Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 10(1): 4860, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32184419

RESUMO

The opportunistic pathogen Malassezia pachydermatis causes bloodstream infections in preterm infants or individuals with immunodeficiency disorders and has been associated with a broad spectrum of diseases in animals such as seborrheic dermatitis, external otitis and fungemia. The current approaches to treat these infections are failing as a consequence of their adverse effects, changes in susceptibility and antifungal resistance. Thus, the identification of novel therapeutic targets against M. pachydermatis infections are highly relevant. Here, Gene Essentiality Analysis and Flux Variability Analysis was applied to a previously reported M. pachydermatis metabolic network to identify enzymes that, when absent, negatively affect biomass production. Three novel therapeutic targets (i.e., homoserine dehydrogenase (MpHSD), homocitrate synthase (MpHCS) and saccharopine dehydrogenase (MpSDH)) were identified that are absent in humans. Notably, L-lysine was shown to be an inhibitor of the enzymatic activity of MpHCS and MpSDH at concentrations of 1 mM and 75 mM, respectively, while L-threonine (1 mM) inhibited MpHSD. Interestingly, L- lysine was also shown to inhibit M. pachydermatis growth during in vitro assays with reference strains and canine isolates, while it had a negligible cytotoxic activity on HEKa cells. Together, our findings form the bases for the development of novel treatments against M. pachydermatis infections.


Assuntos
Dermatomicoses/microbiologia , Proteínas Fúngicas/antagonistas & inibidores , Fungemia/microbiologia , Lisina/farmacologia , Malassezia/crescimento & desenvolvimento , Treonina/farmacologia , Animais , Linhagem Celular , Dermatomicoses/tratamento farmacológico , Dermatomicoses/veterinária , Relação Dose-Resposta a Droga , Fungemia/tratamento farmacológico , Genes Essenciais , Homosserina Desidrogenase/antagonistas & inibidores , Humanos , Malassezia/efeitos dos fármacos , Oxo-Ácido-Liases/antagonistas & inibidores , Sacaropina Desidrogenases/antagonistas & inibidores
2.
J Mol Model ; 25(11): 325, 2019 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-31654136

RESUMO

Paracoccidioidomycosis (PCM) is a systemic mycosis caused by fungi of the genus Paracoccidioides spp., which mainly affects workers in rural regions of Latin America. Although the antifungal agents currently available for the treatment of PCM are effective in controlling the disease, many months are needed for healing, making the side effects and drug interactions relevant. In addition, conventional treatments are not able to control the sequelae left by PCM, even after the cure, justifying the search for new therapeutic options against PCM. In this context, the enzyme homoserine dehydrogenase of P. brasiliensis (PbHSD) was used to screen a library of natural products from the Zinc database using three different docking programs, i.e. Autodock, Molegro, and CLC Drugdiscovery Workbench. Three molecules (Zinc codes 2123137, 15967722, and 20611644) were better ranked than the homoserine substrate (HSE) and were used for in vitro trials of the minimum inhibitory concentration (MIC) and minimal fungicidal concentration (MCF). All three molecules presented a fungicidal profile with MICs/MCFs of 8, 32, and 128 µg mL-1, respectively. The two most promising molecules presented satisfactory results with wide therapeutic ranges in the cytotoxicity assays. Molecular dynamics simulations of PbHSD indicated that the ligands remained bound to the protein by a common mechanism throughout the simulation. The molecule with the lowest MIC value presented the highest number of contacts with the protein. The results presented in this work suggest that the molecule Zinc2123137 may be considered as a hit in the development of new therapeutic options for PCM.


Assuntos
Antifúngicos/farmacologia , Homosserina Desidrogenase/antagonistas & inibidores , Paracoccidioides/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Células HeLa , Humanos , Ligantes , Testes de Sensibilidade Microbiana/métodos , Simulação de Dinâmica Molecular , Células Vero
3.
J Pept Sci ; 24(1)2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29322651

RESUMO

The antifungal activity of 5-hydroxy-4-oxo-l-norvaline (HONV), exhibited under conditions mimicking human serum, may be improved upon incorporation of this amino acid into a dipeptide structure. Several HONV-containing dipeptides inhibited growth of human pathogenic yeasts of the Candida genus in the RPMI-1640 medium, with minimal inhibitory concentration values in the 32 to 64 µg mL-1 range. This activity was not affected by multidrug resistance that is caused by overexpression of genes encoding drug efflux proteins. The mechanism of antifungal action of HONV dipeptides involved uptake by the oligopeptide transport system, subsequent intracellular cleavage by cytosolic peptidases, and inhibition of homoserine dehydrogenase by the released HONV. The relative transport rates determined the anticandidal activity of HONV dipeptides.


Assuntos
Antifúngicos/farmacologia , Candida albicans/efeitos dos fármacos , Candida albicans/enzimologia , Dipeptídeos/farmacologia , Inibidores Enzimáticos/farmacologia , Homosserina Desidrogenase/antagonistas & inibidores , Valina/análogos & derivados , Valina/farmacologia , Antifúngicos/síntese química , Antifúngicos/química , Dipeptídeos/síntese química , Dipeptídeos/química , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Homosserina Desidrogenase/metabolismo , Testes de Sensibilidade Microbiana , Conformação Molecular , Relação Estrutura-Atividade , Valina/síntese química , Valina/química
4.
ACS Synth Biol ; 4(2): 126-31, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24344690

RESUMO

Allosteric proteins, which can sense different signals, are interesting biological parts for synthetic biology. In particular, the design of an artificial allosteric enzyme to sense an unnatural signal is both challenging and highly desired, for example, for a precise and dynamical control of fluxes of growth-essential but byproduct pathways in metabolic engineering of industrial microorganisms. In this work, we used homoserine dehydrogenase (HSDH) of Corynebacterium glutamicum, which is naturally allosterically regulated by threonine and isoleucine, as an example to demonstrate the feasibility of reengineering an allosteric enzyme to respond to an unnatural inhibitor L-lysine. For this purpose, the natural threonine binding sites of HSD were first predicted and verified by mutagenesis experiments. The threonine binding sites were then engineered to a lysine binding pocket. The reengineered HSD only responds to lysine inhibition but not to threonine. This is a significant step toward the construction of artificial molecular circuits for dynamic control of growth-essential byproduct formation pathway for lysine biosynthesis.


Assuntos
Inibidores Enzimáticos/química , Homosserina Desidrogenase/antagonistas & inibidores , Lisina/química , Regulação Alostérica , Substituição de Aminoácidos , Sítios de Ligação , Corynebacterium/enzimologia , Desenho de Fármacos , Inibidores Enzimáticos/metabolismo , Homosserina Desidrogenase/genética , Homosserina Desidrogenase/metabolismo , Isoleucina/química , Isoleucina/metabolismo , Cinética , Lisina/metabolismo , Simulação de Dinâmica Molecular , Ligação Proteica , Engenharia de Proteínas , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Treonina/química , Treonina/metabolismo
5.
J Biol Chem ; 285(2): 827-34, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19897476

RESUMO

Aspartate kinase (AK) and homoserine dehydrogenase (HSD) function as key regulatory enzymes at branch points in the aspartate amino acid pathway and are feedback-inhibited by threonine. In plants the biochemical features of AK and bifunctional AK-HSD enzymes have been characterized, but the molecular properties of the monofunctional HSD remain unexamined. To investigate the role of HSD, we have cloned the cDNA and gene encoding the monofunctional HSD (GmHSD) from soybean. Using heterologously expressed and purified GmHSD, initial velocity and product inhibition studies support an ordered bi bi kinetic mechanism in which nicotinamide cofactor binds first and leaves last in the reaction sequence. Threonine inhibition of GmHSD occurs at concentrations (K(i) = 160-240 mM) more than 1000-fold above physiological levels. This is in contrast to the two AK-HSD isoforms in soybean that are sensitive to threonine inhibition (K(i) approximately 150 microM). In addition, GmHSD is not inhibited by other aspartate-derived amino acids. The ratio of threonine-resistant to threonine-sensitive HSD activity in soybean tissues varies and likely reflects different demands for amino acid biosynthesis. This is the first cloning and detailed biochemical characterization of a monofunctional feedback-insensitive HSD from any plant. Threonine-resistant HSD offers a useful biotechnology tool for manipulating the aspartate amino acid pathway to increase threonine and methionine production in plants for improved nutritional content.


Assuntos
Glycine max/enzimologia , Glycine max/genética , Homosserina Desidrogenase/química , Homosserina Desidrogenase/genética , Homosserina Desidrogenase/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Sequência de Bases , Clonagem Molecular , Homosserina Desidrogenase/antagonistas & inibidores , Cinética , Dados de Sequência Molecular , Proteínas de Plantas/antagonistas & inibidores , Treonina/química
6.
Bioorg Med Chem ; 12(14): 3825-30, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15210149

RESUMO

A relatively unexploited potential target for antimicrobial agents is the biosynthesis of essential amino acids. Homoserine dehydrogenase, which reduces aspartate semi-aldehyde to homoserine in a NAD(P)H-dependent reaction, is one such target that is required for the biosynthesis of Met, Thr, and Ile from Asp. We report a small molecule screen of yeast homoserine dehydrogenase that has identified a new class of phenolic inhibitors of this class of enzyme. X-ray crystal structural analysis of one of the inhibitors in complex with homoserine dehydrogenase reveals that these molecules bind in the amino acid binding region of the active site and that the phenolic hydroxyl group interacts specifically with the backbone amide of Gly175. These results provide the first nonamino acid inhibitors of this class of enzyme and have the potential to be exploited as leads in antifungal compound design.


Assuntos
Inibidores Enzimáticos/farmacologia , Homosserina Desidrogenase/antagonistas & inibidores , Fenóis/farmacologia , Saccharomyces cerevisiae/enzimologia , Antifúngicos/química , Antifúngicos/farmacologia , Candida/efeitos dos fármacos , Cristalografia por Raios X , Inibidores Enzimáticos/química , Espectroscopia de Ressonância Magnética , Testes de Sensibilidade Microbiana , Fenóis/química , Saccharomyces cerevisiae/efeitos dos fármacos
7.
Chem Biol ; 10(10): 989-95, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14583265

RESUMO

The structure of the antifungal drug 5-hydroxy-4-oxonorvaline (HON) in complex with its target homoserine dehydrogenase (HSD) has been determined by X-ray diffraction to 2.6 A resolution. HON shows potent in vitro and in vivo activity against various fungal pathogens despite its weak (2 mM) affinity for HSD in the steady state. The structure together with structure-activity relationship studies, mass spectrometry experiments, and spectroscopic data reveals that the molecular mechanism of antifungal action conferred by HON involves enzyme-dependent formation of a covalent adduct between C4 of the nicotinamide ring of NAD(+) and C5 of HON. Furthermore, novel interactions are involved in stabilizing the (HON*NAD)-adduct, which are not observed in the enzyme's ternary complex structure. These findings clarify the apparent paradox of the potent antifungal actions of HON given its weak steady-state inhibition characteristics.


Assuntos
Ácido Aminolevulínico/farmacologia , Antifúngicos/farmacologia , Homosserina Desidrogenase/antagonistas & inibidores , Ácido Aminolevulínico/análogos & derivados , Ácido Aminolevulínico/química , Sítios de Ligação , Cristalografia por Raios X , Inibidores Enzimáticos/farmacologia , Homosserina Desidrogenase/química , Homosserina Desidrogenase/metabolismo , Cinética , NAD/química , NAD/metabolismo , Relação Estrutura-Atividade
8.
Biochim Biophys Acta ; 1544(1-2): 28-41, 2001 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-11341914

RESUMO

Fungal homoserine dehydrogenase (HSD) is required for the biosynthesis of threonine, isoleucine and methionine from aspartic acid, and is a target for antifungal agents. HSD from the yeast Saccharomyces cerevisiae was overproduced in Escherichia coli and 25 mg of soluble dimeric enzyme was purified per liter of cell culture in two steps. HSD efficiently reduces aspartate semialdehyde to homoserine (Hse) using either NADH or NADPH with kcat/Km in the order of 10(6-7) M(-1) x s(-1) at pH 7.5. The rate constant of the reverse direction (Hse oxidation) was also significant at pH 9.0 (kcat/Km approximately 10(4-5) M(-1) x s(-1)) but was minimal at pH 7.5. Chemical modification of HSD with diethyl pyrocarbonate (DEPC) resulted in a loss of activity that could be obviated by the presence of substrates. UV difference spectra revealed an increase in absorbance at 240 nm for DEPC-modified HSD consistent with the modification of two histidines (His) per subunit. Amino acid sequence alignment of HSD illustrated the conservation of two His residues among HSDs. These residues, His79 and His309, were substituted to alanine (Ala) using site directed mutagenesis. HSD H79A had similar steady state kinetics to wild type, while kcat/Km for HSD H309A decreased by almost two orders of magnitude. The recent determination of the X-ray structure of HSD revealed that His309 is located at the dimer interface [B. DeLaBarre, P.R. Thompson, G.D. Wright, A.M. Berghuis, Nat. Struct. Biol. 7 (2000) 238-244]. The His309Ala mutant enzyme was found in very high molecular weight complexes rather than the expected dimer by analytical gel filtration chromatography analysis. Thus the invariant His309 plays a structural rather than catalytic role in these enzymes.


Assuntos
Antifúngicos/farmacologia , Homosserina Desidrogenase/antagonistas & inibidores , Saccharomyces cerevisiae/enzimologia , Sequência de Aminoácidos , Sequência de Bases , Primers do DNA , Homosserina Desidrogenase/química , Homosserina Desidrogenase/genética , Homosserina Desidrogenase/metabolismo , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Filogenia , Homologia de Sequência de Aminoácidos
9.
Biochim Biophys Acta ; 1544(1-2): 42-54, 2001 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-11341915

RESUMO

Homoserine dehydrogenase (HSD), which is required for the synthesis of threonine, isoleucine and methionine in fungi, is a potential target for novel antifungal drugs. In order to design effective inhibitors, the kinetic mechanism of Saccharomyces cerevisiae HSD and the stereochemistry of hydride transfer were examined. Product inhibition experiments revealed that yeast HSD follows an ordered Bi Bi kinetic mechanism, where NAD(P)H must bind the enzyme prior to aspartate semialdehyde (ASA) and homoserine is released first followed by NAD(P)+. H-(1,2,4-triazol-3-yl)-D,L-alanine was an uncompetitive inhibitor of HSD with respect to NADPH (K(ii)=3.04+/-0.18 mM) and a noncompetitive inhibitor with respect to ASA (K(is)=1.64+/-0.36 mM, K(ii)=3.84+/-0.46 mM), in agreement with the proposed substrate order. Both kinetic isotope and viscosity experiments provided evidence for a very rapid catalytic step and suggest nicotinamide release to be primarily rate limiting. Incubation of HSD with stereospecifically deuterated NADP[2H] and subsaturating amounts of aspartate semialdehyde revealed that the pro-S NADPH hydride is transferred to the aldehyde. The pH dependence of steady state kinetic parameters indicate that ionizable groups with basic pKs may be involved in substrate binding, consistent with the observation of Lys223 at the enzyme active site in the recently determined 3D structure [B. DeLaBarre, P.R. Thompson, G.D. Wright, A.M. Berghuis, Nat. Struct. Biol. 7 (2000) 238-244]. These findings provide the requisite foundation for future exploitation of fungal HSD in inhibitor design.


Assuntos
Homosserina Desidrogenase/metabolismo , Hidrogênio/metabolismo , Saccharomyces cerevisiae/enzimologia , Homosserina Desidrogenase/antagonistas & inibidores , Homosserina Desidrogenase/química , Concentração de Íons de Hidrogênio , Cinética , NAD/metabolismo , Estereoisomerismo , Viscosidade
10.
Arch Microbiol ; 161(4): 359-62, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-8002714

RESUMO

The enzymes aspartokinase and homoserine dehydrogenase catalyze the reaction at key branching points in the aspartate pathway of amino acid biosynthesis. Enterococcus faecium has been found to contain two distinct aspartokinases and a single homoserine dehydrogenase. Aspartokinase isozymes eluted on gel filtration chromatography at molecular weights greater than 250,000 and about 125,000. The molecular weight of homoserine dehydrogenase was determined to be 220,000. One aspartokinase isozyme was slightly inhibited by meso-diaminopimelic acid. Another aspartokinase was repressed and inhibited by lysine. Although the level of diaminopimelate-sensitive (DAPs) enzyme was not much affected by growth conditions, the activity of lysine-sensitive (Lyss) aspartokinase disappeared rapidly during the stationary phase and was depressed in rich media. The synthesis of homoserine dehydrogenase was controlled by threonine and methionine. Threonine also inhibited the specific activity of this enzyme. The regulatory properties of aspartokinase isozymes and homoserine dehydrogenase from E. faecium are discussed and compared with those from Bacillus subtilis.


Assuntos
Aspartato Quinase/metabolismo , Ácido Aspártico/metabolismo , Enterococcus faecium/enzimologia , Homosserina Desidrogenase/metabolismo , Isoenzimas/metabolismo , Aminoácidos/farmacologia , Animais , Aspartato Quinase/antagonistas & inibidores , Aspartato Quinase/isolamento & purificação , Divisão Celular , Enterococcus faecium/crescimento & desenvolvimento , Homosserina Desidrogenase/antagonistas & inibidores , Homosserina Desidrogenase/isolamento & purificação , Isoenzimas/antagonistas & inibidores , Isoenzimas/isolamento & purificação , Rúmen/microbiologia
11.
J Antibiot (Tokyo) ; 45(5): 750-5, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1352515

RESUMO

An antifungal antibiotic (S) 2-amino-4-oxo-5-hydroxypentanoic acid, inhibited the biosynthesis of the aspartate family of amino acids (methionine, isoleucine and threonine) followed by the inhibition of protein biosynthesis in Saccharomyces cerevisiae. This inhibition was effected by impeding the biosynthesis of their common intermediate precursor, homoserine. The inhibition of biosynthesis of homoserine by the antibiotic was attributable to inactivation of homoserine dehydrogenase [EC 1.1.1.3], which is involved in the conversion of aspartate semialdehyde to homoserine in the metabolic pathway leading to threonine, methionine and isoleucine. Since such enzymic activity is not present in animal cells, the selective antifungal activity of the antibiotic is thus explained.


Assuntos
Antifúngicos/farmacologia , Homosserina Desidrogenase/antagonistas & inibidores , Saccharomyces cerevisiae/química , Ácido Aminolevulínico/análogos & derivados , Ácido Aminolevulínico/isolamento & purificação , Ácido Aminolevulínico/farmacologia , Antifúngicos/isolamento & purificação , Aspartato Quinase/efeitos dos fármacos , Aspartato-Semialdeído Desidrogenase/efeitos dos fármacos , Cinética
12.
Gene ; 107(1): 53-9, 1991 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-1743520

RESUMO

In Escherichia coli, Bacillus subtilis and Corynebacterium glutamicum, homoserine dehydrogenase (HD), the enzyme after the branch point of the threonine/methionine and lysine biosynthetic pathways, is allosterically inhibited by L-threonine. To investigate the regulation of the C. glutamicum HD enzyme by L-threonine, the structural gene, hom, was mutated by UV irradiation of whole cells to obtain a deregulated allele, homdr. L-Threonine inhibits the wild-type (wt) enzyme with a Ki of 0.16 mM. The deregulated enzyme remains 80% active in the presence of 50 mM L-threonine. The homdr gene mutant was isolated and cloned in E. coli. In a C. glutamicum wt host background, but not in E. coli, the cloned homdr gene is genetically unstable. The cloned homdr gene is overexpressed tenfold in C. glutamicum and is active in the presence of over 60 mM L-threonine. Sequence analysis revealed that the homdr mutation is a single nucleotide (G1964) deletion in codon 429 within the hom reading frame. The resulting frame-shift mutation radically alters the structure of the C terminus, resulting in ten amino acid (aa) changes and a deletion of the last 7 aa relative to the wt protein. These observations suggest that the C terminus may be associated with the L-threonine allosteric response. The homdr mutation is unstable and probably deleterious to the cell. This may explain why only one mutation was obtained despite repeated mutagenesis.


Assuntos
Corynebacterium/enzimologia , Regulação Bacteriana da Expressão Gênica/genética , Homosserina Desidrogenase/genética , Treonina/farmacologia , Regulação Alostérica/genética , Sequência de Aminoácidos , Sequência de Bases , Deleção Cromossômica , Clonagem Molecular , Corynebacterium/genética , Homosserina Desidrogenase/antagonistas & inibidores , Dados de Sequência Molecular , Mutagênese , Mutação/genética , Mapeamento por Restrição
13.
J Biochem ; 109(4): 604-8, 1991 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1907968

RESUMO

We have reported that a major cause of growth inhibition of Escherichia coli by L-serine is its inhibition of homoserine dehydrogenase I (HDH I), which is involved in the biosyntheses of threonine and isoleucine [Hama, H., Sumita, Y., Kakutani, Y., Tsuda, M., & Tsuchiya, T. (1990) Biochem. Biophys. Res. Commun. 168, 1211-1216]. However, Patte et al. reported that L-serine does not inhibit HDH I [Patte, J.-C., Truffa-Bachi, P., & Cohen, G.N. (1966) Biochim. Biophys. Acta 128, 426-439]. In studies on the reason for these discrepant results, we found that the concentration of K+ and the pH in the assay mixture strongly influenced the inhibitory effect of L-serine. L-Serine strongly inhibited the HDH I activities in both the forward and reverse reactions between aspartate semialdehyde and homoserine at a physiological K+ concentration (100 to 200 mM) and physiological pH (7.5) for E. coli cells. On the other hand, two well-known inhibitors of HDH I, L-threonine and L-cysteine, strongly inhibited the activity regardless of the K+ concentration and pH.


Assuntos
Escherichia coli/enzimologia , Homosserina Desidrogenase/antagonistas & inibidores , Serina/farmacologia , Cátions Monovalentes , Cisteína/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Concentração de Íons de Hidrogênio , Cinética , Potássio/farmacologia , Treonina/farmacologia
14.
Biochem Biophys Res Commun ; 168(3): 1211-6, 1990 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-2111991

RESUMO

L-serine has long been known to inhibit growth of Escherichia coli cells cultured in minimal medium supplemented with glucose, lactate, or another carbohydrate as the sole source of carbon. However, the target of serine inhibition was not known. The growth inhibition was released by adding isoleucine, 2-ketobutyric acid, threonine or homoserine, but not by aspartate. Thus the inhibition site must be between aspartate and homoserine in the isoleucine biosynthetic pathway. We found that homoserine dehydrogenase I was strongly inhibited by serine. We isolated serine-resistant mutants, and found that in these mutants homoserine dehydrogenase I was resistant to serine. Thus, we conclude that the target of serine inhibition in Escherichia coli is homoserine dehydrogenase I.


Assuntos
Oxirredutases do Álcool/antagonistas & inibidores , Escherichia coli/efeitos dos fármacos , Homosserina Desidrogenase/antagonistas & inibidores , Serina/farmacologia , Divisão Celular/efeitos dos fármacos , Escherichia coli/enzimologia , Escherichia coli/crescimento & desenvolvimento , Homosserina Desidrogenase/isolamento & purificação , Isoleucina/biossíntese , Oxaloacetatos/metabolismo
15.
Biochemistry ; 28(22): 8771-7, 1989 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-2557908

RESUMO

The pH variation of the kinetic parameters was examined for the kinase activity of the bifunctional enzyme aspartokinase--homoserine dehydrogenase I isolated from Escherichia coli. The V/K profile for L-aspartic acid indicates the loss of activity upon protonation of a cationic acid type group with a pK value near neutrality. Incubation of the enzyme with diethyl pyrocarbonate at pH 6.0 results in a loss of enzymic activity. The reversal of this reaction by neutral hydroxylamine, the appearance of a peak at 242 nm for the inactivated enzyme, and the observation of a pK value of 7.0 obtained from variation of the inactivation rate with pH all suggest that enzyme inactivation occurs by modification of histidine residues. The substrate L-aspartic acid protects one residue against inactivation, which implies that this histidine may participate in substrate binding or catalysis. Activity loss was also observed at high pH due to the ionization of a neutral acid group with a pK value of 9.8. The reactions of AK-HSD I with N-acetylimidazole and tetranitromethane have been investigated to obtain information about the functional role of tyrosyl residues in the enzyme. The acylation of tyrosines leads to inactivation of the enzyme, which can then be fully reversed by treatment with hydroxylamine. Incubation of the enzyme with tetranitromethane at pH 9.5 also leads to rapid inactivation, and the substrates of the kinase reaction provide substantial protection against inactivation. However, three tyrosines are protected by substrates, implying a structural role for these amino acids.


Assuntos
Oxirredutases do Álcool/metabolismo , Aspartato Quinase/metabolismo , Escherichia coli/enzimologia , Homosserina Desidrogenase/metabolismo , Fosfotransferases/metabolismo , Aspartato Quinase/antagonistas & inibidores , Dietil Pirocarbonato/farmacologia , Ativação Enzimática/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Homosserina Desidrogenase/antagonistas & inibidores , Concentração de Íons de Hidrogênio , Imidazóis/farmacologia , Cinética , Tetranitrometano/farmacologia , Raios Ultravioleta
16.
Eur J Biochem ; 180(3): 587-93, 1989 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-2496980

RESUMO

Mycobacterium smegmatis grows best on L-asparagine as a sole nitrogen source; this was confirmed. [14C]Aspartate was taken up rapidly (46 nmol.mg dry cells-1.h-1 from 1 mM L-asparagine) and metabolised to CO2 as well as to amino acids synthesised through the aspartate pathway. Proportionately more radioactivity appeared in the amino acids in bacteria grown in medium containing low nitrogen. Activities of aspartokinase and homoserine dehydrogenase, the initial enzymes of the aspartate pathway, were carried by separate proteins. Aspartokinase was purified as three isoenzymes and represented up to 8% of the soluble protein of M. smegmatis. All three isoenzymes contained molecular mass subunits of 50 kDa and 11 kDa which showed no activity individually; full enzyme activity was recovered on pooling the subunits. Km values for aspartate were: aspartokinases I and III, 2.4 mM; aspartokinase II, 6.4 mM. Aspartokinase I was inhibited by threonine and homoserine and aspartokinase III by lysine, but aspartokinase II was not inhibited by any amino acids. Aspartokinase activity was repressed by methionine and lysine with a small residue of activity attributable to unrepressed aspartokinase I. Homoserine dehydrogenase activity was 96% inhibited by 2 mM threonine; isoleucine, cysteine and valine had lesser effects and in combination gave additive inhibition. Homoserine dehydrogenase was repressed by threonine and leucine. Only amino acids synthesised through the aspartate pathway were tested for inhibition and repression. Of these, only one, meso-diaminopimilate, had no discernable effect on either enzyme activity.


Assuntos
Ácido Aspártico/metabolismo , Mycobacterium/metabolismo , Aspartato Quinase/antagonistas & inibidores , Aspartato Quinase/metabolismo , Sítios de Ligação/efeitos dos fármacos , Ligação Competitiva , Dióxido de Carbono/biossíntese , Homosserina/farmacologia , Homosserina Desidrogenase/antagonistas & inibidores , Homosserina Desidrogenase/metabolismo , Isoenzimas/metabolismo , Lisina/farmacologia , Mycobacterium/enzimologia , Mycobacterium/crescimento & desenvolvimento , Nitrogênio/metabolismo , Treonina/farmacologia
17.
Arch Biochem Biophys ; 227(1): 210-24, 1983 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-6357097

RESUMO

The threonine-sensitive homoserine dehydrogenase (L-homoserine: NAD(P)+ oxido-reductase), isolated from seedlings of Zea mays L., is characterized by variable kinetic and regulatory properties. Previous analysis of this enzyme suggested that it is capable of ligand-mediated interconversions among four kinetically distinct states (S. Krishnaswamy and J. K. Bryan (1983) Arch. Biochem. Biophys. 222, 449-463). These forms of the enzyme have been identified and found to differ in oligomeric configuration and conformation. In the presence of KCl and threonine a rapid equilibrium among three species of the enzyme (B, T, and K) is established. Each of these species can undergo a unique slow transition to a steady-state form under assay conditions. Results obtained from gel-filtration chromatography and sucrose density centrifugation indicate that the B and steady-state forms are tetramers and the T and K states are dimers. Evidence is presented to indicate that the rapid conversion from one dimeric species to the other can only occur via formation of the tetrameric B state. Chromatography under reacting-enzyme conditions provides direct support for the slow formation of a common steady-state species from any one of the other forms of the enzyme. The rate of transition is influenced by threonine, homoserine, NAD+, and, for transitions involving association reactions, by enzyme concentration. Small, reproducible differences in the apparent size of the T and K forms, and the B and steady-state species, are attributed to changes in conformation. This conclusion is supported by differential susceptibility of the enzymic states to proteolytic inactivation, by different rates of inactivation by dithio-bis-nitrobenzoate, and by alterations in their thermal stability. In addition, the B, T, and K states of the enzyme exhibit unique intrinsic fluorescence spectra. Spectral changes are shown to closely parallel changes in kinetic and hysteretic properties of the enzyme. The results of diverse methods of analysis are internally consistent, and provide considerable support for the conclusion that this pleiotropic regulatory enzyme can exist in any of several physically distinct states.


Assuntos
Oxirredutases do Álcool/biossíntese , Homosserina Desidrogenase/biossíntese , Ligantes/farmacologia , Zea mays/enzimologia , Centrifugação com Gradiente de Concentração , Cromatografia em Gel , Eletroforese em Gel de Poliacrilamida , Indução Enzimática/efeitos dos fármacos , Homosserina Desidrogenase/antagonistas & inibidores , Cinética , Peso Molecular , Conformação Proteica , Espectrometria de Fluorescência , Compostos de Sulfidrila/metabolismo , Temperatura
18.
Can J Microbiol ; 26(12): 1386-91, 1980 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-6786715

RESUMO

Homoserine dehydrogenase in dialyzed cell extracts of Bacillus subtilis 168 was studied, particularly with regard to inhibition, repression, and level of activity as a function of stage of development (growth and sporulation). It was assayed in the "forward direction" using L-aspartic semialdehyde and NADPH as substrates. Of the potentials inhibitors tested, only cysteine and NADP were found to be effective. Both L- and D-cysteine were equally effective. Therefore, the physiological significance of cysteine as an inhibitor is somewhat questionable. Amino acids involved in repression of homoserine dehydrogenase included methionine, isoleucine, possibly threonine, and one or more unidentified components of Casamino acids. The specific activity of homoserine dehydrogenase was highest during the exponential phase of growth and declined steadily during the stationary phase of growth. The low specific activity during late sporulation may favor preferential funnelling of L-aspartic semialdehyde into the lysine pathway, where it is needed for synthesis of large amounts of dipicolinic acid and diaminopimelic acid.


Assuntos
Oxirredutases do Álcool/metabolismo , Bacillus subtilis/enzimologia , Homosserina Desidrogenase/metabolismo , Bacillus subtilis/fisiologia , Repressão Enzimática , Homosserina Desidrogenase/antagonistas & inibidores , Esporos Fúngicos
20.
Proc Natl Acad Sci U S A ; 74(11): 4862-6, 1977 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-270718

RESUMO

Incubation of Rhodospirillum rubrum homoserine dehydrogenase (L-homoserine:NAD+ oxidoreductase, EC 1.1.1.3) with p-mercuribenzoate (PMB) in the presence of 0.2 M KCl and 2 mM L-threonine resulted in complete loss of enzyme activity. Upon removal of excess PMB, KCl, and L-threonine, a time-dependent recovery of enzyme activity was observed in 25 mM phosphate/I mM EDTA buffer, pH 7.5. Circular dichroism studies indicated that the transition from inactive to reactivated form of the enzyme was accompanied by a conformational change in the protein. Experiments with [14C]PMB revealed loss of enzyme-bound radioactivity during reactivation. Increase in ionic strength of the phosphate buffer and/or addition of L-threonine, leading to enzyme aggregation, decreased the rate of enzyme reactivation, aggregated enzyme that remained inactive retained [14C]PMB on the enzyme. Sulfhydryl titration of various forms of the enzyme suggested a preferential release of PMB from a sulfhydryl group essential to enzymic activity. We conclude that reactivation of the inactive enzyme is due to dissociation of PMB from an "active-site" sulfhydryl group and that changes in the protein structure influence the rate of dissociation of the enzyme-PMB complex.


Assuntos
Oxirredutases do Álcool/metabolismo , Homosserina Desidrogenase/metabolismo , Mercurobenzoatos/metabolismo , Dicroísmo Circular , Ativação Enzimática , Homosserina Desidrogenase/antagonistas & inibidores , Cinética , Ligantes , Mercurobenzoatos/farmacologia , Conformação Proteica , Compostos de Sulfidrila/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...