Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 67(2): e0087122, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36719223

RESUMO

Ampicillin-ceftriaxone has become a first-line therapy for Enterococcus faecalis endocarditis. We characterized the penicillin-binding protein (PBP) profiles of various E. faecalis strains and tested for synergy to better inform beta-lactam options for the treatment of E. faecalis infections. We assessed the affinity of PBP2B from elevated-MIC strain E. faecalis LS4828 compared to type strain JH2-2 using the fluorescent beta-lactam Bocillin FL. We also characterized pbp4 and pbpA structures and PBP4 and PBP2B expression and used deletion and complementation studies to assess the impact of PBP2B on the levels of resistance. We tested penicillin-susceptible and -resistant E. faecalis isolates against ceftriaxone or ceftaroline combinations with other beta-lactams in 24-h time-kill studies. Two penicillin-susceptible strains (JH2-2 and L2052) had identical pbp sequences and similar PBP expression levels. One reduced-penicillin-susceptibility strain (L2068) had pbp sequences identical to those of the susceptible strains but expressed more PBP4. The second decreased-penicillin-susceptibility strain (LS4828) had amino acid substitutions in both PBP4 and PBP2B and expressed increased quantities of both proteins. PBP2B did not appear to contribute significantly to the elevated beta-lactam MICs. No synergy was demonstrable against the strains with both mutated PBPs and increased expression (L2068 and LS4828). Meropenem plus ceftriaxone or ertapenem plus ceftriaxone demonstrated the most consistent synergistic activity. PBP2B of strain LS4828 does not contribute significantly to reduced penicillin susceptibility. Neither the MIC nor the level of PBP expression correlated directly with the identified synergistic combinations when tested at static subinhibitory concentrations.


Assuntos
Enterococcus faecalis , beta-Lactamas , Proteínas de Ligação às Penicilinas/genética , Proteínas de Ligação às Penicilinas/metabolismo , beta-Lactamas/farmacologia , beta-Lactamas/metabolismo , Enterococcus faecalis/genética , Enterococcus faecalis/metabolismo , Ceftriaxona/farmacologia , Penicilinas/farmacologia , Penicilinas/metabolismo , Testes de Sensibilidade Microbiana , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Antibacterianos/farmacologia , Antibacterianos/metabolismo
2.
Clin Infect Dis ; 75(9): 1661-1664, 2022 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-35653393

RESUMO

During the past 4 decades, oral vancomycin has been a mainstay of Clostridioides difficile infection (CDI) therapy with no reports of treatment failure due to emergence of vancomycin resistance. However, C. difficile isolates with high-level phenotypic resistance to vancomycin have recently been reported in 3 distinct geographic regions. There is an urgent need for surveillance to determine if strains with reduced vancomycin susceptibility are circulating in other areas. In a Cleveland-area hospital, screening of 176 CDI stool specimens yielded no C. difficile isolates with reduced vancomycin susceptibility and highlighted the potential for false-positive results due to contamination with vancomycin-resistant enterococci. Additional studies are needed to clarify whether reduced vancomycin susceptibility is an emerging problem that will alter clinical practice. Clinicians should alert their health department if they observe a substantial increase in the frequency of vancomycin treatment failure in patients diagnosed with CDI with no alternative explanation for diarrhea.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Humanos , Vancomicina/farmacologia , Vancomicina/uso terapêutico , Clostridioides , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/epidemiologia
3.
Antimicrob Agents Chemother ; 66(10): e0042622, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36154173

RESUMO

The standard of care for serious Enterococcus faecalis infections is ampicillin plus ceftriaxone. Ampicillin's inconvenient dosing schedule, drug instability, allergy potential, along with ceftriaxone's high risk for Clostridioides difficile infection and its promotion of vancomycin-resistant enterococci (VRE), led our team to explore alternative options. This work aimed to understand the role of carbapenems in combination with cephalosporins in these infections. We selected two ampicillin and penicillin susceptible E. faecalis strains (AMP-MIC 0.5-2 µg/mL; PCN-MIC 2 µg/mL) and simulated human therapeutic dosing regimens in a 48-h in vitro pharmacodynamic model (IVPD) with ampicillin (2g q4h), ertapenem (1g q24h), meropenem (2g q8h), ceftriaxone (2g q12h), and ceftaroline (600 mg q8h). As expected, ampicillin plus ceftriaxone demonstrated enhanced activity compared with ampicillin monotherapy with no MIC increases in either isolate. Meropenem and ceftaroline demonstrated significant kill against both isolates, with no regrowth or MIC increases occurring. Meropenem plus ceftriaxone also demonstrated significant kill, and while no MIC increases were identified for meropenem, there was minor regrowth and larger standard deviations. Ertapenem combined with either ceftriaxone or ceftaroline enhanced activity at 24 h, but at 48 h, regrowth occurred, and ertapenem MIC increases were noted. Meropenem-based combination therapy against E. faecalis may provide clinicians with another regimen to treat severe E. faecalis infections. Meropenem plus ceftaroline was as active as the standard of care treatment (ampicillin plus ceftriaxone) and may serve as an alternative for serious E. faecalis infections. Further studies are warranted to determine the clinical efficacy.


Assuntos
Ceftriaxona , Enterococcus faecalis , Humanos , Monofosfato de Adenosina , Ampicilina/farmacologia , Antibacterianos/farmacologia , Ceftriaxona/farmacologia , Cefalosporinas/farmacologia , Sinergismo Farmacológico , Ertapenem , Meropeném/farmacologia , Testes de Sensibilidade Microbiana , Ceftarolina
4.
Artigo em Inglês | MEDLINE | ID: mdl-33361289

RESUMO

Since its inaugural issue nearly half a century ago, Antimicrobial Agents and Chemotherapy has served as a premier source for reports on scientific and clinical advances in the field of antimicrobial chemotherapy. As a follow-up to the previous "History of Antimicrobial Agents and Chemotherapy from 1972 to 1998" written by George A. Jacoby (Antimicrob Agents Chemother 43:999-1002, 1999, https://doi.org/10.1128/AAC.43.5.999), we herein highlight the further evolution of this comprehensive and authoritative journal in response to changing science, demographics, and information technology.


Assuntos
Anti-Infecciosos , Antibacterianos , História do Século XX
5.
Clin Microbiol Rev ; 32(2)2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30700430

RESUMO

The genus Enterococcus comprises a ubiquitous group of Gram-positive bacteria that are of great relevance to human health for their role as major causative agents of health care-associated infections. The enterococci are resilient and versatile species able to survive under harsh conditions, making them well adapted to the health care environment. Two species cause the majority of enterococcal infections: Enterococcus faecalis and Enterococcus faecium Both species demonstrate intrinsic resistance to common antibiotics, such as virtually all cephalosporins, aminoglycosides, clindamycin, and trimethoprim-sulfamethoxazole. Additionally, a remarkably plastic genome allows these two species to readily acquire resistance to further antibiotics, such as high-level aminoglycoside resistance, high-level ampicillin resistance, and vancomycin resistance, either through mutation or by horizontal transfer of genetic elements conferring resistance determinants.


Assuntos
Farmacorresistência Bacteriana Múltipla , Enterococcus/crescimento & desenvolvimento , Infecções por Bactérias Gram-Positivas/microbiologia , Antibacterianos/farmacologia , Enterococcus/efeitos dos fármacos , Enterococcus/genética , Transferência Genética Horizontal , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Humanos , Testes de Sensibilidade Microbiana , Mutação
8.
Clin Infect Dis ; 69(12): 2045-2056, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30861061

RESUMO

BACKGROUND: ZTI-01 (fosfomycin for injection) is an epoxide antibiotic with a differentiated mechanism of action (MOA) inhibiting an early step in bacterial cell wall synthesis. ZTI-01 has broad in vitro spectrum of activity, including multidrug-resistant Gram-negative pathogens, and is being developed for treatment of complicated urinary tract infection (cUTI) and acute pyelonephritis (AP) in the United States. METHODS: Hospitalized adults with suspected or microbiologically confirmed cUTI/AP were randomized 1:1 to 6 g ZTI-01 q8h or 4.5 g intravenous (IV) piperacillin-tazobactam (PIP-TAZ) q8h for a fixed 7-day course (no oral switch); patients with concomitant bacteremia could receive up to 14 days. RESULTS: Of 465 randomized patients, 233 and 231 were treated with ZTI-01 and PIP-TAZ, respectively. In the microbiologic modified intent-to-treat (m-MITT) population, ZTI-01 met the primary objective of noninferiority compared with PIP-TAZ with overall success rates of 64.7% (119/184 patients) vs 54.5% (97/178 patients), respectively; treatment difference was 10.2% (95% confidence interval [CI]: -0.4, 20.8). Clinical cure rates at test of cure (TOC, day 19-21) were high and similar between treatments (90.8% [167/184] vs 91.6% [163/178], respectively). In post hoc analysis using unique pathogens typed by pulsed-field gel electrophoresis, overall success rates at TOC in m-MITT were 69.0% (127/184) for ZTI-01 versus 57.3% (102/178) for PIP-TAZ (difference 11.7% 95% CI: 1.3, 22.1). ZTI-01 was well tolerated. Most treatment-emergent adverse events, including hypokalemia and elevated serum aminotransferases, were mild and transient. CONCLUSIONS: ZTI-01 was effective for treatment of cUTI including AP and offers a new IV therapeutic option with a differentiated MOA for patients with serious Gram-negative infections. CLINICAL TRIAL REGISTRATION: NCT02753946.


Assuntos
Antibacterianos/administração & dosagem , Fosfomicina/administração & dosagem , Combinação Piperacilina e Tazobactam/uso terapêutico , Pielonefrite/tratamento farmacológico , Infecções Urinárias/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Carga Bacteriana , Farmacorresistência Bacteriana , Feminino , Humanos , Injeções , Masculino , Testes de Sensibilidade Microbiana , Pessoa de Meia-Idade , Combinação Piperacilina e Tazobactam/administração & dosagem , Combinação Piperacilina e Tazobactam/efeitos adversos , Pielonefrite/etiologia , Resultado do Tratamento , Infecções Urinárias/etiologia , Adulto Jovem
9.
Clin Infect Dis ; 67(2): 303-309, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29390132

RESUMO

Enterococci, one of the most common causes of hospital-associated infections, are responsible for substantial morbidity and mortality. Enterococcus faecalis, the more common and virulent species, causes serious high-inoculum infections, namely infective endocarditis, that are associated with cardiac surgery and mortality rates that remained unchanged for the last 30 years. The best cures for these infections are observed with combination antibiotic therapy; however, optimal treatment has not been fully elucidated. It is the purpose of this review to highlight treatment options and their limitations, and provide direction for future investigative efforts to aid in the treatment of these severe infections. While ampicillin plus ceftriaxone has emerged as a preferred treatment option, mortality rates continue to be high, and from a safety standpoint, ceftriaxone, unlike other cephalosporins, promotes colonization with vancomycin resistant-enterococci due to high biliary concentrations. More research is needed to improve patient outcomes from this high-mortality disease.


Assuntos
Antibacterianos/uso terapêutico , Bacteriemia/tratamento farmacológico , Endocardite Bacteriana/tratamento farmacológico , Enterococcus faecalis/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Ampicilina/uso terapêutico , Ceftriaxona/uso terapêutico , Cefalosporinas/uso terapêutico , Ensaios Clínicos como Assunto , Sinergismo Farmacológico , Quimioterapia Combinada , Infecções por Bactérias Gram-Positivas/complicações , Humanos , Testes de Sensibilidade Microbiana , Enterococos Resistentes à Vancomicina/efeitos dos fármacos
10.
Emerg Infect Dis ; 23(11): 1902-1904, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29048285

RESUMO

Of 890 vancomycin-resistant Enterococcus faecium isolates obtained by rectal screening from patients in Pittsburgh, Pennsylvania, USA, 4 had MICs >1,024 µg/mL for fosfomycin. These isolates had a Cys119Asp substitution in the active site of UDP-N-acetylglucosamine enolpyruvyl transferase. This substitution increased the fosfomycin MIC >4-fold and rendered this drug inactive in biochemical assays.


Assuntos
Alquil e Aril Transferases/genética , Antibacterianos/farmacologia , Enterococcus faecium/enzimologia , Fosfomicina/farmacologia , Infecções por Bactérias Gram-Positivas/microbiologia , Substituição de Aminoácidos , Proteínas de Bactérias/genética , Farmacorresistência Bacteriana , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/genética , Humanos , Testes de Sensibilidade Microbiana , Mutação , Pennsylvania , Vancomicina/farmacologia
11.
Antimicrob Agents Chemother ; 60(5): 3178-82, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26926624

RESUMO

Ampicillin-ceftriaxone combination therapy has become a predominant treatment for serious Enterococcus faecalis infections, such as endocarditis. Unfortunately, ceftriaxone use is associated with future vancomycin-resistant enterococcus colonization. We evaluated E. faecalis in an in vitro pharmacodynamic model against simulated human concentration-time profiles of ampicillin plus ceftaroline, cefepime, ceftriaxone, or gentamicin. Ampicillin-cefepime and ampicillin-ceftaroline demonstrated activities similar to those of ampicillin-ceftriaxone against E. faecalis.


Assuntos
Ampicilina/farmacologia , Antibacterianos/farmacologia , Ceftriaxona/farmacologia , Cefalosporinas/farmacologia , Enterococcus faecalis/efeitos dos fármacos , Cefepima , Gentamicinas/farmacologia , Testes de Sensibilidade Microbiana , Ceftarolina
12.
Antimicrob Agents Chemother ; 60(10): 5777-86, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27431230

RESUMO

The transfer of DNA between Enterococcus faecium strains has been characterized both by the movement of well-defined genetic elements and by the large-scale transfer of genomic DNA fragments. In this work, we report on the whole-genome analysis of transconjugants resulting from mating events between the vancomycin-resistant E. faecium C68 strain and the vancomycin-susceptible D344RRF strain to discern the mechanism by which the transferred regions enter the recipient chromosome. Vancomycin-resistant transconjugants from five independent matings were analyzed by whole-genome sequencing. In all cases but one, the penicillin binding protein 5 (pbp5) gene and the Tn5382 vancomycin resistance transposon were transferred together and replaced the corresponding pbp5 region of D344RRF. In one instance, Tn5382 inserted independently downstream of the D344RRF pbp5 gene. Single nucleotide variant (SNV) analysis suggested that entry of donor DNA into the recipient chromosome occurred by recombination across regions of homology between donor and recipient chromosomes, rather than through insertion sequence-mediated transposition. The transfer of genomic DNA was also associated with the transfer of C68 plasmid pLRM23 and another putative plasmid. Our data are consistent with the initiation of transfer by cointegration of a transferable plasmid with the donor chromosome, with subsequent circularization of the plasmid-chromosome cointegrant in the donor prior to transfer. Entry into the recipient chromosome most commonly occurred across regions of homology between donor and recipient chromosomes.


Assuntos
Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/genética , Resistência a Vancomicina/genética , Resistência beta-Lactâmica/genética , Proteínas de Bactérias/genética , Cromossomos Bacterianos , Conjugação Genética , Elementos de DNA Transponíveis , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Recombinação Homóloga , Óperon , Proteínas de Ligação às Penicilinas/genética , Plasmídeos , Resistência a Vancomicina/efeitos dos fármacos , Resistência beta-Lactâmica/efeitos dos fármacos
13.
Antimicrob Agents Chemother ; 59(10): 6132-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26195528

RESUMO

Beta-lactam antibiotics sensitize Enterococcus faecium to killing by endogenous antimicrobial peptides (AMPs) of the innate immune system and daptomycin through mechanisms yet to be elucidated. It has been speculated that beta-lactam inactivation of select E. faecium penicillin binding proteins (PBPs) may play a pivotal role in this sensitization process. To characterize the specific PBP inactivation that may be responsible for these phenotypes, we utilized a previously characterized set of E. faecium PBP knockout mutants to determine the effects of such mutations on the activity of daptomycin and the AMP human cathelicidin (LL-37). Enhanced susceptibility to daptomycin was dependent more on a cumulative effect of multiple PBP deletions than on inactivation of any single specific PBP. Selective knockout of PBPZ rendered E. faecium more vulnerable to killing by both recombinant LL-37 and human neutrophils, which produce the antimicrobial peptide in high quantities. Pharmacotherapy targeting multiple PBPs may be used as adjunctive therapy with daptomycin to treat difficult E. faecium infections.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/metabolismo , Proteínas de Ligação às Penicilinas/metabolismo , Daptomicina/farmacologia , Enterococcus faecium/genética , Testes de Sensibilidade Microbiana , Proteínas de Ligação às Penicilinas/genética
14.
Antimicrob Agents Chemother ; 59(9): 5306-15, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26077262

RESUMO

Bypass of the d,d-transpeptidase activity of penicillin-binding proteins by an l,d-transpeptidase (Ldtfm) results in resistance to ampicillin and glycopeptides in Enterococcus faecium M9, a mutant obtained by nine consecutive selection steps. Resistance requires activation of a cryptic locus for production of the essential tetrapeptide-containing substrate of Ldtfm and impaired activity of protein phosphatase StpA. Here, whole-genome sequencing revealed a high mutation rate for the entire selection procedure (79 mutations in 900 generations). Acquisition of a mutation in the mismatch repair gene mutL had little impact on the frequency of rifampin-resistant mutants although the mutation spectrum of M9 was typical of impaired MutL with high transversion to transition (40/11) and substitution to deletion (51/28) ratios. M9 did not mainly accumulate neutral mutations since base substitutions occurred more frequently in coding sequences than expected (χ(2) = 5.0; P < 0.05) and silent mutations were underrepresented (χ(2) = 5.72; P < 0.02). None of the mutations directly affected recognition of the tetrapeptide substrate of Ldtfm by peptidoglycan synthesis enzymes. Instead, mutations appear to remodel regulatory circuits involving two-component regulatory systems and sugar metabolism. The high number of mutations required for activation of the l,d-transpeptidase pathway may strongly limit emergence of cross-resistance to ampicillin and glycopeptides by this mechanism.


Assuntos
Enterococcus faecium/efeitos dos fármacos , Glicopeptídeos/farmacologia , beta-Lactamas/farmacologia , Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla/genética , Enterococcus faecium/genética , Mutação
15.
Am J Kidney Dis ; 65(1): 88-97, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25042816

RESUMO

BACKGROUND: Vancomycin-resistant enterococci (VRE) have become important nosocomial pathogens causing outbreaks worldwide. Patients undergoing dialysis represent a vulnerable population due to their comorbid conditions, frequent use of antibacterial agents, and frequent contact with health care settings. STUDY DESIGN: Systematic review and meta-analysis of cross-sectional studies of screening for VRE colonization. SETTING & POPULATION: Patients receiving long-term dialysis treatment. SELECTION CRITERIA FOR STUDIES: We performed a systematic literature search of PubMed and EMBASE databases to identify studies performing screening for VRE colonization among dialysis patients. PREDICTOR: Region, recent use of vancomycin or other antibiotics, previous hospitalization. OUTCOMES: (1) VRE colonization and (2) rate of VRE infection among colonized and noncolonized individuals. Relative effects were expressed as ORs and 95% CIs. RESULTS: We identified 23 studies that fulfilled the inclusion criteria and provided data for 4,842 dialysis patients from 100 dialysis centers. The pooled prevalence of VRE colonization was 6.2% (95% CI, 2.8%-10.8%), with significant variability between centers. The corresponding number for North American centers was 5.2% (95% CI, 2.8%-8.2%). Recent use of any antibiotic (OR, 3.62; 95% CI, 1.22-10.75), particularly vancomycin (OR, 5.15; 95% CI, 1.56-17.02), but also use of antibiotics other than vancomycin (OR, 2.92; 95% CI, 0.99-8.55) and recent hospitalization (OR, 4.55; 95% CI, 1.93-10.74) significantly increased the possibility of a VRE-positive surveillance culture. Colonized patients had a significantly higher risk of VRE infection (OR, 21.62; 95% CI, 5.33-87.69) than their noncolonized counterparts. LIMITATIONS: In 19 of 23 studies, a low percentage of dialysis patients (<80%) consented to participate in the screening procedure. 4 of 8 studies in which patients were followed up for more than 1 month reported VRE infections and only 5 of 23 studies provided extractable data for antibiotic consumption prior to screening. CONCLUSIONS: VRE colonization is prevalent in dialysis centers. Previous antibiotic use, in particular vancomycin, and recent hospitalization are important predicting factors of colonization, whereas the risk of VRE infection is significantly higher for colonized patients.


Assuntos
Infecção Hospitalar , Falência Renal Crônica/terapia , Diálise Renal , Enterococos Resistentes à Vancomicina/isolamento & purificação , Vancomicina/uso terapêutico , Antibacterianos/uso terapêutico , Contagem de Colônia Microbiana/estatística & dados numéricos , Infecção Hospitalar/epidemiologia , Infecção Hospitalar/etiologia , Infecção Hospitalar/microbiologia , Estudos Transversais , Hospitalização/estatística & dados numéricos , Humanos , Prevalência , Diálise Renal/efeitos adversos , Diálise Renal/métodos , Fatores de Risco
18.
Antimicrob Agents Chemother ; 58(3): 1749-56, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24395229

RESUMO

Synthesis of peptidoglycan precursors ending in D-lactate (D-Lac) is thought to be responsible for glycopeptide resistance in members of the order Actinomycetales that produce these drugs and in related soil bacteria. More recently, the peptidoglycan of several members of the order Actinomycetales was shown to be cross-linked by L,D-transpeptidases that use tetrapeptide acyl donors devoid of the target of glycopeptides. To evaluate the contribution of these resistance mechanisms, we have determined the peptidoglycan structure of Streptomyces coelicolor A(3)2, which harbors a vanHAX gene cluster for the production of precursors ending in D-Lac, and Nonomuraea sp. strain ATCC 39727, which is devoid of vanHAX and produces the glycopeptide A40296. Vancomycin retained residual activity against S. coelicolor A(3)2 despite efficient incorporation of D-Lac into cytoplasmic precursors. This was due to a D,D-transpeptidase-catalyzed reaction that generated a stem pentapeptide recognized by glycopeptides by the exchange of D-Lac for D-Ala and Gly. The contribution of L,D-transpeptidases to resistance was limited by the supply of tetrapeptide acyl donors, which are essential for the formation of peptidoglycan cross-links by these enzymes. In the absence of a cytoplasmic metallo-D,D-carboxypeptidase, the tetrapeptide substrate was generated by hydrolysis of the C-terminal D-Lac residue of the stem pentadepsipeptide in the periplasm in competition with the exchange reaction catalyzed by D,D-transpeptidases. In Nonomuraea sp. strain ATCC 39727, the contribution of L,D-transpeptidases to glycopeptide resistance was limited by the incomplete conversion of pentapeptides into tetrapeptides despite the production of a cytoplasmic metallo-D,D-carboxypeptidase. Since the level of drug production exceeds the level of resistance, we propose that L,D-transpeptidases merely act as a tolerance mechanism in this bacterium.


Assuntos
Actinomycetales/efeitos dos fármacos , Antibacterianos/farmacologia , Glicopeptídeos/farmacologia , Peptidoglicano/metabolismo , Actinomycetales/metabolismo , Farmacorresistência Bacteriana , Peptidoglicano/química , Streptomyces coelicolor/efeitos dos fármacos , Streptomyces coelicolor/metabolismo
19.
Trans Am Clin Climatol Assoc ; 124: 123-32, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874016

RESUMO

The human gastrointestinal tract is a complex environment of mutualistic associations. As bacteria form a major component of fecal content, the natural balance of the colon can be significantly altered by exposure to antimicrobial agents. However, the effects of antimicrobial therapy on fecal content are difficult to predict and can at times be quite surprising. The emergence and spread of vancomycin-resistant enterococci are cases in point. Resistance to the glycopeptide vancomycin emerged in enterococci (primarily in Enterococcus faecium) in the late 1980s in both Europe and the United States. In Europe, this emergence was tied to the use of the glycopeptide antibiotic avoparcin to promote growth in food animals and had little actual impact on hospital infections. In the United States, where avoparcin has never been licensed, vancomycin-resistant enterococci (VRE) emerged as a major hospital pathogen. Paradoxically, while the initial entry of the vancomycin resistance determinants into enterococci was almost certainly driven by high fecal concentrations of vancomycin associated with treatment for Clostridium difficile colitis, clinical infection and outbreaks were more frequently tied to use of extended-spectrum cephalosporins and agents with potent activity against anaerobic bacteria. Animal studies suggest that cephalosporins promote initial VRE colonization because of the frequent concomitant high-level resistance to ß-lactam antibiotics expressed by these strains. Anti-anaerobic agents appear to increase the output of VRE in the feces, presumably by reducing the number of competitive flora in the colon. Intravenously administered vancomycin appears to have little impact because it achieves negligible concentrations in the feces after short courses. Thus, the spread of glycopeptide resistance in enterococci is promoted in a large measure by the administration of non-glycopeptide antibiotics.


Assuntos
Antibacterianos/farmacologia , Enterococcus/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Vancomicina/farmacologia , Humanos , Resistência a Vancomicina
20.
Nat Commun ; 14(1): 4268, 2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37460557

RESUMO

Penicillin-binding proteins (PBPs) are essential for the formation of the bacterial cell wall. They are also the targets of ß-lactam antibiotics. In Enterococcus faecium, high levels of resistance to ß-lactams are associated with the expression of PBP5, with higher levels of resistance associated with distinct PBP5 variants. To define the molecular mechanism of PBP5-mediated resistance we leveraged biomolecular NMR spectroscopy of PBP5 - due to its size (>70 kDa) a challenging NMR target. Our data show that resistant PBP5 variants show significantly increased dynamics either alone or upon formation of the acyl-enzyme inhibitor complex. Furthermore, these variants also exhibit increased acyl-enzyme hydrolysis. Thus, reducing sidechain bulkiness and expanding surface loops results in increased dynamics that facilitates acyl-enzyme hydrolysis and, via increased ß-lactam antibiotic turnover, facilitates ß-lactam resistance. Together, these data provide the molecular basis of resistance of clinical E. faecium PBP5 variants, results that are likely applicable to the PBP family.


Assuntos
Antibacterianos , Hexosiltransferases , Proteínas de Ligação às Penicilinas/genética , Proteínas de Ligação às Penicilinas/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Resistência beta-Lactâmica/genética , Monobactamas , beta-Lactamas/farmacologia , Testes de Sensibilidade Microbiana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA