Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Chem Inf Model ; 63(20): 6354-6365, 2023 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-37791530

RESUMO

Due to the emergence of antibiotic resistance, the need to explore novel antibiotics and/or novel strategies to counter antibiotic resistance is of utmost importance. In this work, we explored the molecular and mechanistic details of the degradation of a streptogramin B antibiotic by virginiamycin B (Vgb) lyase of Staphylococcus aureus using classical molecular dynamics simulations and multiscale quantum mechanics/molecular mechanics methods. Our results were in line with available experimental kinetic information. Although we were able to identify a stepwise mechanism, in the wild-type enzyme, the intermediate is short-lived, showing a small barrier to decay to the product state. The impact of point mutations on the reaction was also assessed, showing not only the importance of active site residues to the reaction catalyzed by Vgb lyase but also of near positive and negative residues surrounding the active site. Using molecular dynamics simulations, we also predicted the most likely protonation state of the 3-hydroxypicolinic moiety of the antibiotic and the impact of mutants on antibiotic binding. All this information will expand our understanding of linearization reactions of cyclic antibiotics, which are crucial for the development of novel strategies that aim to tackle antibiotic resistance.


Assuntos
Liases , Virginiamicina , Virginiamicina/química , Virginiamicina/metabolismo , Simulação de Dinâmica Molecular , Liases/metabolismo , Antibacterianos/química , Catálise
2.
Acc Chem Res ; 54(8): 1891-1908, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33792282

RESUMO

Continued, rapid development of antimicrobial resistance has become worldwide health crisis and a burden on the global economy. Decisive and comprehensive action is required to slow down the spread of antibiotic resistance, including increased investment in antibiotic discovery, sustainable policies that provide returns on investment for newly launched antibiotics, and public education to reduce the overusage of antibiotics, especially in livestock and agriculture. Without significant changes in the current antibiotic pipeline, we are in danger of entering a post-antibiotic era.In this Account, we summarize our recent efforts to develop next-generation streptogramin and lankacidin antibiotics that overcome bacterial resistance by means of modular chemical synthesis. First, we describe our highly modular, scalable route to four natural group A streptogramins antibiotics in 6-8 steps from seven simple chemical building blocks. We next describe the application of this route to the synthesis of a novel library of streptogramin antibiotics informed by in vitro and in vivo biological evaluation and high-resolution cryo-electron microscopy. One lead compound showed excellent inhibitory activity in vitro and in vivo against a longstanding streptogramin-resistance mechanism, virginiamycin acetyltransferase. Our results demonstrate that the combination of rational design and modular chemical synthesis can revitalize classes of antibiotics that are limited by naturally arising resistance mechanisms.Second, we recount our modular approaches toward lankacidin antibiotics. Lankacidins are a group of polyketide natural products with activity against several strains of Gram-positive bacteria but have not been deployed as therapeutics due to their chemical instability. We describe a route to several diastereomers of 2,18-seco-lankacidinol B in a linear sequence of ≤8 steps from simple building blocks, resulting in a revision of the C4 stereochemistry. We next detail our modular synthesis of several diastereoisomers of iso-lankacidinol that resulted in the structural reassignment of this natural product. These structural revisions raise interesting questions about the biosynthetic origin of lankacidins, all of which possessed uniform stereochemistry prior to these findings. Finally, we summarize the ability of several iso- and seco-lankacidins to inhibit the growth of bacteria and to inhibit translation in vitro, providing important insights into structure-function relationships for the class.


Assuntos
Antibacterianos/síntese química , Macrolídeos/síntese química , Estreptograminas/síntese química , Antibacterianos/química , Antibacterianos/farmacologia , Farmacorresistência Bacteriana/efeitos dos fármacos , Escherichia coli/metabolismo , Bactérias Gram-Negativas , Bactérias Gram-Positivas/efeitos dos fármacos , Macrolídeos/química , Macrolídeos/farmacologia , Testes de Sensibilidade Microbiana , Conformação Molecular , Simulação de Dinâmica Molecular , Ribossomos/química , Ribossomos/metabolismo , Estreptograminas/química , Estreptograminas/farmacologia , Virginiamicina/análogos & derivados , Virginiamicina/síntese química , Virginiamicina/metabolismo , Virginiamicina/farmacologia
3.
J Asian Nat Prod Res ; 23(10): 992-1000, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32924591

RESUMO

One new virginiamycin derivative, 'beilunmycin' (1), and three known virginiamycin antibiotics, 16-hydroxy-virginiamycin M1 (2), virginiamycin M2 (3), and virginiamycin M1 (4), were isolated from the culture of a mangrove-derived endophytic Streptomyces sp. 2BBP-J2. The structures were characterized on the basis of their spectroscopic data, and the absolute configuration of 1 was established by ECD calculations. Compounds 1-4 exhibited antibacterial activities against Gram-positive bacteria, with MIC values in the range of 0.5-16 µg/ml. All the compounds demonstrated strong protein translation-stalling activity, with minimal concentrations detected with pDualrep2 in the range of 1.9-5.9 nmol.


