formed. The K77M, H976Q, and K77M/H96Q mutant enzymes give 145-, 28-, and 700-fold decreases in V/E(t) and >10³-fold increases in V2/K(Lys)E(t) and V2/K(α-kg)E(t) (the double mutation gives >105-fold decreases in the second-order rate constants). In addition, the K77M mutant enzyme exhibits a primary deuterium kinetic isotope effect of 2.0 and an inverse solvent deuterium isotope effect of 0.77 on V2/K(Lys). A value of 2.0 was also observed for (D)(V2/K(Lys))(D2O) when the primary deuterium kinetic isotope effect was repeated in D2O, consistent with a rate-limiting hydride transfer step. A viscosity effect of 0.8 was observed on V2/K(Lys), indicating the solvent deuterium isotope effect resulted from stabilization of an enzyme form prior to hydride transfer. A small normal solvent isotope effect is observed on V, which decreases slightly when repeated with NADD, consistent with a contribution from product release to rate limitation. In addition, V2/K(Lys)E(t) is pH-independent, which is consistent with the loss of an acid-base catalyst and perturbation of the pK(a) of the second catalytic group to a higher pH, likely a result of a change in the overall charge of the active site. The primary deuterium kinetic isotope effect for H96Q, measured in H2O or D2O, is within error equal to 1. A solvent deuterium isotope effect of 2.4 is observed with NADH or NADD as the dinucleotide substrate. Data suggest rate-limiting imine formation, consistent with the proposed role of H96 in protonating the leaving hydroxyl as the imine is formed. The pH-rate profile for V2/K(Lys)E(t) exhibits the pK(a) for K77, perturbed to a value of ∼9, which must be unprotonated to accept a proton from the ε-amine of the substrate Lys so that it can act as a nucleophile. Overall, data are consistent with a role for K77 acting as the base that accepts a proton from the ε-amine of the substrate lysine prior to nucleophilic attack on the α-oxo group of α-ketoglutarate, and finally donating a proton to the imine nitrogen as it is reduced to give saccharopine. In addition, data indicate a role for H96 acting as a general acid-base catalyst in the formation of the imine between the ε-amine of lysine and the α-oxo group of α-ketoglutarate.


Asunto(s)
Histidina/química , Lisina/química , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Sacaropina Deshidrogenasas/química , Sacaropina Deshidrogenasas/metabolismo , Sustitución de Aminoácidos , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Deuterio , Estabilidad de Enzimas , Concentración de Iones de Hidrógeno , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , NAD/química , NAD/metabolismo , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Sacaropina Deshidrogenasas/genética , Viscosidad
7.
Molecules ; 16(10): 8569-89, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21993247

RESUMEN

Saccharopine reductase from Magnaporthe grisea, an NADPH-containing enzyme in the α-aminoadipate pathway, catalyses the formation of saccharopine, a precursor to L-lysine, from the substrates glutamate and α-aminoadipate-δ-semialdehyde. Its catalytic mechanism has been investigated using quantum mechanics/molecular mechanics (QM/MM) ONIOM-based approaches. In particular, the overall catalytic pathway has been elucidated and the effects of electron correlation and the anisotropic polar protein environment have been examined via the use of the ONIOM(HF/6-31G(d):AMBER94) and ONIOM(MP2/6-31G(d)//HF/6-31G(d):AMBER94) methods within the mechanical embedding formulism and ONIOM(MP2/6-31G(d)//HF/6-31G(d):AMBER94) and ONIOM(MP2/6-311G(d,p)//HF/6-31G(d):AMBER94) within the electronic embedding formulism. The results of the present study suggest that saccharopine reductase utilises a substrate-assisted catalytic pathway in which acid/base groups within the cosubstrates themselves facilitate the mechanistically required proton transfers. Thus, the enzyme appears to act most likely by binding the three required reactant molecules glutamate, α-aminoadipate-δ-semialdehyde and NADPH in a manner and polar environment conducive to reaction.


Asunto(s)
Lisina/análogos & derivados , Sacaropina Deshidrogenasas/metabolismo , Ácido 2-Aminoadípico/metabolismo , Catálisis , Ácido Glutámico/metabolismo , Lisina/metabolismo , Magnaporthe/enzimología , Magnaporthe/metabolismo , NADP/metabolismo , Sacaropina Deshidrogenasas/química
8.
Arch Biochem Biophys ; 513(2): 71-80, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21798231

RESUMEN

Saccharopine dehydrogenase catalyzes the NAD-dependent conversion of saccharopine to generate L-lysine and α-ketoglutarate. A disulfide bond between cysteine 205 and cysteine 249, in the vicinity of the dinucleotide-binding site, is observed in structures of the apoenzyme, while a dithiol is observed in a structure with AMP bound, suggesting preferential binding of the dinucleotide to reduced enzyme. Mutation of C205 to S gave increased values of V/E(t) and V/KE(t) at pH 7 compared to wild type. Primary deuterium and solvent deuterium kinetic isotope effects suggest the catalytic pathway, which includes the hydride transfer and hydrolysis steps, contributes more to rate limitation in C205S, but the rates of the two steps relative to one another remain the same. There is a large increase in the rate constants V1/E(t) and V1/K(NAD)Et at pH values below 7 compared to WT. Data indicate the low pH increase in activity results from a decreased sensitivity of the C205S mutant enzyme to the protonation state of an enzyme group with a pK(a) of about 7, likely responsible for a pH-dependent conformational change. Reduction of WT and C205S mutant enzymes with TCEP gives equal activities at pH 6, consistent with the increased activity observed for the C205S mutant enzyme.


Asunto(s)
Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Sacaropina Deshidrogenasas/química , Sacaropina Deshidrogenasas/metabolismo , Sustitución de Aminoácidos , Apoenzimas/química , Apoenzimas/genética , Apoenzimas/metabolismo , Secuencia de Bases , Dominio Catalítico , Cisteína/química , ADN de Hongos/genética , Medición de Intercambio de Deuterio , Concentración de Iones de Hidrógeno , Cinética , Lisina/análogos & derivados , Lisina/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , NAD/metabolismo , Oxidación-Reducción , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Sacaropina Deshidrogenasas/genética , Compuestos de Sulfhidrilo/química
9.
FEMS Microbiol Lett ; 312(1): 77-83, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20846141

RESUMEN

By means of an in silico analysis, we demonstrated that a previously described chimeric gene (Spe-Sdh) encoding spermidine synthase, a key enzyme involved in the synthesis of polyamines, and saccharopine dehydrogenase, an enzyme involved in lysine synthesis in fungi, were present exclusively in members of all Basidiomycota subphyla, but not in any other group of living organisms. We used this feature to design degenerated primers to amplify a specific fragment of the Spe-Sdh gene by PCR, as a tool to unequivocally identify Basidiomycota isolates. The specificity of this procedure was tested using different fungal species. As expected, positive results were obtained only with Basidiomycota species, whereas no amplification was achieved with species belonging to other fungal phyla.


Asunto(s)
Basidiomycota/enzimología , Proteínas Fúngicas/genética , Sondas Moleculares/genética , Sacaropina Deshidrogenasas/genética , Espermidina Sintasa/genética , Secuencia de Aminoácidos , Secuencia de Bases , Basidiomycota/química , Basidiomycota/clasificación , Basidiomycota/genética , Cartilla de ADN/genética , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena de la Polimerasa , Sacaropina Deshidrogenasas/química , Sacaropina Deshidrogenasas/metabolismo , Alineación de Secuencia , Especificidad de la Especie , Espermidina Sintasa/química , Espermidina Sintasa/metabolismo
10.
J Biol Chem ; 285(27): 20756-68, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20427272

RESUMEN

Saccharopine dehydrogenase catalyzes the NAD-dependent oxidative deamination of saccharopine to give l-lysine and alpha-ketoglutarate. There are a number of conserved hydrophilic, ionizable residues in the active site, all of which must be important to the overall reaction. In an attempt to determine the contribution to binding and rate enhancement of each of the residues in the active site, mutations at each residue are being made, and double mutants are being made to estimate the interrelationship between residues. Here, we report the effects of mutations of active site glutamate residues, Glu(78) and Glu(122), on reactant binding and catalysis. Site-directed mutagenesis was used to generate E78Q, E122Q, E78Q/E122Q, E78A, E122A, and E78A/E122A mutant enzymes. Mutation of these residues increases the positive charge of the active site and is expected to affect the pK(a) values of the catalytic groups. Each mutant enzyme was completely characterized with respect to its kinetic and chemical mechanism. The kinetic mechanism remains the same as that of wild type enzymes for all of the mutant enzymes, with the exception of E78A, which exhibits binding of alpha-ketoglutarate to E and E.NADH. Large changes in V/K(Lys), but not V, suggest that Glu(78) and Glu(122) contribute binding energy for lysine. Shifts of more than a pH unit to higher and lower pH of the pK(a) values observed in the V/K(Lys) pH-rate profile of the mutant enzymes suggests that the presence of Glu(78) and Glu(122) modulates the basicity of the catalytic groups.


Asunto(s)
Ácido Glutámico/análisis , Sacaropina Deshidrogenasas/química , Equilibrio Ácido-Base , Alanina/genética , Sustitución de Aminoácidos , Unión Competitiva , Catálisis , Dominio Catalítico , Glutamina/genética , Cinética , Lisina/biosíntesis , Modelos Moleculares , Mutagénesis Sitio-Dirigida , NAD/química , NAD/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Sacaropina Deshidrogenasas/genética , Sacaropina Deshidrogenasas/aislamiento & purificación , Sacaropina Deshidrogenasas/metabolismo
11.
Biochemistry ; 48(25): 5899-907, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19449898

RESUMEN

Saccharopine reductase (SR) [saccharopine dehydrogenase (l-glutamate forming), EC 1.5.1.10] catalyzes the condensation of l-alpha-aminoadipate-delta-semialdehyde (AASA) with l-glutamate to give an imine, which is reduced by NADPH to give saccharopine. An acid-base chemical mechanism has been proposed for SR on the basis of pH-rate profiles and solvent deuterium kinetic isotope effects. A finite solvent isotope effect is observed indicating that proton(s) are in flight in the rate-limiting step(s) and likely the same step is limiting under both limiting and saturating substrate concentrations. A concave upward proton inventory suggests that more than one proton is transferred in a single transition state, likely a conformation change required to open the site and release products. Two groups are involved in the acid-base chemistry of the reaction. One of these groups catalyzes the steps involved in forming the imine between the alpha-amine of glutamate and the aldehyde of AASA. The group, which has a pK(a) of about 8, is observed in the pH-rate profiles for V(1) and V(1)/K(Glu) and must be protonated for optimal activity. It is also observed in the V(2) and V(2)/K(Sacc) pH-rate profiles and is required unprotonated. The second group, which has a pK(a) of 5.6, accepts a proton from the alpha-amine of glutamate so that it can act as a nucleophile in forming a carbinolamine upon attack of the carbonyl of AASA.


Asunto(s)
Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Sacaropina Deshidrogenasas/química , Medición de Intercambio de Deuterio , Concentración de Iones de Hidrógeno , Cinética , Lisina/análogos & derivados , Lisina/química , Lisina/metabolismo , NADP/química , NADP/metabolismo , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Proteínas de Saccharomyces cerevisiae/metabolismo , Sacaropina Deshidrogenasas/antagonistas & inhibidores , Sacaropina Deshidrogenasas/metabolismo , Solventes
12.
Biochemistry ; 46(44): 12512-21, 2007 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-17939687

RESUMEN

Three structures of saccharopine dehydrogenase (l-lysine-forming) (SDH) have been determined in the presence of sulfate, adenosine monophosphate (AMP), and oxalylglycine (OxGly). In the sulfate-bound structure, a sulfate ion binds in a cleft between the two domains of SDH, occupies one of the substrate carboxylate binding sites, and results in partial closure of the active site of the enzyme due to a domain rotation of almost 12 degrees in comparison to the apoenzyme structure. In the second structure, AMP binds to the active site in an area where the NAD+ cofactor is expected to bind. All of the AMP moieties (adenine ring, ribose, and phosphate) interact with specific residues of the enzyme. In the OxGly-bound structure, carboxylates of OxGly interact with arginine residues representative of the manner in which substrate (alpha-ketoglutarate and saccharopine) may bind. The alpha-keto group of OxGly interacts with Lys77 and His96, which are candidates for acid-base catalysis. Analysis of ligand-enzyme interactions, comparative structural analysis, corroboration with kinetic data, and discussion of a ternary complex model are presented in this study.


Asunto(s)
Ligandos , Lisina/análogos & derivados , Saccharomyces cerevisiae/enzimología , Sacaropina Deshidrogenasas/química , Sitios de Unión , Cristalografía por Rayos X , Lisina/química , Lisina/metabolismo , Modelos Biológicos , Modelos Moleculares , Unión Proteica , Sacaropina Deshidrogenasas/aislamiento & purificación , Sacaropina Deshidrogenasas/metabolismo
13.
J Mol Biol ; 373(3): 745-54, 2007 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-17854830

RESUMEN

The 1.64 A structure of the apoenzyme form of saccharopine dehydrogenase (SDH) from Saccharomyces cerevisiae shows the enzyme to be composed of two domains with similar dinucleotide binding folds with a deep cleft at the interface. The structure reveals homology to alanine dehydrogenase, despite low primary sequence similarity. A model of the ternary complex of SDH, NAD, and saccharopine identifies residues Lys77 and Glu122 as potentially important for substrate binding and/or catalysis, consistent with a proton shuttle mechanism. Furthermore, the model suggests that a conformational change is required for catalysis and that residues Lys99 and Asp281 may be instrumental in mediating this change. Analysis of the crystal structure in the context of other homologous enzymes from pathogenic fungi and human sources sheds light into the suitability of SDH as a target for antimicrobial drug development.


Asunto(s)
Lisina/análogos & derivados , NAD/metabolismo , Saccharomyces cerevisiae/enzimología , Sacaropina Deshidrogenasas/química , Alanina-Deshidrogenasa/química , Alanina-Deshidrogenasa/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Lisina/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Saccharomyces cerevisiae/crecimiento & desarrollo , Sacaropina Deshidrogenasas/aislamiento & purificación , Sacaropina Deshidrogenasas/metabolismo , Homología de Secuencia de Aminoácido
14.
Biochemistry ; 46(25): 7625-36, 2007 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-17542618

RESUMEN

A survey of NADH, alpha-Kg, and lysine analogues has been undertaken in an attempt to define the substrate specificity of saccharopine dehydrogenase and to identify functional groups on all substrates and dinucleotides important for substrate binding. A number of NAD analogues, including NADP, 3-acetylpyridine adenine dinucleotide (3-APAD), 3-pyridinealdehyde adenine dinucleotide (3-PAAD), and thionicotinamide adenine dinucleotide (thio-NAD), can serve as a substrate in the oxidative deamination reaction, as can a number of alpha-keto analogues, including glyoxylate, pyruvate, alpha-ketobutyrate, alpha-ketovalerate, alpha-ketomalonate, and alpha-ketoadipate. Inhibition studies using nucleotide analogues suggest that the majority of the binding energy of the dinucleotides comes from the AMP portion and that distinctly different conformations are generated upon binding of the oxidized and reduced dinucleotides. Addition of the 2'-phosphate as in NADPH causes poor binding of subsequent substrates but has little effect on coenzyme binding and catalysis. In addition, the 10-fold decrease in affinity of 3-APAD in comparison to NAD suggests that the nicotinamide ring binding pocket is hydrophilic. Extensive inhibition studies using aliphatic and aromatic keto acid analogues have been carried out to gain insight into the keto acid binding pocket. Data suggest that a side chain with three carbons (from the alpha-keto group up to and including the side chain carboxylate) is optimal. In addition, the distance between the C1-C2 unit and the C5 carboxylate of the alpha-keto acid is also important for binding; the alpha-oxo group contributes a factor of 10 to affinity. The keto acid binding pocket is relatively large and flexible and can accommodate the bulky aromatic ring of a pyridine dicarboxylic acid and a negative charge at the C3 but not the C4 position. However, the amino acid binding site is hydrophobic, and the optimal length of the hydrophobic portion of the amino acid carbon side chain is three or four carbons. In addition, the amino acid binding pocket can accommodate a branch at the gamma-carbon, but not at the beta-carbon.


Asunto(s)
Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Sacaropina Deshidrogenasas/química , Sitios de Unión , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Estructura Molecular , NAD/análogos & derivados , NAD/química , NAD/metabolismo , Oxidación-Reducción , Unión Proteica , Conformación Proteica , Estructura Secundaria de Proteína , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Sacaropina Deshidrogenasas/metabolismo , Especificidad por Sustrato
15.
Biochemistry ; 46(3): 871-82, 2007 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-17223709

RESUMEN

Saccharopine dehydrogenase [N6-(glutaryl-2)-L-lysine:NAD oxidoreductase (L-lysine forming)] catalyzes the final step in the alpha-aminoadipate pathway for lysine biosynthesis. It catalyzes the reversible pyridine nucleotide-dependent oxidative deamination of saccharopine to generate alpha-Kg and lysine using NAD+ as an oxidizing agent. The proton shuttle chemical mechanism is proposed on the basis of the pH dependence of kinetic parameters, dissociation constants for competitive inhibitors, and isotope effects. In the direction of lysine formation, once NAD+ and saccharopine bind, a group with a pKa of 6.2 accepts a proton from the secondary amine of saccharopine as it is oxidized. This protonated general base then does not participate in the reaction again until lysine is formed at the completion of the reaction. A general base with a pKa of 7.2 accepts a proton from H2O as it attacks the Schiff base carbon of saccharopine to form the carbinolamine intermediate. The same residue then serves as a general acid and donates a proton to the carbinolamine nitrogen to give the protonated carbinolamine. Collapse of the carbinolamine is then facilitated by the same group accepting a proton from the carbinolamine hydroxyl to generate alpha-Kg and lysine. The amine nitrogen is then protonated by the group that originally accepted a proton from the secondary amine of saccharopine, and products are released. In the reverse reaction direction, finite primary deuterium kinetic isotope effects were observed for all parameters with the exception of V2/K(NADH), consistent with a steady-state random mechanism and indicative of a contribution from hydride transfer to rate limitation. The pH dependence, as determined from the primary isotope effect on DV2 and D(V2/K(Lys)), suggests that a step other than hydride transfer becomes rate-limiting as the pH is increased. This step is likely protonation/deprotonation of the carbinolamine nitrogen formed as an intermediate in imine hydrolysis. The observed solvent isotope effect indicates that proton transfer also contributes to rate limitation. A concerted proton and hydride transfer is suggested by multiple substrate/solvent isotope effects, as well as a proton transfer in another step, likely hydrolysis of the carbinolamine. In agreement, dome-shaped proton inventories are observed for V2 and V2/K(Lys), suggesting that proton transfer exists in at least two sequential transition states.


Asunto(s)
Deuterio/metabolismo , Protones , Saccharomyces cerevisiae/enzimología , Sacaropina Deshidrogenasas/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Redes y Vías Metabólicas , Modelos Químicos , Resonancia Magnética Nuclear Biomolecular , Sacaropina Deshidrogenasas/química
16.
Biochemistry ; 45(39): 12156-66, 2006 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-17002315

RESUMEN

Kinetic data have been measured for the histidine-tagged saccharopine dehydrogenase from Saccharomyces cerevisiae, suggesting the ordered addition of nicotinamide adenine dinucleotide (NAD) followed by saccharopine in the physiologic reaction direction. In the opposite direction, the reduced nicotinamide adenine dinucleotide (NADH) adds to the enzyme first, while there is no preference for the order of binding of alpha-ketoglutarate (alpha-Kg) and lysine. In the direction of saccharopine formation, data also suggest that, at high concentrations, lysine inhibits the reaction by binding to free enzyme. In addition, uncompetitive substrate inhibition by alpha-Kg and double inhibition by NAD and alpha-Kg suggest the existence of an abortive E:NAD:alpha-Kg complex. Product inhibition by saccharopine is uncompetitive versus NADH, suggesting a practical irreversibility of the reaction at pH 7.0 in agreement with the overall K(eq). Saccharopine is noncompetitive versus lysine or alpha-Kg, suggesting the existence of both E:NADH:saccharopine and E:NAD:saccharopine complexes. NAD is competitive versus NADH, and noncompetitive versus lysine and alpha-Kg, indicating the combination of the dinucleotides with free enzyme. Dead-end inhibition studies are also consistent with the random addition of alpha-Kg and lysine. Leucine and oxalylglycine serve as lysine and alpha-Kg dead-end analogues, respectively, and are uncompetitive against NADH and noncompetitive against alpha-Kg and lysine, respectively. Oxaloacetate (OAA), pyruvate, and glutarate behave as dead-end analogues of lysine, which suggests that the lysine-binding site has a higher affinity for keto acid analogues than does the alpha-Kg site or that dicarboxylic acids have more than one binding mode on the enzyme. In addition, OAA and glutarate also bind to free enzyme as does lysine at high concentrations. Glutarate gives S-parabolic noncompetitive inhibition versus NADH, indicating the formation of a E:(glutarate)2 complex as a result of occupying both the lysine- and alpha-Kg-binding sites. Pyruvate, a slow alternative keto acid substrate, exhibits competitive inhibition versus both lysine and alpha-Kg, suggesting the combination to the E:NADH:alpha-Kg and E:NADH:lysine enzyme forms. The equilibrium constant for the reaction has been measured at pH 7.0 as 3.9 x 10(-7) M by monitoring the change in NADH upon the addition of the enzyme. The Haldane relationship is in very good agreement with the directly measured value.


Asunto(s)
Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Sacaropina Deshidrogenasas/química , Sitios de Unión , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Lisina/análogos & derivados , Lisina/química , Lisina/metabolismo , NAD/química , NAD/metabolismo , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Sacaropina Deshidrogenasas/metabolismo
17.
Cell Biochem Biophys ; 46(1): 17-26, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16943620

RESUMEN

The three-dimensional structure of the saccharopine reductase enzyme from the budding yeast Saccharomyces cerevisiae was determined to 1.7-A resolution in the apo form by using molecular replacement. The enzyme monomer consists of three domains: domain I is a variant of the Rossmann fold, domain II folds into a alpha/beta structure containing a mixed seven-stranded beta-sheet as the central core, and domain III has an all-helical fold. Comparative fold alignment with the enzyme from Magnaporthe grisea suggests that domain I binds to NADPH, and domain II binds to saccharopine and is involved in dimer formation. Domain III is involved in closing the active site of the enzyme once substrates are bound. Structural comparison of the saccharopine reductase enzymes from S. cerevisiae and M. grisea indicates that domain II has the highest number of conserved residues, suggesting that it plays an important role in substrate binding and in spatially orienting domains I and III.


Asunto(s)
Modelos Moleculares , Proteínas de Saccharomyces cerevisiae/química , Sacaropina Deshidrogenasas/química , Histidina/química , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación , Sacaropina Deshidrogenasas/aislamiento & purificación
18.
Mol Genet Genomics ; 275(1): 74-80, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16292576

RESUMEN

Candida albicans LYS1-encoded saccharopine dehydrogenase (CaLys1p, SDH) catalyzes the final biosynthetic step (saccharopine to lysine + alpha-ketoglutarate) of the novel alpha-aminoadipate pathway for lysine synthesis in fungi. The reverse reaction catalyzed by lysine-alpha-ketoglutarate reductase (LKR) is used exclusively in animals and plants for the catabolism of excess lysine. The 1,146 bp C. albicans LYS1 ORF encodes a 382 amino acid SDH. In the present investigation, we have used E. coli-expressed recombinant C. albicans Lys1p for the determination of both forward and reverse SDH activities in vitro, compared the sequence identity of C. albicans Lys1p with other known SDHs and LKRs, performed extensive site-directed mutational analyses of conserved amino acid residues and analyzed the phylogenetic relationship of C. albicans Lys1p to other known SDHs and LKRs. We have identified 14 of the 68 amino acid substitutions as essential for C. albicans Lys1p SDH activity, including two highly conserved functional motifs, H93XXF96XH98 and G138XXXG142XXG145. These results provided new insight into the functional and phylogenetic characteristics of the distinct biosynthetic SDH in fungi and catabolic LKR in higher eukaryotes.


Asunto(s)
Sustitución de Aminoácidos , Candida albicans/enzimología , Proteínas Fúngicas/química , Filogenia , Mutación Puntual , Sacaropina Deshidrogenasas/química , Secuencias de Aminoácidos/genética , Candida albicans/genética , Proteínas Fúngicas/metabolismo , Ácidos Cetoglutáricos/metabolismo , Lisina/biosíntesis , Lisina/química , Mutagénesis Sitio-Dirigida , Sacaropina Deshidrogenasas/genética
19.
IUBMB Life ; 55(8): 459-66, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-14609201

RESUMEN

Although bifunctional enzymes containing two different active centers located within separate domains are quite common in living systems, the significance of this bifunctionality is not always clear, and the molecular mechanisms of site-site interactions in such complex systems have come under the scrutiny of science only in recent years. This review summarizes recent data on the mechanisms of communication between active centers in bifunctional enzymes. Three types of enzymes are considered: (1) those catalyzing consecutive reactions of a metabolic pathway and exhibiting substrate channeling (glutamate synthase and imidazole glycerol phosphate synthase), (2) those catalyzing consecutive reactions without substrate channeling (lysine-ketoglutarate reductase/saccharopine dehydrogenase), and (3) those catalyzing opposed reactions (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase). The functional role of interdomain communications is briefly discussed.


Asunto(s)
Fenómenos Fisiológicos Bacterianos , Enzimas/química , Enzimas/fisiología , Aminohidrolasas/química , Sitios de Unión , Catálisis , Glutamato Sintasa/química , Modelos Biológicos , Fosfofructoquinasa-2/metabolismo , Fosforilación , Estructura Terciaria de Proteína , Sacaropina Deshidrogenasas/química , Sacaropina Deshidrogenasas/fisiología , Transducción de Señal
20.
Amino Acids ; 24(1-2): 179-86, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12624751

RESUMEN

Lysine is catabolyzed by the bifunctional enzyme lysine 2-oxoglutarate reductase-saccharopine dehydrogenase (LOR-SDH) in both animals and plants. LOR condenses lysine and 2-oxoglutarate into saccharopine, using NADPH as cofactor and SDH converts saccharopine into alpha-aminoadipate delta-semialdehyde and glutamic acid, using NAD as cofactor. The distribution pattern of LOR and SDH among different tissues of Phaseolus vulgaris was determined. The hypocotyl contained the highest specific activity, whereas in seeds the activities of LOR and SDH were below the limit of detection. Precipitation of hypocotyl proteins with increasing concentrations of PEG 8000 revealed one broad peak of SDH activity, indicating that two isoforms may be present, a bifunctional LOR-SDH and possibly a monofunctional SDH. During the purification of the hypocotyl enzyme, the LOR activity proved to be very unstable, following ion-exchange chromatography. Depending on the purification procedure, the protein eluted as a monomer of 91-94 kDa containing only SDH activity, or as a dimer of 190 kDa with both, LOR and SDH activities, eluting together.


Asunto(s)
Phaseolus/enzimología , Sacaropina Deshidrogenasas/aislamiento & purificación , Peso Molecular , Sacaropina Deshidrogenasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...