Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Biol Chem ; 288(46): 33439-48, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24064214

RESUMO

Formation of the peptidoglycan stem pentapeptide requires the insertion of both L and D amino acids by the ATP-dependent ligase enzymes MurC, -D, -E, and -F. The stereochemical control of the third position amino acid in the pentapeptide is crucial to maintain the fidelity of later biosynthetic steps contributing to cell morphology, antibiotic resistance, and pathogenesis. Here we determined the x-ray crystal structure of Staphylococcus aureus MurE UDP-N-acetylmuramoyl-L-alanyl-D-glutamate:meso-2,6-diaminopimelate ligase (MurE) (E.C. 6.3.2.7) at 1.8 Šresolution in the presence of ADP and the reaction product, UDP-MurNAc-L-Ala-γ-D-Glu-L-Lys. This structure provides for the first time a molecular understanding of how this Gram-positive enzyme discriminates between L-lysine and D,L-diaminopimelic acid, the predominant amino acid that replaces L-lysine in Gram-negative peptidoglycan. Despite the presence of a consensus sequence previously implicated in the selection of the third position residue in the stem pentapeptide in S. aureus MurE, the structure shows that only part of this sequence is involved in the selection of L-lysine. Instead, other parts of the protein contribute substrate-selecting residues, resulting in a lysine-binding pocket based on charge characteristics. Despite the absolute specificity for L-lysine, S. aureus MurE binds this substrate relatively poorly. In vivo analysis and metabolomic data reveal that this is compensated for by high cytoplasmic L-lysine concentrations. Therefore, both metabolic and structural constraints maintain the structural integrity of the staphylococcal peptidoglycan. This study provides a novel focus for S. aureus-directed antimicrobials based on dual targeting of essential amino acid biogenesis and its linkage to cell wall assembly.


Assuntos
Proteínas de Bactérias/química , Parede Celular/enzimologia , Lisina/química , Peptídeo Sintases/química , Peptidoglicano/química , Staphylococcus aureus/enzimologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Parede Celular/genética , Cristalografia por Raios X , Lisina/genética , Lisina/metabolismo , Metabolômica , Peptídeo Sintases/genética , Peptídeo Sintases/metabolismo , Peptidoglicano/biossíntese , Peptidoglicano/genética , Estrutura Terciária de Proteína , Staphylococcus aureus/genética
2.
Cell Microbiol ; 12(11): 1666-79, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20597908

RESUMO

Pseudomonas entomophila is an entomopathogenic bacterium that is lethal to Drosophila melanogaster within 1-2 days of ingestion of high doses. Flies orally infected with P. entomophila rapidly succumb despite the induction of both local and systemic immune responses. Recent studies suggest that its virulence relies on its ability to cause irreversible damages to the intestinal epithelium, in contrast to what is observed with milder pathogenic bacteria such as Erwinia carotovora carotovora Ecc15 or Pseudomonas aeruginosa PA14. The GacS/GacA two-component system plays a key role in P. entomophila pathogenicity. Here, we report the identification of the pvf genes, whose products are involved in production of a secondary metabolite involved in P. entomophila virulence. A pvf mutant is impaired in its ability to persist within the gut, to trigger the fly immune responses and to inflict gut damages. The expression of several genes is affected in a pvf mutant, independently of the Gac system. Moreover, growing a pvf mutant in medium supplemented with supernatant extracts from either the wild-type strain or a gacA mutant restore its pathogenicity. Collectively, our results indicate that we identified genes involved in the synthesis of a signalling molecule that controls P. entomophila virulence independently from the Gac system.


Assuntos
Proteínas de Bactérias/metabolismo , Drosophila melanogaster/microbiologia , Pseudomonas/metabolismo , Pseudomonas/patogenicidade , Transdução de Sinais , Fatores de Virulência/metabolismo , Animais , Proteínas de Bactérias/genética , Expressão Gênica , Regulação Bacteriana da Expressão Gênica , Genes Bacterianos , Mucosa Intestinal/microbiologia , Mutação , Peptídeo Sintases/metabolismo , Policetídeo Sintases , Pseudomonas/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Virulência/genética
3.
FEMS Microbiol Rev ; 32(2): 168-207, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18266853

RESUMO

The biosynthesis of bacterial cell wall peptidoglycan is a complex process that involves enzyme reactions that take place in the cytoplasm (synthesis of the nucleotide precursors) and on the inner side (synthesis of lipid-linked intermediates) and outer side (polymerization reactions) of the cytoplasmic membrane. This review deals with the cytoplasmic steps of peptidoglycan biosynthesis, which can be divided into four sets of reactions that lead to the syntheses of (1) UDP-N-acetylglucosamine from fructose 6-phosphate, (2) UDP-N-acetylmuramic acid from UDP-N-acetylglucosamine, (3) UDP-N-acetylmuramyl-pentapeptide from UDP-N-acetylmuramic acid and (4) D-glutamic acid and dipeptide D-alanyl-D-alanine. Recent data concerning the different enzymes involved are presented. Moreover, special attention is given to (1) the chemical and enzymatic synthesis of the nucleotide precursor substrates that are not commercially available and (2) the search for specific inhibitors that could act as antibacterial compounds.


Assuntos
Bactérias/metabolismo , Vias Biossintéticas , Citoplasma/metabolismo , Peptidoglicano/biossíntese , Bactérias/enzimologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Fosfoglucomutase/química , Fosfoglucomutase/metabolismo , Uridina Difosfato N-Acetilglicosamina/biossíntese
4.
Bioorg Med Chem ; 17(5): 1884-9, 2009 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-19223185

RESUMO

The ATP-dependent Mur ligases (MurC, MurD, MurE and MurF) successively add L-Ala, D-Glu, meso-A(2)pm or L-Lys, and D-Ala-D-Ala to the nucleotide precursor UDP-MurNAc, and they represent promising targets for antibacterial drug discovery. We have used the molecular docking programme eHiTS for the virtual screening of 1990 compounds from the National Cancer Institute 'Diversity Set' on MurD and MurF. The 50 top-scoring compounds from screening on each enzyme were selected for experimental biochemical evaluation. Our approach of virtual screening and subsequent in vitro biochemical evaluation of the best ranked compounds has provided four novel MurD inhibitors (best IC(50)=10 microM) and one novel MurF inhibitor (IC(50)=63 microM).


Assuntos
Antibacterianos/química , Inibidores Enzimáticos/química , Peptídeo Sintases/antagonistas & inibidores , Antibacterianos/farmacologia , Biologia Computacional , Simulação por Computador , Bases de Dados Factuais , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Peptídeo Sintases/química , Peptídeo Sintases/metabolismo , Peptidoglicano/biossíntese
5.
Biochem Pharmacol ; 84(5): 625-32, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22705647

RESUMO

D-Glutamic acid-adding enzyme (MurD ligase) catalyses the addition of D-glutamic acid to UDP-N-acetylmuramoyl-L-alanine, an essential cytoplasmic step in the pathway for bacterial cell-wall peptidoglycan synthesis. As such, it represents an important antibacterial drug-discovery target enzyme. Recently, several series of compounds have been synthesised and found to inhibit MurD from Escherichia coli, the best one having an IC(50) value of 8 µM. In the present work, we have tested 20 of these compounds against the MurD enzymes from Staphylococcus aureus, Streptococcus pneumoniae, Borrelia burgdorferi and Mycobacterium tuberculosis. Most of the E. coli MurD inhibitors appeared less efficient against the four other orthologues. This divergent result can be explained by the differences in amino acid sequences and topologies of the active sites of the MurD ligases studied.


Assuntos
Inibidores Enzimáticos/farmacologia , Escherichia coli/enzimologia , Peptídeo Sintases/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , Concentração Inibidora 50 , Modelos Moleculares , Peptídeo Sintases/química , Reação em Cadeia da Polimerase
6.
Biochimie ; 92(12): 1793-800, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20659527

RESUMO

The Mur ligases (MurC, MurD, MurE and MurF) catalyze the stepwise synthesis of the UDP-N-acetylmuramoyl-pentapeptide precursor of peptidoglycan. The murC, murD, murE and murF genes from Staphylococcus aureus, a major pathogen, were cloned and the corresponding proteins were overproduced in Escherichia coli and purified as His(6)-tagged forms. Their biochemical properties were investigated and compared to those of the E. coli enzymes. Staphylococcal MurC accepted L-Ala, L-Ser and Gly as substrates, as the E. coli enzyme does, with a strong preference for L-Ala. S. aureus MurE was very specific for L-lysine and in particular did not accept meso-diaminopimelic acid as a substrate. This mirrors the E. coli MurE specificity, for which meso-diaminopimelic acid is the preferred substrate and L-lysine a very poor one. S. aureus MurF appeared less specific and accepted both forms (L-lysine and meso-diaminopimelic acid) of UDP-MurNAc-tripeptide, as the E. coli MurF does. The inverse and strict substrate specificities of the two MurE orthologues is thus responsible for the presence of exclusively meso-diaminopimelic acid and L-lysine at the third position of the peptide in the peptidoglycans of E. coli and S. aureus, respectively. The specific activities of the four Mur ligases were also determined in crude extracts of S. aureus and compared to cell requirements for peptidoglycan biosynthesis.


Assuntos
Proteínas de Bactérias/metabolismo , Ligases/metabolismo , Proteínas Recombinantes/metabolismo , Staphylococcus aureus/enzimologia , Trifosfato de Adenosina/metabolismo , Alanina/metabolismo , Proteínas de Bactérias/genética , Ácido Diaminopimélico/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Teste de Complementação Genética , Glicina/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Ligases/genética , Mutação , Oligopeptídeos/metabolismo , Peptidoglicano/metabolismo , Serina/metabolismo , Staphylococcus aureus/metabolismo , Especificidade por Substrato , Temperatura
7.
J Biol Chem ; 284(33): 21856-21862, 2009 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-19542229

RESUMO

Thermotoga maritima is a Gram-negative, hyperthermophilic bacterium whose peptidoglycan contains comparable amounts of L- and D-lysine. We have determined the fine structure of this cell-wall polymer. The muropeptides resulting from the digestion of peptidoglycan by mutanolysin were separated by high-performance liquid chromatography and identified by amino acid analysis after acid hydrolysis, dinitrophenylation, enzymatic determination of the configuration of the chiral amino acids, and mass spectrometry. The high-performance liquid chromatography profile contained four main peaks, two monomers, and two dimers, plus a few minor peaks corresponding to anhydro forms. The first monomer was the d-lysine-containing disaccharide-tripeptide in which the D-Glu-D-Lys bond had the unusual gamma-->epsilon arrangement (GlcNAc-MurNAc-L-Ala-gamma-D-Glu-epsilon-D-Lys). The second monomer was the conventional disaccharide-tetrapeptide (GlcNAc-MurNAc-L-Ala-gamma-D-Glu-L-Lys-D-Ala). The first dimer contained a disaccharide-L-Ala as the acyl donor cross-linked to the alpha-amine of D-Lys in a tripeptide acceptor stem with the sequence of the first monomer. In the second dimer, donor and acceptor stems with the sequences of the second and first monomers, respectively, were connected by a D-Ala4-alpha-D-Lys3 cross-link. The cross-linking index was 10 with an average chain length of 30 disaccharide units. The structure of the peptidoglycan of T. maritima revealed for the first time the key role of D-Lys in peptidoglycan synthesis, both as a surrogate of L-Lys or meso-diaminopimelic acid at the third position of peptide stems and in the formation of novel cross-links of the L-Ala1(alpha-->alpha)D-Lys3 and D-Ala4(alpha-->alpha)D-Lys3 types.


Assuntos
Reagentes de Ligações Cruzadas/farmacologia , Peptidoglicano/química , Thermotoga maritima/metabolismo , Alanina/química , Parede Celular/metabolismo , Cromatografia Líquida de Alta Pressão/métodos , Dissacarídeos/química , Endopeptidases/química , Lisina/química , Espectrometria de Massas/métodos , Modelos Químicos , Peptídeos/química , Polímeros/química , Polissacarídeos/química , Fatores de Tempo
8.
ChemMedChem ; 3(9): 1362-70, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18651694

RESUMO

The Mur ligases have an essential role in the intracellular biosynthesis of bacterial peptidoglycan, and they represent attractive targets for the design of novel antibacterials. A series of compounds with an N-acylhydrazone scaffold were synthesized and screened for inhibition of the MurC and MurD enzymes from Escherichia coli. Compounds with micromolar inhibitory activities against both MurC and MurD were identified, and some of them also showed antibacterial activity.


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Escherichia coli/efeitos dos fármacos , Hidrazonas/síntese química , Hidrazonas/farmacologia , Ligases/antagonistas & inibidores , Antibacterianos/síntese química , Antibacterianos/química , Antibacterianos/farmacologia , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/química , Cristalografia por Raios X , Inibidores Enzimáticos/química , Escherichia coli/enzimologia , Hidrazonas/química , Ligases/química , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Molecular , Peptidoglicano/biossíntese , Staphylococcus aureus/efeitos dos fármacos , Estereoisomerismo , Relação Estrutura-Atividade
9.
Arch Pharm (Weinheim) ; 340(3): 127-34, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17335103

RESUMO

The increasing emergence of pathogenic bacterial strains with high resistance to antibiotic therapy has created an urgent need for the development of new antibacterial agents that are directed towards novel targets. We have focused our attention on the Mur ligases (MurC-F), which catalyze the early steps of bacterial peptidoglycan biosynthesis, and which to date represent under-exploited targets for antibacterial drug design. We show that some of our phosphinate inhibitors of UDP-N-acetylmuramoyl-L-alanyl:D-glutamate ligase (MurD) also inhibits UDP-N-acetylmuramoyl-L-alanyl-D-glutamate:L-lysine ligase (MurE). To obtain new information on their structure-activity relationships, three new, structurally related phosphinates were synthesized and evaluated for inhibition of MurD and MurE.