Assuntos
Streptomyces , Antibacterianos/farmacologia , Testes de Sensibilidade Microbiana , Estrutura Molecular , Biossíntese de Proteínas , Streptomyces/metabolismo , Virginiamicina/metabolismo
4.
Nature ; 586(7827): 145-150, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32968273

RESUMO

Natural products serve as chemical blueprints for most antibiotics in clinical use. The evolutionary process by which these molecules arise is inherently accompanied by the co-evolution of resistance mechanisms that shorten the clinical lifetime of any given class of antibiotics1. Virginiamycin acetyltransferase (Vat) enzymes are resistance proteins that provide protection against streptogramins2, potent antibiotics against Gram-positive bacteria that inhibit the bacterial ribosome3. Owing to the challenge of selectively modifying the chemically complex, 23-membered macrocyclic scaffold of group A streptogramins, analogues that overcome the resistance conferred by Vat enzymes have not been previously developed2. Here we report the design, synthesis, and antibacterial evaluation of group A streptogramin antibiotics with extensive structural variability. Using cryo-electron microscopy and forcefield-based refinement, we characterize the binding of eight analogues to the bacterial ribosome at high resolution, revealing binding interactions that extend into the peptidyl tRNA-binding site and towards synergistic binders that occupy the nascent peptide exit tunnel. One of these analogues has excellent activity against several streptogramin-resistant strains of Staphylococcus aureus, exhibits decreased rates of acetylation in vitro, and is effective at lowering bacterial load in a mouse model of infection. Our results demonstrate that the combination of rational design and modular chemical synthesis can revitalize classes of antibiotics that are limited by naturally arising resistance mechanisms.


Assuntos
Antibacterianos/síntese química , Antibacterianos/farmacologia , Desenho de Fármacos , Farmacorresistência Bacteriana/efeitos dos fármacos , Estreptogramina Grupo A/síntese química , Estreptogramina Grupo A/farmacologia , Acetilação/efeitos dos fármacos , Acetiltransferases/genética , Acetiltransferases/metabolismo , Animais , Antibacterianos/classificação , Carga Bacteriana/efeitos dos fármacos , Sítios de Ligação , Microscopia Crioeletrônica , Feminino , Técnicas In Vitro , Camundongos , Testes de Sensibilidade Microbiana , Modelos Moleculares , RNA de Transferência/metabolismo , Ribossomos/efeitos dos fármacos , Ribossomos/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Estreptogramina Grupo A/química , Estreptogramina Grupo A/classificação , Virginiamicina/análogos & derivados , Virginiamicina/química , Virginiamicina/metabolismo
5.
Sci Rep ; 8(1): 3592, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29483631

RESUMO

Although dietary antibiotic growth promoters have long been used to increase growth performance in commercial food animal production, the biochemical details associated with these effects remain poorly defined. A metabolomics approach was used to characterize and identify the biochemical compounds present in the intestine of broiler chickens fed a standard, unsupplemented diet or a diet supplemented with the antibiotic growth promoters, virginiamycin or bacitracin methylene disalicylate. Compared with unsupplemented controls, the levels of 218 biochemicals were altered (156 increased, 62 decreased) in chickens given the virginiamycin-supplemented diet, while 119 were altered (96 increased, 23 decreased) with the bacitracin-supplemented diet. When compared between antibiotic-supplemented groups, 79 chemicals were altered (43 increased, 36 decreased) in virginiamycin- vs. bacitracin-supplemented chickens. The changes in the levels of intestinal biochemicals provided a distinctive biochemical signature unique to each antibiotic-supplemented group. These biochemical signatures were characterized by increases in the levels of metabolites of amino acids (e.g. 5-hydroxylysine, 2-aminoadipate, 5-hydroxyindoleaceate, 7-hydroxyindole sulfate), fatty acids (e.g. oleate/vaccenate, eicosapentaenoate, 16-hydroxypalmitate, stearate), nucleosides (e.g. inosine, N6-methyladenosine), and vitamins (e.g. nicotinamide). These results provide the framework for future studies to identify natural chemical compounds to improve poultry growth performance without the use of in-feed antibiotics.


Assuntos
Antibacterianos/metabolismo , Bacitracina/metabolismo , Galinhas/crescimento & desenvolvimento , Intestinos/fisiologia , Metaboloma/fisiologia , Salicilatos/metabolismo , Virginiamicina/metabolismo , Aminoácidos/metabolismo , Análise de Variância , Ração Animal/análise , Animais , Antibacterianos/farmacologia , Bacitracina/farmacologia , Suplementos Nutricionais , Ácidos Graxos/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Niacinamida/metabolismo , Nucleosídeos/metabolismo , Salicilatos/farmacologia , Virginiamicina/farmacologia
6.
Biotechnol Lett ; 40(1): 157-163, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29038924

RESUMO

OBJECTIVE: To test the inactivation of the antibiotic, virginiamycin, by laccase-induced culture supernatants of Aureobasidium pullulans. RESULTS: Fourteen strains of A. pullulans from phylogenetic clade 7 were tested for laccase production. Three laccase-producing strains from this group and three previously identified strains from clade 5 were compared for inactivation of virginiamycin. Laccase-induced culture supernatants from clade 7 strains were more effective at inactivation of virginiamycin, particularly at 50 °C. Clade 7 strain NRRL Y-2567 inactivated 6 µg virginiamycin/ml within 24 h. HPLC analyses indicated that virginiamycin was degraded by A. pullulans. CONCLUSIONS: A. pullulans has the potential for the bioremediation of virginiamycin-contaminated materials, such as distiller's dry grains with solubles (DDGS) animal feed produced from corn-based fuel ethanol production.


Assuntos
Antibacterianos/metabolismo , Ascomicetos/metabolismo , Glucanos/metabolismo , Virginiamicina/metabolismo , Ascomicetos/crescimento & desenvolvimento , Biotransformação , Cromatografia Líquida de Alta Pressão , Meios de Cultura , Temperatura Alta
7.
World J Microbiol Biotechnol ; 32(5): 76, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27038946

RESUMO

Antibiotics are frequently used to prevent and treat bacterial contamination of commercial fuel ethanol fermentations, but there is concern that antibiotic residues may persist in the distillers grains coproducts. A study to evaluate the fate of virginiamycin during the ethanol production process was conducted in the pilot plant facilities at the National Corn to Ethanol Research Center, Edwardsville, IL. Three 15,000-liter fermentor runs were performed: one with no antibiotic (F1), one dosed with 2 parts per million (ppm) of a commercial virginiamycin product (F2), and one dosed at 20 ppm of virginiamycin product (F3). Fermentor samples, distillers dried grains with solubles (DDGS), and process intermediates (whole stillage, thin stillage, syrup, and wet cake) were collected from each run and analyzed for virginiamycin M and virginiamycin S using a liquid chromatography-mass spectrometry method. Virginiamycin M was detected in all process intermediates of the F3 run. On a dry-weight basis, virginiamycin M concentrations decreased approximately 97 %, from 41 µg/g in the fermentor to 1.4 µg/g in the DDGS. Using a disc plate bioassay, antibiotic activity was detected in DDGS from both the F2 and F3 runs, with values of 0.69 µg virginiamycin equivalent/g sample and 8.9 µg/g, respectively. No antibiotic activity (<0.6 µg/g) was detected in any of the F1 samples or in the fermentor and process intermediate samples from the F2 run. These results demonstrate that low concentrations of biologically active antibiotic may persist in distillers grains coproducts produced from fermentations treated with virginiamycin.


Assuntos
Antibacterianos/metabolismo , Biocombustíveis/análise , Etanol/metabolismo , Saccharomyces cerevisiae/metabolismo , Virginiamicina/metabolismo , Zea mays/metabolismo , Antibacterianos/análise , Cromatografia Líquida , Etanol/análise , Fermentação , Espectrometria de Massas , Virginiamicina/análise , Zea mays/química
8.
J Am Chem Soc ; 138(12): 4155-67, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-26982529

RESUMO

Modular polyketide synthases (PKSs) direct the biosynthesis of clinically valuable secondary metabolites in bacteria. The fidelity of chain growth depends on specific recognition between successive subunits in each assembly line: interactions mediated by C- and N-terminal "docking domains" (DDs). We have identified a new family of DDs in trans-acyl transferase PKSs, exemplified by a matched pair from the virginiamycin (Vir) system. In the absence of C-terminal partner (VirA (C)DD) or a downstream catalytic domain, the N-terminal DD (VirFG (N)DD) exhibits multiple characteristics of an intrinsically disordered protein. Fusion of the two docking domains results in a stable fold for VirFG (N)DD and an overall protein-protein complex of unique topology whose structure we support by site-directed mutagenesis. Furthermore, using small-angle X-ray scattering (SAXS), the positions of the flanking acyl carrier protein and ketosynthase domains have been identified, allowing modeling of the complete intersubunit interface.


Assuntos
Aciltransferases/metabolismo , Policetídeo Sintases/metabolismo , Virginiamicina/química , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Virginiamicina/metabolismo
9.
J Anim Sci ; 92(10): 4566-76, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25184842

RESUMO

The intent of this study was to establish a fecal sampling procedure for the indicator method (IM) to provide digestibility values similar to those obtained by the total collection (TC) method. A total of 24 pigs (52.6 ± 1.5 kg) were fed 1 of 4 diets with a 2 × 2 factorial arrangement of virginiamycin and phytase (PHY) added to a corn-soybean meal diet with no inorganic P supplement. Pigs were housed in metabolism crates for a 5-d TC period after 7 d of adaptation. Immediately after the TC, a fecal collection period followed, using the IM by including 0.25% of Cr2O3 in the feed for 10 d. Fecal collection for the IM started the day after diets containing Cr2O3 were first fed, and continued for 9 consecutive days with a single grab sample per day. Similar portions of feces from d 5 to 9 were also composited into 4 samples to evaluate multi-day pooling combinations. Highly variable means and CV among samples for apparent total tract digestibility (ATTD) were observed at d 1 and 2 using the IM. The mean ATTD for DM, GE, and nutrients appeared to be stabilized by d 5 or 6 in all dietary treatments. The TC data seemed to have lower CV than the IM data for many components. Based on the linear broken-line analysis, fecal Cr concentration plateaued at d 3.75 (P < 0.001) after the first feeding of Cr. Mean ATTD values by the IM were lower than those by the TC method for DM (P < 0.05), GE (P < 0.01), P (P < 0.01), and Ca (P < 0.001). The PHY supplementation improved ATTD of P (P < 0.001) and Ca (P < 0.001) in both collection methods, whereas the PHY effect on ATTD of DM was observed only for the IM (P < 0.05). Differences related to PHY effect on ATTD were detected from d 4 to 9 in a single grab sample for P and DM but the ATTD of DM had inconsistent P-values by day. Fecal sampling after 4 d of initial feeding of marker always allowed detection of treatment effects on ATTD of P but not on ATTD of DM. Results indicated that the IM results in lower digestibility values than the TC method and does not provide the same treatment difference as the TC digestibility for energy and nutrients that are not highly impacted by the dietary treatment. For the IM, ATTD values and fecal Cr concentration stabilize at least on d 5 after initial feeding of diets containing Cr2O3. At least 2-d pooling of feces for the IM appears to be needed to provide greater accuracy and lower variations than a single grab sample.


Assuntos
Ração Animal/análise , Coleta de Dados/métodos , Dieta/veterinária , Digestão/fisiologia , Sus scrofa/fisiologia , 6-Fitase/metabolismo , 6-Fitase/farmacologia , Animais , Compostos de Cromo/metabolismo , Suplementos Nutricionais , Fezes/química , Suínos , Virginiamicina/metabolismo , Virginiamicina/farmacologia
10.
Lett Appl Microbiol ; 57(5): 393-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23815812

RESUMO

A newly reduced macrocyclic lactone antibiotic streptogramin A, 5,6-dihydrovirginiamycin M1 was created by feeding virginiamycin M1 into a culture of recombinant Streptomyces venezuelae. Its chemical structure was spectroscopically elucidated, and this streptogramin A analogue showed twofold higher antibacterial activities against methicillin-resistant Staphylococcus aureus (MRSA) compared with its parent molecule virginiamycin M1. Docking studies using the model of streptogramin A acetyltransferase (VatA) suggested that the newly generated analogue binds tighter with overall lower free energy compared with the parent molecule virginiamycin M1. This hypothesis was validated experimentally through the improvement of efficacy of the new analogue against MRSA strains. The biotransformation approach presented herein could have a broad application in the production of reduced macrocyclic molecules.


Assuntos
Antibacterianos/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Estreptogramina A/análogos & derivados , Estreptogramina A/biossíntese , Estreptogramina A/química , Estreptogramina A/farmacologia , Virginiamicina/metabolismo
11.
Proc Natl Acad Sci U S A ; 104(25): 10388-93, 2007 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-17563376

RESUMO

The streptogramin combination therapy of quinupristin-dalfopristin (Synercid) is used to treat infections caused by bacterial pathogens, such as methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus faecium. However, the effectiveness of this therapy is being compromised because of an increased incidence of streptogramin resistance. One of the clinically observed mechanisms of resistance is enzymatic inactivation of the type B streptogramins, such as quinupristin, by a streptogramin B lyase, i.e., virginiamycin B lyase (Vgb). The enzyme catalyzes the linearization of the cyclic antibiotic via a cleavage that requires a divalent metal ion. Here, we present crystal structures of Vgb from S. aureus in its apoenzyme form and in complex with quinupristin and Mg2+ at 1.65- and 2.8-A resolution, respectively. The fold of the enzyme is that of a seven-bladed beta-propeller, although the sequence reveals no similarity to other known members of this structural family. Quinupristin binds to a large depression on the surface of the enzyme, where it predominantly forms van der Waals interactions. Validated by site-directed mutagenesis studies, a reaction mechanism is proposed in which the initial abstraction of a proton is facilitated by a Mg2+ -linked conjugated system. Analysis of the Vgb-quinupristin structure and comparison with the complex between quinupristin and its natural target, the 50S ribosomal subunit, reveals features that can be exploited for developing streptogramins that are impervious to Vgb-mediated resistance.


Assuntos
Antibacterianos/farmacologia , Liases/química , Staphylococcus aureus/efeitos dos fármacos , Estreptogramina B/farmacologia , Virginiamicina/análogos & derivados , Antibacterianos/metabolismo , Apoenzimas/química , Sítios de Ligação , Catálise , Cristalografia por Raios X , Resistência Microbiana a Medicamentos , Cinética , Liases/metabolismo , Modelos Químicos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Estreptogramina B/metabolismo , Virginiamicina/metabolismo , Difração de Raios X
12.
Science ; 311(5759): 374-7, 2006 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-16424339

RESUMO

Microbial resistance to antibiotics currently spans all known classes of natural and synthetic compounds. It has not only hindered our treatment of infections but also dramatically reshaped drug discovery, yet its origins have not been systematically studied. Soil-dwelling bacteria produce and encounter a myriad of antibiotics, evolving corresponding sensing and evading strategies. They are a reservoir of resistance determinants that can be mobilized into the microbial community. Study of this reservoir could provide an early warning system for future clinically relevant antibiotic resistance mechanisms.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla , Microbiologia do Solo , Streptomyces/efeitos dos fármacos , Substituição de Aminoácidos , Antibacterianos/metabolismo , Ciprofloxacina/farmacologia , Daptomicina/metabolismo , Daptomicina/farmacologia , Farmacorresistência Bacteriana Múltipla/genética , Eritromicina/metabolismo , Eritromicina/farmacologia , Genes Bacterianos , Cetolídeos/metabolismo , Cetolídeos/farmacologia , Macrolídeos/farmacologia , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Mutação , Rifampina/metabolismo , Rifampina/farmacologia , Streptomyces/enzimologia , Streptomyces/genética , Streptomyces/isolamento & purificação , Resistência a Trimetoprima , Resistência a Vancomicina/genética , Virginiamicina/metabolismo , Virginiamicina/farmacologia
13.
Cell ; 121(2): 257-70, 2005 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-15851032

RESUMO

Crystal structures of H. marismortui large ribosomal subunits containing the mutation G2099A (A2058 in E. coli) with erythromycin, azithromycin, clindamycin, virginiamycin S, and telithromycin bound explain why eubacterial ribosomes containing the mutation A2058G are resistant to them. Azithromycin binds almost identically to both G2099A and wild-type subunits, but the erythromycin affinity increases by more than 10(4)-fold, implying that desolvation of the N2 of G2099 accounts for the low wild-type affinity for macrolides. All macrolides bind similarly to the H. marismortui subunit, but their binding differs significantly from what has been reported in the D. radioidurans subunit. The synergy in the binding of streptogramins A and B appears to result from a reorientation of the base of A2103 (A2062, E. coli) that stacks between them. The structure of large subunit containing a three residue deletion mutant of L22 shows a change in the L22 structure and exit tunnel shape that illuminates its macrolide resistance phenotype.


Assuntos
Antibacterianos/química , Farmacorresistência Bacteriana/fisiologia , Eritromicina/química , Haloarcula marismortui/química , Ribossomos/química , Antibacterianos/metabolismo , Azitromicina/química , Azitromicina/metabolismo , Sítios de Ligação , Clindamicina/química , Clindamicina/metabolismo , Cristalografia , Eritromicina/metabolismo , Escherichia coli/química , Escherichia coli/genética , Escherichia coli/metabolismo , Haloarcula marismortui/genética , Haloarcula marismortui/metabolismo , Cetolídeos/química , Cetolídeos/metabolismo , Mutação , Ligação Proteica , RNA Ribossômico 23S/química , RNA Ribossômico 23S/genética , RNA Ribossômico 23S/metabolismo , Ribossomos/genética , Ribossomos/metabolismo , Virginiamicina/química , Virginiamicina/metabolismo
14.
BMC Biol ; 2: 4, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15059283

RESUMO

BACKGROUND: The bacterial ribosome is a primary target of several classes of antibiotics. Investigation of the structure of the ribosomal subunits in complex with different antibiotics can reveal the mode of inhibition of ribosomal protein synthesis. Analysis of the interactions between antibiotics and the ribosome permits investigation of the specific effect of modifications leading to antimicrobial resistances. Streptogramins are unique among the ribosome-targeting antibiotics because they consist of two components, streptogramins A and B, which act synergistically. Each compound alone exhibits a weak bacteriostatic activity, whereas the combination can act bactericidal. The streptogramins A display a prolonged activity that even persists after removal of the drug. However, the mode of activity of the streptogramins has not yet been fully elucidated, despite a plethora of biochemical and structural data. RESULTS: The investigation of the crystal structure of the 50S ribosomal subunit from Deinococcus radiodurans in complex with the clinically relevant streptogramins quinupristin and dalfopristin reveals their unique inhibitory mechanism. Quinupristin, a streptogramin B compound, binds in the ribosomal exit tunnel in a similar manner and position as the macrolides, suggesting a similar inhibitory mechanism, namely blockage of the ribosomal tunnel. Dalfopristin, the corresponding streptogramin A compound, binds close to quinupristin directly within the peptidyl transferase centre affecting both A- and P-site occupation by tRNA molecules. CONCLUSIONS: The crystal structure indicates that the synergistic effect derives from direct interaction between both compounds and shared contacts with a single nucleotide, A2062. Upon binding of the streptogramins, the peptidyl transferase centre undergoes a significant conformational transition, which leads to a stable, non-productive orientation of the universally conserved U2585. Mutations of this rRNA base are known to yield dominant lethal phenotypes. It seems, therefore, plausible to conclude that the conformational change within the peptidyl transferase centre is mainly responsible for the bactericidal activity of the streptogramins and the post-antibiotic inhibition of protein synthesis.


Assuntos
Antibacterianos/farmacologia , Peptidil Transferases/metabolismo , Ribossomos/efeitos dos fármacos , Virginiamicina/análogos & derivados , Antibacterianos/metabolismo , Sítios de Ligação/efeitos dos fármacos , Cristalização , Deinococcus/efeitos dos fármacos , Deinococcus/enzimologia , Sinergismo Farmacológico , Peptidil Transferases/química , RNA Ribossômico 23S/química , RNA Ribossômico 23S/metabolismo , Proteínas Ribossômicas/química , Proteínas Ribossômicas/metabolismo , Ribossomos/química , Ribossomos/enzimologia , Virginiamicina/metabolismo , Virginiamicina/farmacologia
15.
Gene ; 286(2): 283-90, 2002 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-11943483

RESUMO

Streptomyces virginiae produces -butyrolactone autoregulators (virginiae butanolide, VB), which control the biosynthesis of virginiamycin M1 and S. A 6.3-kb region downstream of the virginiamycin S (VS)-resistance operon in S. virginiae was sequenced, and four plausible open reading frames (ORFs) (visA, 1,260 bp; visB, 1,656 bp; visC, 888 bp; visD, 1209 bp) were identified. Homology analysis revealed significant similarities with enzymes involved in the biosynthesis of cyclopeptolide antibiotics: VisA (53% identity, 65% similarity) to -lysine 2-aminotransferase (NikC) of nikkomycin D biosynthesis, VisB (66% identity, 72% similarity) to 3-hydroxypicolinic acid:AMP ligase of pristinamycin I biosynthesis, VisC (48% identity, 59% similarity) to lysine cyclodeaminase of ascomycin biosynthesis, and VisD (43% identity, 56% similarity) to erythromycin C-22 hydroxylase of erythromycin biosynthesis. Northern blotting as well as high-resolution S1 analysis of the ORFs revealed that they were transcribed as two bicistronic transcripts, namely 3.0-kb visB-visA and another 2.7-kb visC-visD transcript, with promoters locating upstream of visB and visC, respectively. Transcription of the two operons was observed only 1 h after the VB production, which was 2 h before the virginiamycin production. Furthermore, prompt induction of the transcription was observed as a result of external VB addition, suggesting that the expression of the two operons was under the control of VB.


Assuntos
Proteínas de Bactérias/genética , Genes Bacterianos/genética , Streptomyces/genética , Virginiamicina/biossíntese , Amônia-Liases/genética , Amônia-Liases/metabolismo , Proteínas de Bactérias/metabolismo , Sequência de Bases , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , DNA Bacteriano/química , DNA Bacteriano/genética , Ordem dos Genes , Ligases/genética , Ligases/metabolismo , Dados de Sequência Molecular , Análise de Sequência de DNA , Streptomyces/metabolismo , Transaminases/genética , Transaminases/metabolismo , Sítio de Iniciação de Transcrição , Transcrição Gênica , Virginiamicina/análogos & derivados , Virginiamicina/metabolismo
16.
Int J Artif Organs ; 25(1): 33-9, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11853069

RESUMO

BACKGROUND: Quinupristin-dalfopristin (Q/D) is often utilized in critically ill patients, some of whom require CVVH. This study was undertaken to determine the clearance of O/D and their main active metabolites (RPR 100391, RP 69012, RP 12536) via CVVH in the swine model. METHODS: Q/D 7.5 mg/kg was intravenously administered over 0.5 h to 12 swine after induction of acute renal failure by ligation of the renal arteries. At 0.5 h post injection, the CVVH procedure was initiated and continued for 8 hours at the following pump rates: (1)100 mL/min, (2)180 rnL/min, and (3)100 mL/min with dialysis (flow rate: 1 L/h). Blood and ultrafiltrate samples were collected at 1 h intervals and assessed by a validated HPLC method. RESULTS: Plasma analysis suggests rapid metabolism to the main active metabolites which are appreciably cleared as demonstrated by high clearance and sieving coefficient estimates. Mean clearance estimates for RP 69012, RP 100391, and RP 12536 are 729, 777, and 578 mL/h in the 100 mL/min CVVH group, 772, 785, 685 mL/min in the 180 mL/min CVVH group, and 753, 791, 616 mL/min in the 100 mL/min CVVH group with 1 L/h dialysis, respectively. CONCLUSION: These data reveal that Q/D is rapidly metabolized and the metabolites are cleared to a large extent via CVVH. Due to the considerable contribution of the metabolites to overall in vivo activities, additional studies are required to fully quantify their removal before final dosage modifications for patients undergoing CVVH can be recommended.


Assuntos
Quimioterapia Combinada/farmacocinética , Hemodiafiltração/métodos , Hemofiltração/métodos , Virginiamicina/farmacocinética , Animais , Quimioterapia Combinada/metabolismo , Modelos Animais , Suínos , Virginiamicina/metabolismo
17.
Biochemistry ; 40(30): 8877-86, 2001 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-11467949

RESUMO

The streptogramin antibiotics were identified almost 50 years ago but have only recently found clinical use as a consequence of the increase in multidrug-resistant bacteria. Despite the fact that these antibiotics have historically not found intense clinical use, resistance to streptogramins exists. Streptogramins consist of a mixture of two components: cyclic polyunsaturated macrolactones (group A) and cyclic hexadepsipeptides (group B). The latter are cyclized through an ester bond between the hydroxyl group of an N-terminal threonine and the C-terminal carboxyl. Resistance to the B streptogramins can occur through the production of enzymes such as Vgb from Staphylococcus aureus. This enzyme had been assumed to be a lactonase that inactivates the cyclic antibiotic by linearization through hydrolytic cleavage of the ester bond. We have expressed recombinant Vgb in quantity and, using a combination of mass spectrometry, NMR, and synthesis of model depsipeptides, show unequivocally that streptogramin B inactivation does not involve hydrolysis of the ester bond. Rather, the hexadepsipeptide is linearized through an elimination reaction across the ester bond generating an N-terminal dehydrobutyrine group. Therefore, Vgb is not a hydrolase but a lyase. We also have explored the activity of Vgb orthologues present in the chromosomes of various bacteria including Bordetella pertussis and Streptomyces coelicolor and have determined that these enzymes also show streptogramin B inactivation through an elimination mechanism indistinguishable to that used by Vgb. These results demonstrate that Vgb is a member of a large group of streptogramin B lyases that are present not only in resistant clinical isolates but also in the chromosomes of many bacteria. There is therefore a significant reservoir of streptogramin resistance enzymes in the environment, which has the potential to impact the long-term utility of these antibiotics. This research establishing the molecular mechanism of streptogramin resistance therefore has the potential to be exploited in the discovery of inhibitory compounds that could rescue antibiotic activity even in the presence of resistance elements.


Assuntos
Antibacterianos/antagonistas & inibidores , Antibacterianos/metabolismo , Proteínas de Bactérias/fisiologia , Hemeproteínas/fisiologia , Staphylococcus aureus/fisiologia , Virginiamicina/análogos & derivados , Virginiamicina/antagonistas & inibidores , Virginiamicina/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bordetella pertussis/enzimologia , Bordetella pertussis/genética , Catálise , Cátions Bivalentes/química , Resistência Microbiana a Medicamentos , Hemeproteínas/química , Hemeproteínas/genética , Hemeproteínas/metabolismo , Hidrólise , Metais/química , Conformação Molecular , Ressonância Magnética Nuclear Biomolecular , Peptídeos Cíclicos/antagonistas & inibidores , Peptídeos Cíclicos/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética , Streptomyces/enzimologia , Streptomyces/genética , Especificidade por Substrato
18.
Antimicrob Agents Chemother ; 45(1): 319-23, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11120988

RESUMO

Streptococcus pneumoniae clinical isolate BM4455 was resistant to 16-membered macrolides and to streptogramins. This unusual resistance phenotype was due to an A(2062)C (Escherichia coli numbering) mutation in domain V of the four copies of 23S rRNA.


Assuntos
Antibacterianos/farmacologia , RNA Ribossômico 23S/genética , Streptococcus pneumoniae/efeitos dos fármacos , Streptococcus pneumoniae/genética , Virginiamicina/farmacologia , Antibacterianos/metabolismo , Resistência Microbiana a Medicamentos , Macrolídeos , Testes de Sensibilidade Microbiana , Mutação/genética , Oligonucleotídeos/farmacologia , Fenótipo , Virginiamicina/metabolismo
19.
Nat Biotechnol ; 18(11): 1203-8, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11062442

RESUMO

Here we describe repressible (PipOFF) as well as inducible (PipON) systems for regulated gene expression in mammalian cells, based on the repressor Pip (pristinamycin-induced protein), which is encoded by the streptogramin resistance operon of Streptomyces coelicolor. Expression of genes placed under control of these systems was responsive to clinically approved antibiotics belonging to the streptogramin group (pristinamycin, virginiamycin, and Synercid). The versatility of these systems was demonstrated by streptogramin-regulated expression of mouse erythropoietin (EPO), human placental secreted alkaline phosphatase (SEAP), or green fluorescent protein (GFP) in diverse cell lines (BHK, CHO, HeLa, and mouse myoblasts). Analysis of isogenic constructs in CHO cells demonstrated the PipOFF system gave lower background and higher induction ratios than the widely used tetracycline-repressible (TetOFF) expression systems. The streptogramin-based expression technology was functionally compatible with the TetOFF system, thus enabling the selective use of different antibiotics to independently control two different gene activities in the same cell.


Assuntos
Antibacterianos/metabolismo , Regulação da Expressão Gênica , Técnicas Genéticas , Virginiamicina/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Antibacterianos/farmacologia , Sítios de Ligação , Células CHO , Linhagem Celular , Cricetinae , Relação Dose-Resposta a Droga , Eritropoetina/metabolismo , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Proteínas Luminescentes/metabolismo , Camundongos , Microscopia de Fluorescência , Modelos Genéticos , Músculos/citologia , Músculos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Streptomyces/genética , Fatores de Tempo , Transfecção , Virginiamicina/farmacologia
20.
Curr Opin Investig Drugs ; 1(2): 173-80, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11249570

RESUMO

Rhône-Poulenc Rorer (RPR) developed Synercid (RP-59500), an injectable synergistic combination of quinupristin and dalfopristin as a treatment for a variety of infections caused by Gram-positive anaerobic bacteria. The treatment was approved in the UK in July 1999, for use in patients with nosocomial pneumonia, skin and soft tissue infections and clinically significant infections due to Enterococcus faecium when there is no other active antibacterial agent [337556,335257]. It was launched in the UK and the US in September 1999 [342899]. In December 1999, Synercid successfully completed the Mutual Recognition Procedure in the EU under Aventis Pharma for use in patients with these infections [351525]. In September 2000, Merrill Lynch predicted first-year sales in 1999 of Euro 15 million, rising to Euro 171 million in 2004 [384874]. In January 1999, BT Alex Brown predicted sales of US $88 million in 1999 rising to US $450 million in 2002 [318220]. In April 1999, ABN Amro predicted annual sales of DM 30 million in 1999, rising to DM 150 million in 2002 [328676].


Assuntos
Infecções Bacterianas/tratamento farmacológico , Quimioterapia Combinada/uso terapêutico , Virginiamicina/uso terapêutico , Animais , Infecções Bacterianas/microbiologia , Ensaios Clínicos como Assunto , Contraindicações , Quimioterapia Combinada/efeitos adversos , Quimioterapia Combinada/síntese química , Quimioterapia Combinada/metabolismo , Quimioterapia Combinada/farmacologia , Quimioterapia Combinada/toxicidade , Humanos , Relação Estrutura-Atividade , Virginiamicina/efeitos adversos , Virginiamicina/síntese química , Virginiamicina/metabolismo , Virginiamicina/farmacologia , Virginiamicina/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...