Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Environ Microbiol ; 24(9): 4466-4488, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35688634

RESUMO

The human pathogen Listeria monocytogenes synthesizes and degrades c-di-AMP using the diadenylate cyclase CdaA and the phosphodiesterases PdeA and PgpH respectively. c-di-AMP is essential because it prevents the uncontrolled uptake of osmolytes. Here, we studied the phenotypes of cdaA, pdeA, pgpH and pdeA pgpH mutants with defects in c-di-AMP metabolism and characterized suppressor mutants restoring their growth defects. The characterization of the pdeA pgpH mutant revealed that the bacteria show growth defects in defined medium, a phenotype that is invariably suppressed by mutations in cdaA. The previously reported growth defect of the cdaA mutant in rich medium is suppressed by mutations that osmotically stabilize the c-di-AMP-free strain. We also found that the cdaA mutant has an increased sensitivity against isoleucine. The isoleucine-dependent growth inhibition of the cdaA mutant is suppressed by codY mutations that likely reduce the DNA-binding activity of encoded CodY variants. Moreover, the characterization of the cdaA suppressor mutants revealed that the Opp oligopeptide transport system is involved in the uptake of the antibiotic fosfomycin. In conclusion, the suppressor analysis corroborates a key function of c-di-AMP in controlling osmolyte homeostasis in L. monocytogenes.


Assuntos
Fosfomicina , Listeria monocytogenes , Acetamidas , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , DNA/metabolismo , Fosfatos de Dinucleosídeos/metabolismo , Fosfomicina/metabolismo , Fosfomicina/farmacologia , Humanos , Isoleucina/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Oligopeptídeos/metabolismo , Diester Fosfórico Hidrolases/genética , Fósforo-Oxigênio Liases/genética
2.
Methods Enzymol ; 669: 45-70, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35644180

RESUMO

Fosfomycin is a clinically used broad-spectrum antibiotic that has the structure of an oxirane ring with a phosphonic acid substituent and a methyl substituent. In nature, fosfomycin is produced by Streptomyces spp. and Pseudomonas sp., but biosynthesis of fosfomycin significantly differs between the two bacteria, especially in the incorporation mechanism of the methyl group. It has been proposed that the cobalamin-dependent radical S-adenosyl-l-methionine (SAM) enzyme Fom3 is responsible for the methyl-transfer reaction in Streptomyces fosfomycin biosynthesis. In this chapter, we describe the experimental methods to characterize Fom3. We performed the methylation reaction with the purified recombinant Fom3, revealing that Fom3 recognizes a cytidylylated 2-hydroxyethylphosphonate as a substrate and catalyzes stereoselective methylation of the sp3 carbon at the C2 position to afford cytidylylated (S)-2-hydroxypropylphosphonate. Reaction analysis using deuterium-labeled substrates showed that the 5'-deoxyadenosyl radical generated by reductive cleavage of SAM stereoselectively abstracts the pro-R hydrogen atom of the CH bond at the C2 position of cytidylylated 2-hydroxyethylphosphonate. Therefore, the C-methylation reaction catalyzed by Fom3 proceeds with inversion of the configuration at the C2 position. Experimental methods to elucidate the chemical structures of the substrate and products and the stereochemical course in the Fom3-catalyzed reaction could give information to progress investigation of cobalamin-dependent radical SAM C-methyltransferases.


Assuntos
Fosfomicina , Streptomyces , Fosfomicina/química , Fosfomicina/metabolismo , Metiltransferases/metabolismo , S-Adenosilmetionina/metabolismo , Streptomyces/metabolismo , Vitamina B 12/metabolismo
3.
Mol Microbiol ; 116(1): 97-108, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33561903

RESUMO

The Gram-negative bacterium Pseudomonas aeruginosa is an opportunistic pathogen, responsible for many hospital-acquired infections. The bacterium is quite resistant toward many antibiotics, in particular because of the fine-tuned permeability of its outer membrane (OM). General diffusion outer membrane pores are quite rare in this organism. Instead, its OM contains many substrate-specific porins. Their expression is varying according to growth conditions and virulence. Phosphate limitations, as well as pathogenicity factors, result in the induction of the two mono- and polyphosphate-specific porins, OprP and OprO, respectively, together with an inner membrane uptake mechanism and a periplasmic binding protein. These outer membrane channels could serve as outer membrane pathways for the uptake of phosphonates. Among them are not only herbicides, but also potent antibiotics, such as fosfomycin and fosmidomycin. In this study, we investigated the interaction between OprP and OprO and fosmidomycin in detail. We could demonstrate that fosmidomycin is able to bind to the phosphate-specific binding site inside the two porins. The inhibition of chloride conductance of OprP and OprO by fosmidomycin is considerably less than that of phosphate or diphosphate, but it can be measured in titration experiments of chloride conductance and also in single-channel experiments. The results suggest that fosmidomycin transport across the OM of P. aeruginosa occurs through OprP and OprO. Our data with the ones already known in the literature show that phosphonic acid-containing antibiotics are in general good candidates to treat the infections of P. aeruginosa at the very beginning through a favorable OM transport system.


Assuntos
Antibacterianos/metabolismo , Proteínas de Bactérias/metabolismo , Fosfomicina/análogos & derivados , Transporte de Íons/fisiologia , Porinas/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/genética , Sítios de Ligação/fisiologia , Cloretos/metabolismo , Farmacorresistência Bacteriana Múltipla/genética , Fosfomicina/metabolismo , Ácidos Fosforosos/metabolismo , Porinas/genética , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética
5.
Biochemistry ; 56(28): 3519-3522, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28678474

RESUMO

A methylcobalamin (MeCbl)-dependent radical S-adenosyl-l-methionine (SAM) methyltransferase Fom3 was found to catalyze the C-methylation of cytidylyl-2-hydroxyethylphosphonate (HEP-CMP) to give cytidylyl-2-hydroxypropylphosphonate (HPP-CMP), although it was originally proposed to catalyze the C-methylation of 2-hydroxyethylphosphonate to give 2-hydroxypropylphosphonate in the biosynthesis of a unique C-P bond containing antibiotic fosfomycin in Streptomyces. Unexpectedly, the Fom3 reaction product from HEP-CMP was almost a 1:1 diastereomeric mixture of HPP-CMP, indicating that the C-methylation is not stereoselective. Presumably, only the CMP moiety of HEP-CMP is critical for substrate recognition; on the other hand, the enzyme does not fix the 2-hydroxy group of the substrate and either of the prochiral hydrogen atoms at the C2 position can be abstracted by the 5'-deoxyadenosyl radical generated from SAM to form the substrate radical intermediates, which react with MeCbl to afford the corresponding products. This strict substrate recognition mechanism with no stereoselectivity of a MeCbl-dependent radical SAM methyltransferase is remarkable in natural product biosynthetic chemistry, because such a hidden clue for selective substrate recognition is likely to be found in the other biosynthetic pathways.


Assuntos
Fosfomicina/metabolismo , Metiltransferases/metabolismo , Organofosfonatos/metabolismo , Streptomyces/enzimologia , Vitamina B 12/análogos & derivados , Vias Biossintéticas , Monofosfato de Citidina/metabolismo , Metilação , S-Adenosilmetionina/metabolismo , Streptomyces/metabolismo , Especificidade por Substrato , Vitamina B 12/metabolismo
6.
Antimicrob Agents Chemother ; 60(4): 2281-91, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26833150

RESUMO

Deciphering the mode of action (MOA) of new antibiotics discovered through phenotypic screening is of increasing importance. Metabolomics offers a potentially rapid and cost-effective means of identifying modes of action of drugs whose effects are mediated through changes in metabolism. Metabolomics techniques also collect data on off-target effects and drug modifications. Here, we present data from an untargeted liquid chromatography-mass spectrometry approach to identify the modes of action of eight compounds: 1-[3-fluoro-4-(5-methyl-2,4-dioxo-pyrimidin-1-yl)phenyl]-3-[2-(trifluoromethyl)phenyl]urea (AZ1), 2-(cyclobutylmethoxy)-5'-deoxyadenosine, triclosan, fosmidomycin, CHIR-090, carbonyl cyanidem-chlorophenylhydrazone (CCCP), 5-chloro-2-(methylsulfonyl)-N-(1,3-thiazol-2-yl)-4-pyrimidinecarboxamide (AZ7), and ceftazidime. Data analysts were blind to the compound identities but managed to identify the target as thymidylate kinase for AZ1, isoprenoid biosynthesis for fosmidomycin, acyl-transferase for CHIR-090, and DNA metabolism for 2-(cyclobutylmethoxy)-5'-deoxyadenosine. Changes to cell wall metabolites were seen in ceftazidime treatments, although other changes, presumably relating to off-target effects, dominated spectral outputs in the untargeted approach. Drugs which do not work through metabolic pathways, such as the proton carrier CCCP, have no discernible impact on the metabolome. The untargeted metabolomics approach also revealed modifications to two compounds, namely, fosmidomycin and AZ7. An untreated control was also analyzed, and changes to the metabolome were seen over 4 h, highlighting the necessity for careful controls in these types of studies. Metabolomics is a useful tool in the analysis of drug modes of action and can complement other technologies already in use.


Assuntos
Antibacterianos/farmacologia , Escherichia coli/efeitos dos fármacos , Metaboloma/efeitos dos fármacos , Metabolômica , Aciltransferases/antagonistas & inibidores , Aciltransferases/genética , Aciltransferases/metabolismo , Adenosina/metabolismo , Adenosina/farmacologia , Antibacterianos/metabolismo , Carbonil Cianeto m-Clorofenil Hidrazona/metabolismo , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Ceftazidima/metabolismo , Ceftazidima/farmacologia , Parede Celular/química , Parede Celular/efeitos dos fármacos , Parede Celular/metabolismo , Cromatografia Líquida , DNA Bacteriano/antagonistas & inibidores , DNA Bacteriano/biossíntese , Escherichia coli/genética , Escherichia coli/metabolismo , Fosfomicina/análogos & derivados , Fosfomicina/metabolismo , Fosfomicina/farmacologia , Expressão Gênica , Células HEK293 , Humanos , Ácidos Hidroxâmicos/metabolismo , Ácidos Hidroxâmicos/farmacologia , Espectrometria de Massas , Núcleosídeo-Fosfato Quinase/antagonistas & inibidores , Núcleosídeo-Fosfato Quinase/genética , Núcleosídeo-Fosfato Quinase/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacologia , Terpenos/antagonistas & inibidores , Terpenos/metabolismo , Treonina/análogos & derivados , Treonina/metabolismo , Treonina/farmacologia , Triclosan/metabolismo , Triclosan/farmacologia
7.
Biochem Pharmacol ; 102: 45-63, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26707799

RESUMO

Cucurbitacins are cytotoxic triterpenoid sterols isolated from plants. One of their earliest cellular effect is the aggregation of actin associated with blockage of cell migration and division that eventually lead to apoptosis. We unravel here that cucurbitacin I actually induces the co-aggregation of actin with phospho-myosin II. This co-aggregation most probably results from the stimulation of the Rho/ROCK pathway and the direct inhibition of the LIMKinase. We further provide data that suggest that the formation of these co-aggregates is independent of a putative pro-oxidant status of cucurbitacin I. The results help to understand the impact of cucurbitacins on signal transduction and actin dynamics and open novel perspectives to use it as drug candidates for cancer research.


Assuntos
Actinas/metabolismo , Quinases Lim/antagonistas & inibidores , Quinases Lim/metabolismo , Miosina Tipo II/metabolismo , Triterpenos/farmacologia , Quinases Associadas a rho/metabolismo , Actinas/química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/isolamento & purificação , Inibidores Enzimáticos/farmacologia , Fosfomicina/química , Fosfomicina/metabolismo , Células HeLa , Humanos , Miosina Tipo II/química , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/farmacologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Sementes , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Triterpenos/química , Triterpenos/isolamento & purificação , Quinases Associadas a rho/química
8.
J Mol Model ; 20(5): 2236, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24770549

RESUMO

Fosfomycin resistance kinases FomA, one of the key enzymes responsible for bacterial resistances to fosfomycin, has gained much attention recently due to the raising public concern for multi-drug resistant bacteria. Using molecular docking followed by molecular dynamics simulations, our group illustrated the process of fosfomycin induced conformational change of FomA. The detailed roles of the catalytic residues (Lys18, His58 and Thr210) during the formation of the enzyme-substrate complex were shown in our research. The organization functions of Gly53, Gly54, Ile61 and Leu75 were also highlighted. Furthermore, the cation-π interaction between Arg62 and Trp207 was observed and speculated to play an auxiliary role in the conformation change process of the enzyme. This detailed molecular level illustration of the formation of FomA·ATP·Mg·Fosfomycin complex could provide insight for both anti-biotic discovery and improvement of fosfomycin in the future.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Proteínas de Bactérias/química , Farmacorresistência Bacteriana Múltipla , Fosfomicina/química , Fosfomicina/farmacologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas Quinases/química , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Antibacterianos/metabolismo , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Catálise , Domínio Catalítico , Fosfomicina/metabolismo , Ligantes , Magnésio/química , Magnésio/metabolismo , Estrutura Molecular , Ligação Proteica , Conformação Proteica , Proteínas Quinases/metabolismo , Relação Estrutura-Atividade
9.
Biochemistry ; 52(41): 7350-62, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24004181

RESUMO

The fosfomycin resistance enzymes, FosB, from Gram-positive organisms, are M(2+)-dependent thiol tranferases that catalyze nucleophilic addition of either L-cysteine (L-Cys) or bacillithiol (BSH) to the antibiotic, resulting in a modified compound with no bacteriacidal properties. Here we report the structural and functional characterization of FosB from Bacillus cereus (FosB(Bc)). The overall structure of FosB(Bc), at 1.27 Å resolution, reveals that the enzyme belongs to the vicinal oxygen chelate (VOC) superfamily. Crystal structures of FosB(Bc) cocrystallized with fosfomycin and a variety of divalent metals, including Ni(2+), Mn(2+), Co(2+), and Zn(2+), indicate that the antibiotic coordinates to the active site metal center in an orientation similar to that found in the structurally homologous manganese-dependent fosfomycin resistance enzyme, FosA. Surface analysis of the FosB(Bc) structures show a well-defined binding pocket and an access channel to C1 of fosfomycin, the carbon to which nucleophilic addition of the thiol occurs. The pocket and access channel are appropriate in size and shape to accommodate L-Cys or BSH. Further investigation of the structures revealed that the fosfomycin molecule, anchored by the metal, is surrounded by a cage of amino acids that hold the antibiotic in an orientation such that C1 is centered at the end of the solvent channel, positioning the compound for direct nucleophilic attack by the thiol substrate. In addition, the structures of FosB(Bc) in complex with the L-Cys-fosfomycin product (1.55 Å resolution) and in complex with the bacillithiol-fosfomycin product (1.77 Å resolution) coordinated to a Mn(2+) metal in the active site have been determined. The L-Cys moiety of either product is located in the solvent channel, where the thiol has added to the backside of fosfomycin C1 located at the end of the channel. Concomitant kinetic analyses of FosB(Bc) indicated that the enzyme has a preference for BSH over L-Cys when activated by Mn(2+) and is inhibited by Zn(2+). The fact that Zn(2+) is an inhibitor of FosB(Bc) was used to obtain a ternary complex structure of the enzyme with both fosfomycin and L-Cys bound.


Assuntos
Antibacterianos/química , Bacillus cereus/enzimologia , Proteínas de Bactérias/química , Fosfomicina/metabolismo , Transferases/química , Antibacterianos/metabolismo , Bacillus cereus/química , Bacillus cereus/genética , Bacillus cereus/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , Cisteína/análogos & derivados , Cisteína/metabolismo , Fosfomicina/química , Glucosamina/análogos & derivados , Glucosamina/metabolismo , Cinética , Especificidade por Substrato , Transferases/genética , Transferases/metabolismo
10.
Org Lett ; 14(20): 5207-9, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-23030527

RESUMO

Bacillithiol (BSH) has been prepared on the gram scale from the inexpensive starting material, D-glucosamine hydrochloride, in 11 steps and 8-9% overall yield. The BSH was used to survey the substrate and metal-ion selectivity of FosB enzymes from four Gram-positive microorganisms associated with the deactivation of the antibiotic fosfomycin. The in vitro results indicate that the preferred thiol substrate and metal ion for the FosB from Staphylococcus aureus are BSH and Ni(II), respectively. However, the metal-ion selectivity is less distinct with FosB from Bacillus subtilis, Bacillus anthracis, or Bacillus cereus.


Assuntos
Biocatálise , Cisteína/análogos & derivados , Glucosamina/análogos & derivados , Proteínas Proto-Oncogênicas c-fos/metabolismo , Staphylococcus aureus/metabolismo , Bacillus/metabolismo , Cisteína/síntese química , Cisteína/metabolismo , Fosfomicina/química , Fosfomicina/metabolismo , Glucosamina/síntese química , Glucosamina/metabolismo , Estrutura Molecular , Especificidade por Substrato
11.
Artigo em Inglês | MEDLINE | ID: mdl-22505403

RESUMO

The development of new antibiotics is necessitated by the rapid development of resistance to current therapies. UDP-N-acetylglucosamine enolpyruvyl transferase (MurA), which catalyzes the first committed step of bacterial peptidoglycan biosynthesis, is a prime candidate for therapeutic intervention. MurA is the target of the antibiotic fosfomycin, a natural product produced by Streptomyces. Despite possessing a high degree of sequence conservation with MurA enzymes from fosfomycin-susceptible organisms, recent microbiological studies suggest that MurA from Vibrio fischeri (VfiMurA) may confer fosfomycin resistance via a mechanism that is not yet understood. The crystal structure of VfiMurA in a ternary complex with the substrate UDP-N-acetylglucosamine (UNAG) and fosfomycin has been solved to a resolution of 1.93 Å. Fosfomycin is known to inhibit MurA by covalently binding to a highly conserved cysteine in the active site of the enzyme. A comparison of the title structure with the structure of fosfomycin-susceptible Haemophilus influenzae MurA (PDB entry 2rl2) revealed strikingly similar conformations of the mobile substrate-binding loop and clear electron density for a fosfomycin-cysteine adduct. Based on these results, there are no distinguishing sequence/structural features in VfiMurA that would translate to a diminished sensitivity to fosfomycin. However, VfiMurA is a robust crystallizer and shares high sequence identity with many clinically relevant bacterial pathogens. Thus, it would serve as an ideal system for use in the structure-guided optimization of new antibacterial agents.


Assuntos
Aliivibrio fischeri/enzimologia , Alquil e Aril Transferases/química , Fosfomicina/química , Domínios e Motivos de Interação entre Proteínas , Uridina Difosfato N-Acetilglicosamina/química , Alquil e Aril Transferases/metabolismo , Fosfomicina/metabolismo , Modelos Moleculares , Especificidade por Substrato , Uridina Difosfato N-Acetilglicosamina/metabolismo
12.
J Biol Chem ; 287(19): 15803-9, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22442144

RESUMO

Most bacteria use the 2-C-methyl-D-erythritol 4-phosphate (MEP) pathway for the synthesis of their essential isoprenoid precursors. The absence of the MEP pathway in humans makes it a promising new target for the development of much needed new and safe antimicrobial drugs. However, bacteria show a remarkable metabolic plasticity for isoprenoid production. For example, the NADPH-dependent production of MEP from 1-deoxy-D-xylulose 5-phosphate in the first committed step of the MEP pathway is catalyzed by 1-deoxy-D-xylulose-5-phosphate reductoisomerase (DXR) in most bacteria, whereas an unrelated DXR-like (DRL) protein was recently found to catalyze the same reaction in some organisms, including the emerging human and animal pathogens Bartonella and Brucella. Here, we report the x-ray crystal structures of the Brucella abortus DRL enzyme in its apo form and in complex with the broad-spectrum antibiotic fosmidomycin solved to 1.5 and 1.8 Å resolution, respectively. DRL is a dimer, with each polypeptide folding into three distinct domains starting with the NADPH-binding domain, in resemblance to the structure of bacterial DXR enzymes. Other than that, DRL and DXR show a low structural relationship, with a different disposition of the domains and a topologically unrelated C-terminal domain. In particular, the active site of DRL presents a unique arrangement, suggesting that the design of drugs that would selectively inhibit DRL-harboring pathogens without affecting beneficial or innocuous bacteria harboring DXR should be feasible. As a proof of concept, we identified two strong DXR inhibitors that have virtually no effect on DRL activity.


Assuntos
Aldose-Cetose Isomerases/metabolismo , Proteínas de Bactérias/metabolismo , Brucella abortus/enzimologia , Complexos Multienzimáticos/metabolismo , Oxirredutases/metabolismo , Terpenos/metabolismo , Aldose-Cetose Isomerases/química , Aldose-Cetose Isomerases/genética , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação/genética , Biocatálise/efeitos dos fármacos , Brucella abortus/genética , Brucella abortus/metabolismo , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Fosfomicina/análogos & derivados , Fosfomicina/química , Fosfomicina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multienzimáticos/química , Complexos Multienzimáticos/genética , Oxirredutases/química , Oxirredutases/genética , Ligação Proteica , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
13.
Biochemistry ; 51(4): 917-25, 2012 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22148590

RESUMO

Isopentenyl phosphate kinase (IPK) catalyzes the phosphorylation of isopentenyl phosphate to form the isoprenoid precursor isopentenyl diphosphate in the archaeal mevalonate pathway. This enzyme is highly homologous to fosfomycin kinase (FomA), an antibiotic resistance enzyme found in a few strains of Streptomyces and Pseudomonas whose mode of action is inactivation by phosphorylation. Superposition of Thermoplasma acidophilum (THA) IPK and FomA structures aligns their respective substrates and catalytic residues, including H50 and K14 in THA IPK and H58 and K18 in Streptomyces wedmorensis FomA. These residues are conserved only in the IPK and FomA members of the phosphate subdivision of the amino acid kinase family. We measured the fosfomycin kinase activity of THA IPK [K(m) = 15.1 ± 1.0 mM, and k(cat) = (4.0 ± 0.1) × 10⁻² s⁻¹], resulting in a catalytic efficiency (k(cat)/K(m) = 2.6 M⁻¹ s⁻¹) that is 5 orders of magnitude lower than that of the native reaction. Fosfomycin is a competitive inhibitor of IPK (K(i) = 3.6 ± 0.2 mM). Molecular dynamics simulation of the IPK·fosfomycin·MgATP complex identified two binding poses for fosfomycin in the IP binding site, one of which results in a complex analogous to the native IPK·IP·ATP complex that engages H50 and the lysine triangle formed by K5, K14, and K205. The other binding pose leads to a dead-end complex that engages K204 near the IP binding site to bind fosfomycin. Our findings suggest a mechanism for acquisition of FomA-based antibiotic resistance in fosfomycin-producing organisms.


Assuntos
Antibacterianos/metabolismo , Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Fosfomicina/metabolismo , Proteínas Quinases/química , Proteínas Quinases/metabolismo , Thermoplasma/enzimologia , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Antibacterianos/química , Antibacterianos/farmacologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Bases de Dados de Proteínas , Farmacorresistência Bacteriana/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fosfomicina/química , Fosfomicina/farmacologia , Cinética , Magnésio/química , Magnésio/metabolismo , Conformação Molecular , Simulação de Dinâmica Molecular , Fosforilação/efeitos dos fármacos , Fosfotransferases/química , Fosfotransferases/metabolismo , Estabilidade Proteica , Streptomyces/enzimologia , Homologia Estrutural de Proteína , Especificidade por Substrato
15.
Biochemistry ; 50(32): 6909-19, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21728358

RESUMO

We present here the crystal structures of fosfomycin resistance protein (FomA) complexed with MgATP, with ATP and fosfomycin, with MgADP and fosfomycin vanadate, with MgADP and the product of the enzymatic reaction, fosfomycin monophosphate, and with ADP at 1.87, 1.58, 1.85, 1.57, and 1.85 Å resolution, respectively. Structures of these complexes that approximate different reaction steps allowed us to distinguish the catalytically active conformation of ATP and to reconstruct the model of the MgATP·fosfomycin complex. According to the model, the triphosphate tail of the nucleotide is aligned toward the phosphonate moiety of fosfomycin, in contest to the previously published MgAMPPNP complex, with the attacking fosfomycin oxygen positioned 4 Å from the γ-phosphorus of ATP. Site-directed mutagenesis studies and comparison of these structures with that of homologous N-acetyl-l-glutamate and isopentenyl phosphate kinases allowed us to propose a model of phosphorylation of fosfomycin by FomA enzyme. A Mg cation ligates all three phosphate groups of ATP and together with positively charged K216, K9, K18, and H58 participates in the dissipation of negative charge during phosphoryl transfer, indicating that the transferred phosphate group is highly negatively charged, which would be expected for an associative mechanism. K216 polarizes the γ-phosphoryl group of ATP. K9, K18, and H58 participate in stabilization of the transition state. D150 and D208 play organizational roles in catalysis. S148, S149, and T210 participate in fosfomycin binding, with T210 being crucial for catalysis. Hence, it appears that as in the homologous enzymes, FomA-catalyzed phosphoryl transfer takes place by an in-line predominantly associative mechanism.


Assuntos
Fosfomicina/metabolismo , Proteínas Quinases/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Fosforilação , Conformação Proteica , Dobramento de Proteína , Proteínas Quinases/química
16.
J Am Chem Soc ; 133(29): 11262-9, 2011 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-21682308

RESUMO

Hydroxypropylphosphonic acid epoxidase (HppE) is an unusual mononuclear iron enzyme that uses dioxygen to catalyze the oxidative epoxidation of (S)-2-hydroxypropylphosphonic acid (S-HPP) in the biosynthesis of the antibiotic fosfomycin. Additionally, the enzyme converts the R-enantiomer of the substrate (R-HPP) to 2-oxo-propylphosphonic acid. To probe the mechanism of HppE regiospecificity, we determined three X-ray structures: R-HPP with inert cobalt-containing enzyme (Co(II)-HppE) at 2.1 Å resolution; R-HPP with active iron-containing enzyme (Fe(II)-HppE) at 3.0 Å resolution; and S-HPP-Fe(II)-HppE in complex with dioxygen mimic NO at 2.9 Å resolution. These structures, along with previously determined structures of S-HPP-HppE, identify the dioxygen binding site on iron and elegantly illustrate how HppE is able to recognize both substrate enantiomers to catalyze two completely distinct reactions.


Assuntos
Antibacterianos/metabolismo , Bactérias/enzimologia , Fosfomicina/metabolismo , Oxirredutases/química , Oxirredutases/metabolismo , Bactérias/química , Sítios de Ligação , Cobalto/química , Cobalto/metabolismo , Cristalografia por Raios X , Ferro/química , Ferro/metabolismo , Modelos Moleculares , Oxigênio/química , Oxigênio/metabolismo , Conformação Proteica , Estereoisomerismo , Especificidade por Substrato
17.
Proc Natl Acad Sci U S A ; 107(41): 17557-62, 2010 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-20876132

RESUMO

Phosphonate natural products possess a range of biological activities as a consequence of their ability to mimic phosphate esters or tetrahedral intermediates formed in enzymatic reactions involved in carboxyl group metabolism. The dianionic form of these compounds at pH 7 poses a drawback with respect to their ability to mimic carboxylates and tetrahedral intermediates. Microorganisms producing phosphonates have evolved two solutions to overcome this hurdle: biosynthesis of monoanionic phosphinates containing two P-C bonds or esterification of the phosphonate group. The latter solution was first discovered for the antibiotic dehydrophos that contains a methyl ester of a phosphonodehydroalanine group. We report here the expression, purification, substrate scope, and structure of the O-methyltransferase from the dehydrophos biosynthetic gene cluster. The enzyme utilizes S-adenosylmethionine to methylate a variety of phosphonates including 1-hydroxyethylphosphonate, 1,2-dihydroxyethylphosphonate, and acetyl-1-aminoethylphosphonate. Kinetic analysis showed that the best substrates are tripeptides containing as C-terminal residue a phosphonate analog of alanine suggesting the enzyme acts late in the biosynthesis of dehydrophos. These conclusions are corroborated by the X-ray structure that reveals an active site that can accommodate a tripeptide substrate. Furthermore, the structural studies demonstrate a conformational change brought about by substrate or product binding. Interestingly, the enzyme has low substrate specificity and was used to methylate the clinical antibiotic fosfomycin and the antimalaria clinical candidate fosmidomycin, showing its promise for applications in bioengineering.


Assuntos
Vias Biossintéticas/fisiologia , Metiltransferases/química , Metiltransferases/metabolismo , Organofosfonatos/metabolismo , Streptomyces/enzimologia , Bioengenharia , Cromatografia Líquida de Alta Pressão , Cromatografia por Troca Iônica , Cristalografia por Raios X , Dipeptídeos/biossíntese , Esterificação , Fosfomicina/análogos & derivados , Fosfomicina/metabolismo , Cinética , Espectrometria de Massas , Metilação , Estrutura Molecular , S-Adenosilmetionina/metabolismo , Especificidade por Substrato
18.
Biochemistry ; 48(49): 11715-23, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19899805

RESUMO

MurA (enolpyruvyl UDP-GlcNAc synthase) catalyzes the first committed step in peptidoglycan biosynthesis. In this study, MurA-catalyzed breakdown of its tetrahedral intermediate (THI), with a k(cat)/K(M) of 520 M(-1) s(-1), was far slower than the normal reaction, and 3 x 10(5)-fold slower than the homologous enzyme, AroA, reacting with its THI. This provided kinetic evidence of slow binding and a conformationally constrained active site. The MurA cocrystal structure with UDP-N-acetylmuramic acid (UDP-MurNAc), a potent inhibitor, and phosphite revealed a new "staged" MurA conformation in which the Arg397 side chain tracked phosphite out of the catalytic site. The closed-to-staged transition involved breaking eight MurA.ligand ion pairs, and three intraprotein hydrogen bonds helping hold the active site loop closed. These were replaced with only two MurA.UDP-MurNAc ion pairs, two with phosphite, and seven new intraprotein ion pairs or hydrogen bonds. Cys115 appears to have an important role in forming the staged conformation. The staged conformation appears to be one step in a complex choreography of release of the product from MurA.


Assuntos
Alquil e Aril Transferases/química , Alquil e Aril Transferases/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Alquil e Aril Transferases/antagonistas & inibidores , Alquil e Aril Transferases/fisiologia , Catálise , Domínio Catalítico , Cristalografia por Raios X , Cisteína/metabolismo , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/fisiologia , Fosfomicina/química , Fosfomicina/metabolismo , Cinética , Ligantes , Fosfoenolpiruvato/química , Fosfoenolpiruvato/metabolismo , Ligação Proteica , Conformação Proteica
19.
Methods Enzymol ; 401: 367-79, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16399398

RESUMO

Three similar but mechanistically distinct fosfomycin resistance proteins that catalyze the opening of the oxirane ring of the antibiotic are known. FosA is a Mn(II) and K(+)-dependent glutathione transferase. FosB is a Mg(2+)-dependent L-cysteine thiol transferase. FosX is a Mn(II)-dependent fosfomycin-specific epoxide hydrolase. The expression, purification, kinetic, and physical characteristics of six fosfomycin resistance proteins including the FosA proteins from transposon TN2921 and Pseudomonas aeruginosa, the FosB proteins from Bacillus subtilis and Staphylococcus aureus, and the FosX proteins from Mesorhizobium loti and Listeria monocytogenes are reported.


Assuntos
Bactérias/enzimologia , Proteínas de Bactérias/metabolismo , Glutationa Transferase/metabolismo , Metaloproteínas/metabolismo , Proteínas de Bactérias/genética , Resistência Microbiana a Medicamentos , Fosfomicina/metabolismo , Glutationa Transferase/genética , Hidrolases/genética , Hidrolases/metabolismo , Metaloproteínas/genética , Estrutura Molecular
20.
J Biol Chem ; 280(5): 3757-63, 2005 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-15531591

RESUMO

MurA (UDP-N-acetylglucosamine enolpyruvyl transferase, EC 2.5.1.7) is an essential enzyme in the biosynthesis of the peptidoglycan layer of the bacterial cell. It provides an attractive template for the design of novel antibiotic drugs and is the target of the naturally occurring antibiotic fosfomycin, which covalently attaches to Cys115 in the active site of the enzyme. Mutations of Cys115 to Asp exist in pathogens such as Mycobacteria or Chlamydia rendering these organisms resistant to fosfomycin. Thus, there is a need for the elucidation of the role of this cysteine in the MurA reaction. We determined the x-ray structure of the C115S mutant of Enterobacter cloacae MurA, which was crystallized in the presence of the substrates of MurA. The structure depicts the product state of the enzyme with enolpyruvyl-UDP-N-acetylglucosamine and inorganic phosphate trapped in the active site. Kinetic analysis revealed that the Cys-to-Ser mutation results in an enzyme that appears to perform a single turnover of the reaction. Opposing the common view of Cys115 as a key residue in the chemical reaction of enolpyruvyl transfer, we now conclude that the wild-type cysteine is essential for product release only. On the basis of a detailed comparison of the product state with the intermediate state and an unliganded state of MurA, we propose that dissociation of the products is an ordered event with inorganic phosphate leaving first. Phosphate departure appears to trigger a suite of conformational changes, which finally leads to opening of the two-domain structure of MurA and the release of the second product enolpyruvyl-UDP-N-acetylglucosamine.


Assuntos
Alquil e Aril Transferases/metabolismo , Antibacterianos/metabolismo , Enterobacter cloacae/metabolismo , Fosfomicina/metabolismo , Alquil e Aril Transferases/genética , Antibacterianos/química , Sítios de Ligação , Catálise , Cristalografia , Cistina/metabolismo , Enterobacter cloacae/genética , Fosfomicina/química , Mutação , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA