Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
1.
Methods Enzymol ; 669: 45-70, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35644180

RESUMEN

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.


Asunto(s)
Fosfomicina , Streptomyces , Fosfomicina/química , Fosfomicina/metabolismo , Metiltransferasas/metabolismo , S-Adenosilmetionina/metabolismo , Streptomyces/metabolismo , Vitamina B 12/metabolismo
2.
Environ Microbiol ; 24(9): 4466-4488, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35688634

RESUMEN

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.


Asunto(s)
Fosfomicina , Listeria monocytogenes , Acetamidas , Antibacterianos/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , ADN/metabolismo , Fosfatos de Dinucleósidos/metabolismo , Fosfomicina/metabolismo , Fosfomicina/farmacología , Humanos , Isoleucina/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Oligopéptidos/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Liasas de Fósforo-Oxígeno/genética
3.
Microbiology (Reading) ; 168(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35358034

RESUMEN

Burkholderia cenocepacia infections are difficult to treat and there is an urgent need for alternative (combination) treatments. The use of anti-virulence therapies in combination with antibiotics is a possible strategy to increase the antimicrobial susceptibility of the pathogen and to slow down the development of resistance. In the present study we evaluated the ß-lactam and colistin-potentiating activity, and anti-virulence effect of the non-mevalonate pathway inhibitor FR900098 against B. cenocepacia in various in vitro and in vivo models. In addition, we evaluated whether repeated exposure to FR900098 alone or when combined with ceftazidime leads to increased resistance. FR900098 potentiated the activity of colistin and several ß-lactam antibiotics (aztreonam, cefepime, cefotaxime, ceftazidime, mecillinam and piperacillin) but not of imipenem and meropenem. When used alone or in combination with ceftazidime, FR900098 increased the survival of infected Galleria mellonella and Caenorhabditis elegans. Furthermore, combining ceftazidime with FR900098 resulted in a significant inhibition of the biofilm formation of B. cenocepacia. Repeated exposure to FR900098 in the C. elegans infection model did not lead to decreased activity, and the susceptibility of the evolved B. cenocepacia HI2424 lineages to ceftazidime, FR900098 and the combination of both remained unchanged. In conclusion, FR900098 reduces B. cenocepacia virulence and potentiates ceftazidime in an in vivo C. elegans model, and this activity is not lost during the experimental evolution experiment carried out in the present study.


Asunto(s)
Burkholderia cenocepacia , Fosfomicina , Animales , Burkholderia cenocepacia/genética , Burkholderia cenocepacia/metabolismo , Caenorhabditis elegans , Fosfomicina/análogos & derivados , Fosfomicina/metabolismo , Fosfomicina/farmacología , Virulencia
4.
J Phys Chem B ; 126(7): 1388-1403, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35138863

RESUMEN

Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen responsible for many nosocomial infections. It is quite resistant to various antibiotics, caused by the absence of general diffusion pores in the outer membrane. Instead, it contains many substrate-specific channels. Among them are the two phosphate- and pyrophosphate-specific porins OprP and OprO. Phosphonic acid antibiotics such as fosfomycin and fosmidomycin seem to be good candidates for using these channels to enter P. aeruginosa bacteria. Here, we investigated the permeation of fosfomycin through OprP and OprO using electrophysiology and molecular dynamics (MD) simulations. The results were compared to those of the fosmidomycin translocation, for which additional MD simulations were performed. In the electrophysiological approach, we noticed a higher binding affinity of fosfomycin than of fosmidomycin to OprP and OprO. In MD simulations, the ladder of arginine residues and the cluster of lysine residues play an important role in the permeation of fosfomycin through the OprP and OprO channels. Molecular details on the permeation of fosfomycin through OprP and OprO channels were derived from MD simulations and compared to those of fosmidomycin translocation. In summary, this study demonstrates that the selectivity of membrane channels can be employed to improve the permeation of antibiotics into Gram-negative bacteria and especially into resistant P. aeruginosa strains.


Asunto(s)
Fosfomicina , Pseudomonas aeruginosa , Antibacterianos/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/química , Fosfomicina/metabolismo , Fosfatos/metabolismo , Porinas/química , Pseudomonas aeruginosa/química
5.
Mol Microbiol ; 116(1): 97-108, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33561903

RESUMEN

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.


Asunto(s)
Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Fosfomicina/análogos & derivados , Transporte Iónico/fisiología , Porinas/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/genética , Sitios de Unión/fisiología , Cloruros/metabolismo , Farmacorresistencia Bacteriana Múltiple/genética , Fosfomicina/metabolismo , Ácidos Fosforosos/metabolismo , Porinas/genética , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética
6.
J Chem Theory Comput ; 17(1): 549-559, 2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33378186

RESUMEN

To reach their target site inside Gram-negative bacteria, almost all antibiotics need to cross the outer membrane. Computational modeling of such processes can be numerically demanding due to the size of the systems and especially due to the timescales involved. Recently, a hybrid Brownian and molecular dynamics approach, i.e., Brownian dynamics including explicit atoms (BRODEA), has been developed and evaluated for studying the transport of monoatomic ions through membrane channels. Later on, this numerically efficient scheme has been applied to determine the free energy surfaces of the ciprofloxacin and enrofloxacin translocation through the porin OmpC using temperature-accelerated simulations. To improve the usability and accuracy of the approach, schemes to approximate the position-dependent diffusion constant of the molecule while traversing the pore had to be established. To this end, we have studied the translocation of the charged phosphonic acid antibiotic fosfomycin through the porin OmpF from Escherichia coli devising and benchmarking several diffusion models. To test the efficiency and sensitivity of these models, the effect of OmpF mutations on the permeation of fosfomycin was analyzed. Permeation events have been recorded over millisecond-long biased and unbiased simulations, from which thermodynamics and kinetics quantities of the translocation processes were determined. As a result, the use of the BRODEA approach, together with the appropriate diffusion model, was seen to accurately reproduce the findings observed in electrophysiology experiments and all-atom molecular dynamics simulations. These results suggest that the BRODEA approach can become a valuable tool for screening numerous compounds to evaluate their outer membrane permeability, a property important in the development of new antibiotics.


Asunto(s)
Antibacterianos/farmacocinética , Escherichia coli/metabolismo , Fosfomicina/farmacocinética , Porinas/metabolismo , Antibacterianos/metabolismo , Permeabilidad de la Membrana Celular , Difusión , Escherichia coli/química , Escherichia coli/genética , Infecciones por Escherichia coli/microbiología , Fosfomicina/metabolismo , Humanos , Cinética , Modelos Moleculares , Mutación , Porinas/química , Porinas/genética , Conformación Proteica , Termodinámica
7.
J Comput Aided Mol Des ; 33(10): 927-940, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31654265

RESUMEN

Proteins of the independent mevalonate pathway for isoprenoid biosynthesis are important targets for the development of new antibacterial compounds as this pathway is present in most pathogenic organisms such as Mycobacterium tuberculosis, DPlasmodium falciparum and Escherichia coli, but is not present in mammalian species, including humans. Deoxy-D-xylulose 5-phosphate reductoisomerase (DXR) is an important target in this pathway and the most effective DXR inhibitor to date is fosmidomycin, which is used to treat malaria and, more recently, tuberculosis. Recently, Armstrong C. M. et al. showed that a mutant of DXR, S222T, induces a loss of the fosmidomycin inhibition efficiency, even though the bacteria culture is still viable and able to produce isoprenoids. As this represents a potential fosmidomycin-resistant mutation, it is important to understand the mechanism of this apparent mutation-induced resistance to fosmidomycin. Here, we used molecular dynamics simulations and Molecular Mechanics/Poisson Boltzmann Surface Area analysis to understand the structural and energetic basis of the resistance. Our results suggest that the point mutation results in changes to the structural dynamics of an active site loop that probably protects the active site and facilitates enzymatic reaction. From the simulation analysis, we also showed that the mutation results in changes in the interaction energy profiles in a way that can explain the observed activity of the mutant protein toward the natural inhibitor deoxy-D-xylulose 5-phosphate. These results should be taken into consideration in future efforts to develop new therapeutic antibiotic compounds that target DXR.


Asunto(s)
Isomerasas Aldosa-Cetosa/antagonistas & inhibidores , Isomerasas Aldosa-Cetosa/metabolismo , Farmacorresistencia Microbiana , Escherichia coli/enzimología , Fosfomicina/análogos & derivados , Simulación de Dinámica Molecular , Mutación , Isomerasas Aldosa-Cetosa/genética , Antibacterianos/administración & dosificación , Antibacterianos/metabolismo , Sitios de Unión , Escherichia coli/efectos de los fármacos , Fosfomicina/administración & dosificación , Fosfomicina/metabolismo , Ligandos , Modelos Teóricos , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Pentosafosfatos/metabolismo , Conformación Proteica
8.
Biophys J ; 116(2): 258-269, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30616836

RESUMEN

Fosfomycin is a frequently prescribed drug in the treatment of acute urinary tract infections. It enters the bacterial cytoplasm and inhibits the biosynthesis of peptidoglycans by targeting the MurA enzyme. Despite extensive pharmacological studies and clinical use, the permeability of fosfomycin across the bacterial outer membrane is largely unexplored. Here, we investigate the fosfomycin permeability across the outer membrane of Gram-negative bacteria by electrophysiology experiments as well as by all-atom molecular dynamics simulations including free-energy and applied-field techniques. Notably, in an electrophysiological zero-current assay as well as in the molecular simulations, we found that fosfomycin can rapidly permeate the abundant Escherichia coli porin OmpF. Furthermore, two triple mutants in the constriction region of the porin have been investigated. The permeation rates through these mutants are slightly lower than that of the wild type but fosfomycin can still permeate. Altogether, this work unravels molecular details of fosfomycin permeation through the outer membrane porin OmpF of E. coli and moreover provides hints for understanding the translocation of phosphonic acid antibiotics through other outer membrane pores.


Asunto(s)
Antibacterianos/metabolismo , Fosfomicina/química , Simulación de Dinámica Molecular , Porinas/química , Antibacterianos/química , Transporte Biológico , Fosfomicina/metabolismo , Cinética , Porinas/metabolismo
9.
ACS Synth Biol ; 7(12): 2824-2832, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30462485

RESUMEN

Sequence to activity mapping technologies are rapidly developing, enabling the generation and isolation of mutations conferring novel phenotypes. Here we used the CRISPR enabled trackable genome engineering (CREATE) technology to investigate the inhibition of the essential ispC gene in its native genomic context in Escherichia coli. We created a full saturation library of 33 sites proximal to the ligand binding pocket and challenged this library with the antimalarial drug fosmidomycin, which targets the ispC gene product, DXR. This selection is especially challenging since it is relatively weak in E. coli, with multiple naturally occurring pathways for resistance. We identified several previously unreported mutations that confer fosmidomycin resistance, in highly conserved sites that also exist in pathogens including the malaria-inducing Plasmodium falciparum. This approach may have implications for the isolation of resistance-conferring mutations and may affect the design of future generations of fosmidomycin-based drugs.


Asunto(s)
Isomerasas Aldosa-Cetosa/genética , Antimaláricos/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Fosfomicina/análogos & derivados , Isomerasas Aldosa-Cetosa/metabolismo , Antimaláricos/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Escherichia coli/química , Escherichia coli/metabolismo , Fosfomicina/metabolismo , Fosfomicina/farmacología , Ingeniería Genética/métodos , Mutación , Plásmidos/genética , Plásmidos/metabolismo , Plasmodium falciparum/efectos de los fármacos
10.
Artículo en Inglés | MEDLINE | ID: mdl-29891612

RESUMEN

Fosfomycin inhibits MurA following uptake by the GlpT transporter of glycerol-3-phosphate in Escherichia coli In Staphylococcus aureus, plasmid overexpression of the Tet38 efflux pump and a glpT mutant resulted in increased MICs and decreased accumulation of fosfomycin, with MICs affected by glycerol-3-phosphate. In contrast, a tet38 mutant had a lower MIC and increased accumulation of fosfomycin, suggesting that Tet38 acts as an efflux transporter of fosfomycin.


Asunto(s)
Fosfomicina/metabolismo , Fosfomicina/farmacología , Staphylococcus aureus/efectos de los fármacos , Farmacorresistencia Bacteriana , Pruebas de Sensibilidad Microbiana , Staphylococcus aureus/metabolismo
11.
J Vet Pharmacol Ther ; 41(3): 415-427, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29333612

RESUMEN

Fosfomycin is a broad-spectrum bactericidal antibiotic widely used in pig farms for the treatment of a wide variety of bacterial infections. In this study, the elimination of disodium fosfomycin in colostrum/milk of the sow and the impact of this antibiotic on the microbiota and intestinal morpho-physiology of suckling piglets were analyzed. The average amount of fosfomycin eliminated in colostrum (after administration of 15 mg/kg IM) during the first 10 hr postpartum was 0.85 µg/ml, and the mean residual amount ingested by the piglets was 0.26 mg/kg. The elimination profile of fosfomycin concentrations in colostrum occurs at a time of profound changes in the morpho-physiology of the gastrointestinal tract of the piglet. However, the studied concentrations did not produce imbalances on the microbiota or on the morpho-physiology of the gastrointestinal tract of the piglet. Concentrations of fosfomycin were maintained in the mammary gland above the MIC for more than 8 hr for pathogenic bacteria of productive importance. This would indicate that fosfomycin may be considered safe for the specific treatment of bacterial infectious processes in sows during the peri- and postpartum period. This first study with disodium fosfomycin stimulates awareness in the proper use of antimicrobials at farrowing.


Asunto(s)
Antibacterianos/farmacocinética , Calostro/química , Fosfomicina/farmacocinética , Porcinos/metabolismo , Alimentación Animal , Animales , Animales Recién Nacidos , Animales Lactantes , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Residuos de Medicamentos , Femenino , Fosfomicina/química , Fosfomicina/metabolismo , Fosfomicina/farmacología , Pruebas de Sensibilidad Microbiana , Embarazo , Porcinos/microbiología
13.
Emerg Infect Dis ; 23(9): 1564-1567, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28820368
14.
Biochemistry ; 56(28): 3519-3522, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28678474

RESUMEN

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.


Asunto(s)
Fosfomicina/metabolismo , Metiltransferasas/metabolismo , Organofosfonatos/metabolismo , Streptomyces/enzimología , Vitamina B 12/análogos & derivados , Vías Biosintéticas , Citidina Monofosfato/metabolismo , Metilación , S-Adenosilmetionina/metabolismo , Streptomyces/metabolismo , Especificidad por Sustrato , Vitamina B 12/metabolismo
15.
Org Biomol Chem ; 15(15): 3276-3285, 2017 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-28352915

RESUMEN

Three functionalised propylphosphonic acids were synthesised to study C-P bond cleavage in R. huakuii PMY1. (R)-1-Hydroxy-2-oxopropylphosphonic acid [(R)-5] was prepared by chiral resolution of (±)-dimethyl 1-hydroxy-2-methylallyllphosphonate [(±)-12], followed by ozonolysis and deprotection. The N-(l-alanyl)-substituted (1R,2R)-2-amino-1-hydroxypropylphosphonic acid 10, a potential precursor for 2-oxopropylphosphonic acid (5) in cells, was obtained by coupling the aminophosphonic acid with benzotriazole-activated Z-l-alanine and hydrogenolytic deprotection. (1R*,2R*)-1,2-Dihydroxy-3,3,3-trifluoropropylphosphonic acid, a potential inhibitor of C-P bond cleavage after conversion into its 2-oxo derivative in the cell, was accessed from trifluoroacetaldehyde hydrate via hydroxypropanenitrile 21, which was silylated and reduced to the aldehyde (±)-23. Diastereoselective addition of diethyl trimethylsilyl phosphite furnished diastereomeric α-siloxyphosphonates. The less polar one was converted to the desired racemic phosphonic acid (±)-(1R*,2R*)-9 as its ammonium salt.


Asunto(s)
Fosfomicina/metabolismo , Fosfomicina/química , Hidrólisis , Ácidos Fosforosos/química
16.
Int J Biol Macromol ; 97: 106-114, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28064057

RESUMEN

Peptidoglycan (PG) is the key component of the bacterial cell wall. The enzyme UDP-N-Acetylglucosamine Enolpyruvyl Transferase (MurA) catalyzes the transfer of enolpyruvate from phosphoenolpyruvate (PEP) to uridinediphospho-N-acetylglucosamine (UNAG), which is the first committed step of PG biosynthesis. Here, we present the biochemical and structural features of the MurA enzyme of the opportunistic pathogen Acinetobacter baumannii (AbMurA). The recombinant AbMurA exists as a monomer in solution and shows optimal activity at pH 7.5 and 37°C. The Km for UDP-N-acetylglucosamine was 1.062±0.09mM and for PEP was 1.806±0.23mM. The relative enzymatic activity was inhibited ∼3 fold in the presence of 50mM fosfomycin (FFQ). Superimposition of the AbMurA model with E. coli demonstrated key structural similarity in the FFQ-binding site. AbMurA also has a surface loop that contains the active site Cys116 that interact with FFQ. Sequence analysis indicates the presence of the five conserved amino acids, i.e., K22, C116, D306, D370 and L371, required for the functional activity like other MurA enzymes from different bacteria. MurA enzymes are indispensable for cell integrity and their lack of counterparts in eukaryotes suggests them to be a promising drug target.


Asunto(s)
Acinetobacter baumannii/enzimología , Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/metabolismo , Transferasas Alquil y Aril/antagonistas & inhibidores , Secuencia de Aminoácidos , Dominio Catalítico , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Fosfomicina/metabolismo , Fosfomicina/farmacología , Concentración de Iones de Hidrógeno , Cinética , Simulación del Acoplamiento Molecular , Filogenia , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Temperatura
17.
J Antimicrob Chemother ; 72(1): 68-74, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27650185

RESUMEN

OBJECTIVES: To decipher the function of A1S_1331, named AbaF (Acinetobacter baumannii Fosfomycin efflux), one of the primary targets of AbsR25, a small RNA of A. baumannii. METHODS: abaF was cloned in a multicopy plasmid and expressed from its native promoter in an efflux-deficient strain-Escherichia coli KAM32. Drug susceptibility, accumulation and efflux of ethidium bromide (EtBr) were determined in this strain. abaF was disrupted in A. baumannii using homologous recombination and its effect on drug susceptibility, biofilm formation and virulence was studied. Expression of abaF was followed by quantitative PCR in fosfomycin-challenged A. baumannii and fosfomycin-resistant mutants of A. baumannii. Expression of abaF in clinical strains of A. baumannii was determined by RT-PCR. RESULTS: Expression of abaF in E. coli KAM32 resulted in increased resistance to fosfomycin. Lower accumulation and higher efflux of EtBr from this strain confirmed the role of AbaF as an efflux pump. Disruption of abaF in A. baumannii caused an increase in fosfomycin susceptibility and a decrease in biofilm formation and virulence. The expression of abaF was higher in A. baumannii cells exposed to fosfomycin and in cells resistant to higher concentrations of fosfomycin. The clinically relevant strains of A. baumannii also tested positive for the expression of abaF. CONCLUSIONS: The results of this study suggest that efflux is an important mechanism of fosfomycin resistance and AbaF is involved in fosfomycin resistance in A. baumannii. AbaF also seems to play a role in biofilm formation and virulence of A. baumannii.


Asunto(s)
Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/metabolismo , Antibacterianos/metabolismo , Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Fosfomicina/metabolismo , Fosfomicina/farmacología , Acinetobacter baumannii/crecimiento & desarrollo , Acinetobacter baumannii/patogenicidad , Biopelículas/crecimiento & desarrollo , Transporte Biológico Activo , Clonación Molecular , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Genes Bacterianos , Pruebas de Sensibilidad Microbiana , Reacción en Cadena en Tiempo Real de la Polimerasa , Virulencia
18.
Antimicrob Agents Chemother ; 60(4): 2281-91, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26833150

RESUMEN

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.


Asunto(s)
Antibacterianos/farmacología , Escherichia coli/efectos de los fármacos , Metaboloma/efectos de los fármacos , Metabolómica , Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/genética , Aciltransferasas/metabolismo , Adenosina/metabolismo , Adenosina/farmacología , Antibacterianos/metabolismo , Carbonil Cianuro m-Clorofenil Hidrazona/metabolismo , Carbonil Cianuro m-Clorofenil Hidrazona/farmacología , Ceftazidima/metabolismo , Ceftazidima/farmacología , Pared Celular/química , Pared Celular/efectos de los fármacos , Pared Celular/metabolismo , Cromatografía Liquida , ADN Bacteriano/antagonistas & inhibidores , ADN Bacteriano/biosíntesis , Escherichia coli/genética , Escherichia coli/metabolismo , Fosfomicina/análogos & derivados , Fosfomicina/metabolismo , Fosfomicina/farmacología , Expresión Génica , Células HEK293 , Humanos , Ácidos Hidroxámicos/metabolismo , Ácidos Hidroxámicos/farmacología , Espectrometría de Masas , Nucleósido-Fosfato Quinasa/antagonistas & inhibidores , Nucleósido-Fosfato Quinasa/genética , Nucleósido-Fosfato Quinasa/metabolismo , Pirimidinas/metabolismo , Pirimidinas/farmacología , Terpenos/antagonistas & inhibidores , Terpenos/metabolismo , Treonina/análogos & derivados , Treonina/metabolismo , Treonina/farmacología , Triclosán/metabolismo , Triclosán/farmacología
19.
Biochem Pharmacol ; 102: 45-63, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26707799

RESUMEN

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.


Asunto(s)
Actinas/metabolismo , Quinasas Lim/antagonistas & inhibidores , Quinasas Lim/metabolismo , Miosina Tipo II/metabolismo , Triterpenos/farmacología , Quinasas Asociadas a rho/metabolismo , Actinas/química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/aislamiento & purificación , Inhibidores Enzimáticos/farmacología , Fosfomicina/química , Fosfomicina/metabolismo , Células HeLa , Humanos , Miosina Tipo II/química , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Semillas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Triterpenos/química , Triterpenos/aislamiento & purificación , Quinasas Asociadas a rho/química
20.
Bioanalysis ; 7(19): 2585-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26470832

RESUMEN

BACKGROUND: Fosfomycin is an antibiotic of considerable interest for the treatment of infection by multidrug-resistant bacteria. Translating microsampling techniques into clinical PK studies may provide effective dosing information to improve patient outcomes and minimize the potential development of resistance. RESULTS: Accuracy and precision results were within ±15%; the method was validated across the range of 5-2000 µg/ml of fosfomycin using volumetric absorptive microsampling (VAMS) devices. CONCLUSION: The VAMS techniques provide acceptable validation data as assessed for lower limit of quantification, linearity, intra- and interday precision and accuracy, selectivity and matrix effects. Results from the recovery and stability studies suggest challenges remain for the analysis of fosfomycin in whole blood using VAMS.


Asunto(s)
Antibacterianos/sangre , Análisis Químico de la Sangre/métodos , Fosfomicina/sangre , Espectrometría de Masas en Tándem , Antibacterianos/metabolismo , Antibacterianos/normas , Cromatografía Líquida de Alta Presión , Pruebas con Sangre Seca , Fosfomicina/metabolismo , Fosfomicina/normas , Semivida , Humanos , Control de Calidad , Espectrometría de Masas en Tándem/normas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...