Assuntos
Antibacterianos/síntese química , Inibidores Enzimáticos/síntese química , Peptídeo Sintases/química , Ácidos Fosfínicos/síntese química , Staphylococcus aureus/enzimologia , Antibacterianos/farmacologia , Simulação por Computador , Inibidores Enzimáticos/farmacologia , Modelos Moleculares , Estrutura Molecular , Peptídeo Sintases/antagonistas & inibidores , Peptídeo Sintases/isolamento & purificação , Ácidos Fosfínicos/farmacologia , Conformação Proteica , Relação Estrutura-Atividade
10.
J Bacteriol ; 189(11): 3987-95, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17384195

RESUMO

The UDP-N-acetylmuramate:L-alanyl-gamma-D-glutamyl-meso-diaminopimelate ligase (murein peptide ligase [Mpl]) is known to be a recycling enzyme allowing reincorporation into peptidoglycan (murein) of the tripeptide L-alanyl-gamma-D-glutamyl-meso-diaminopimelate released during the maturation and constant remodeling of this bacterial cell wall polymer that occur during cell growth and division. Mpl adds this peptide to UDP-N-acetylmuramic acid, thereby providing an economical additional source of UDP-MurNAc-tripeptide available for de novo peptidoglycan biosynthesis. The Mpl enzyme from Escherichia coli was purified to homogeneity as a His-tagged form, and its kinetic properties and parameters were determined. Mpl was found to accept tri-, tetra-, and pentapeptides as substrates in vitro with similar efficiencies, but it accepted the dipeptide L-Ala-D-Glu and L-Ala very poorly. Replacement of meso-diaminopimelic acid by L-Lys resulted in a significant decrease in the catalytic efficacy. The effects of disruption of the E. coli mpl gene and/or the ldcA gene encoding the LD-carboxypeptidase on peptidoglycan metabolism were investigated. The differences in the pools of UDP-MurNAc peptides and of free peptides between the wild-type and mutant strains demonstrated that the recycling activity of Mpl is not restricted to the tripeptide and that tetra- and pentapeptides are also directly reused by this process in vivo. The relatively broad substrate specificity of the Mpl ligase indicates that it is an interesting potential target for antibacterial compounds.


Assuntos
Ácido Diaminopimélico/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Peptídeo Sintases/metabolismo , Uridina Difosfato Ácido N-Acetilmurâmico/metabolismo , Ácido Diaminopimélico/química , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Teste de Complementação Genética , Cinética , Mutação , Peptídeo Sintases/genética , Peptidoglicano/metabolismo , Especificidade por Substrato , Temperatura , Uridina Difosfato Ácido N-Acetilmurâmico/química
11.
J Biol Chem ; 281(23): 15680-6, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16595662

RESUMO

The peptidoglycan of Thermotoga maritima, an extremely thermophilic eubacterium, was shown to contain no diaminopimelic acid and approximate amounts of both enantiomers of lysine (Huber, R., Langworthy, T. A., König, H., Thomm, M., Woese, C. R., Sleytr, U. B., and Stetter, K. O. (1986) Arch. Microbiol. 144, 324-333). To assess the possible involvement of the MurE activity in the incorporation of D-lysine, the murE gene from this organism was cloned in Escherichia coli, and the corresponding protein was purified as the C-terminal His6-tagged form. In vitro assays showed that D-lysine and meso-diaminopimelic acid were added to UDP-N-acetylmuramoyl-dipeptide with 25 and 10% efficiencies, respectively, relative to L-lysine. The purified enzyme was used to synthesize the L- and D-lysine-containing UDP-N-acetylmuramoyl-tripeptides; chemical analysis revealed an unusual structure for the D-lysine-containing nucleotide, namely acylation of the epsilon-amino function of D-lysine by the D-glutamyl residue. In vitro assays with MurF and MraY enzymes from T. maritima showed that this novel nucleotide was not a substrate for MurF but that it could be directly processed into tripeptide lipid I by MraY, thereby substantiating the role of MurE in the incorporation of D-lysine into peptidoglycan.


Assuntos
Ligases/metabolismo , Lisina/metabolismo , Thermotoga maritima/enzimologia , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , Primers do DNA , Genes Bacterianos , Ligases/química , Ligases/genética , Ligases/isolamento & purificação , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Thermotoga maritima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA