Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 110(51): 20747-52, 2013 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-24248354

RESUMO

An important question regarding the biologic implications of antibiotic-resistant microbes is how resistance impacts the organism's overall fitness and virulence. Currently it is generally thought that antibiotic resistance carries a fitness cost and reduces virulence. For the human pathogen Pseudomonas aeruginosa, treatment with carbapenem antibiotics is a mainstay of therapy that can lead to the emergence of resistance, often through the loss of the carbapenem entry channel OprD. Transposon insertion-site sequencing was used to analyze the fitness of 300,000 mutants of P. aeruginosa strain PA14 in a mouse model for gut colonization and systemic dissemination after induction of neutropenia. Transposon insertions in the oprD gene led not only to carbapenem resistance but also to a dramatic increase in mucosal colonization and dissemination to the spleen. These findings were confirmed in vivo with different oprD mutants of PA14 as well as with related pairs of carbapenem-susceptible and -resistant clinical isolates. Compared with OprD(+) strains, those lacking OprD were more resistant to killing by acidic pH or normal human serum and had increased cytotoxicity against murine macrophages. RNA-sequencing analysis revealed that an oprD mutant showed dramatic changes in the transcription of genes that may contribute to the various phenotypic changes observed. The association between carbapenem resistance and enhanced survival of P. aeruginosa in infected murine hosts suggests that either drug resistance or host colonization can cause the emergence of more pathogenic, drug-resistant P. aeruginosa clones in a single genetic event.


Assuntos
Carbapenêmicos/farmacologia , Farmacorresistência Bacteriana/genética , Mutação , Porinas , Pseudomonas aeruginosa , Animais , Modelos Animais de Doenças , Farmacorresistência Bacteriana/efeitos dos fármacos , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Concentração de Íons de Hidrogênio , Macrófagos/metabolismo , Macrófagos/microbiologia , Macrófagos/patologia , Masculino , Camundongos , Porinas/biossíntese , Porinas/genética , Infecções por Pseudomonas/tratamento farmacológico , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade
2.
PLoS Pathog ; 8(8): e1002834, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22876178

RESUMO

Oxidative stress serves as an important host/environmental signal that triggers a wide range of responses in microorganisms. Here, we identified an oxidative stress sensor and response regulator in the important multidrug-resistant nosocomial pathogen Enterococcus faecium belonging to the MarR family and called AsrR (antibiotic and stress response regulator). The AsrR regulator used cysteine oxidation to sense the hydrogen peroxide which results in its dissociation to promoter DNA. Transcriptome analysis showed that the AsrR regulon was composed of 181 genes, including representing functionally diverse groups involved in pathogenesis, antibiotic and antimicrobial peptide resistance, oxidative stress, and adaptive responses. Consistent with the upregulated expression of the pbp5 gene, encoding a low-affinity penicillin-binding protein, the asrR null mutant was found to be more resistant to ß-lactam antibiotics. Deletion of asrR markedly decreased the bactericidal activity of ampicillin and vancomycin, which are both commonly used to treat infections due to enterococci, and also led to over-expression of two major adhesins, acm and ecbA, which resulted in enhanced in vitro adhesion to human intestinal cells. Additional pathogenic traits were also reinforced in the asrR null mutant including greater capacity than the parental strain to form biofilm in vitro and greater persistance in Galleria mellonella colonization and mouse systemic infection models. Despite overexpression of oxidative stress-response genes, deletion of asrR was associated with a decreased oxidative stress resistance in vitro, which correlated with a reduced resistance to phagocytic killing by murine macrophages. Interestingly, both strains showed similar amounts of intracellular reactive oxygen species. Finally, we observed a mutator phenotype and enhanced DNA transfer frequencies in the asrR deleted strain. These data indicate that AsrR plays a major role in antimicrobial resistance and adaptation for survival within the host, thereby contributes importantly to the opportunistic traits of E. faecium.


Assuntos
Proteínas de Bactérias/metabolismo , Enterococcus faecium/patogenicidade , Infecções por Bactérias Gram-Positivas/metabolismo , Fatores de Transcrição/metabolismo , Resistência beta-Lactâmica , Animais , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Enterococcus faecium/genética , Enterococcus faecium/metabolismo , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/genética , Humanos , Peróxido de Hidrogênio/farmacologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Oxidantes/farmacologia , Oxirredução/efeitos dos fármacos , Característica Quantitativa Herdável , Fatores de Transcrição/genética , beta-Lactamas/farmacologia
3.
Antimicrob Agents Chemother ; 57(9): 4463-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23836170

RESUMO

As opposed to Enterococcus faecalis, which is intrinsically resistant to lincosamides, streptogramins A, and pleuromutilins (LSAP phenotype) by production of the ABC protein Lsa(A), Enterococcus faecium is naturally susceptible. Since this phenotype may be selected for in vivo by quinupristin-dalfopristin (Q-D), the aim of this study was to investigate the molecular mechanism of acquired LSAP resistance in E. faecium. Six LSAP-resistant in vitro mutants of E. faecium HM1070 as well as three different pairs of clinical isolates (pre- and postexposure to Q-D) were studied. The full genome sequence of an in vitro mutant (E. faecium UCN90B) was determined by using 454 sequencing technology and was compared with that of the parental strain. Single-nucleotide replacement was carried out to confirm the role of this mutation. By comparative genomic analysis, a point mutation was found within a 1,503-bp gene coding for an ABC homologue showing 66% amino acid identity with Lsa(A). This mutation (C1349T) led to an amino acid substitution (Thr450Ile). An identical mutation was identified in all in vitro and in vivo resistant strains but was not present in susceptible strains. The wild-type allele was named eat(A) (for Enterococcus ABC transporter), and its mutated allelic variant was named eat(A)v. The introduction of eat(A)v from UCN90B into HM1070 conferred the LSAP phenotype, whereas that of eat(A) from HM1070 into UCN90B restored susceptibility entirely. This is the first description of the molecular mechanism of acquired LSAP resistance in E. faecium. Characterization of the biochemical mechanism of resistance and the physiological role of this ABC protein need further investigations.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Farmacorresistência Bacteriana Múltipla/genética , Enterococcus faecium/genética , Lincosamidas/farmacologia , Estreptogramina A/farmacologia , Transportadores de Cassetes de Ligação de ATP/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Sequência de Bases , Diterpenos/farmacologia , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/metabolismo , Teste de Complementação Genética , Genótipo , Dados de Sequência Molecular , Fenótipo , Mutação Puntual , Compostos Policíclicos , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Pleuromutilinas
4.
Antimicrob Agents Chemother ; 57(10): 4615-21, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23817377

RESUMO

Rhodococcus equi, the causal agent of rhodococcosis, is a major pathogen of foals and is also responsible for severe infections in immunocompromised humans. Of great concern, strains resistant to currently used antibiotics have emerged. As the number of drugs that are efficient in vivo is limited because of the intracellular localization of the bacterium inside macrophages, new active but cell-permeant drugs will be needed in the near future. In the present study, we evaluated, by in vitro and ex vivo experiments, the ability of the alpha-helical equine antimicrobial peptide eCATH1 to kill intracellular bacterial cells. Moreover, the therapeutic potential of the peptide was assessed in experimental rhodococcosis induced in mice, while the in vivo toxicity was evaluated by behavioral and histopathological analysis. The study revealed that eCATH1 significantly reduced the number of bacteria inside macrophages. Furthermore, the bactericidal potential of the peptide was maintained in vivo at doses that appeared to have no visible deleterious effects for the mice even after 7 days of treatment. Indeed, daily subcutaneous injections of 1 mg/kg body weight of eCATH1 led to a significant reduction of the bacterial load in organs comparable to that obtained after treatment with 10 mg/kg body weight of rifampin. Interestingly, the combination of the peptide with rifampin showed a synergistic interaction in both ex vivo and in vivo experiments. These results emphasize the therapeutic potential that eCATH1 represents in the treatment of rhodococcosis.


Assuntos
Infecções por Actinomycetales/tratamento farmacológico , Anti-Infecciosos/uso terapêutico , Rhodococcus equi/patogenicidade , Animais , Linhagem Celular , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Rhodococcus equi/efeitos dos fármacos , Rifampina/uso terapêutico
5.
J Antimicrob Chemother ; 68(4): 731-42, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23208830

RESUMO

Twenty-five years ago, isolation of vancomycin-resistant Enterococcus faecium (VREm) was reported both in the UK and in France. Since then, VREm has spread worldwide in hospitals. Hospital outbreaks appeared to be related to the evolution since the end of 1980s of a subpopulation of E. faecium highly resistant to ampicillin and fluoroquinolones (the so-called clonal complex CC17) that later acquired resistance to vancomycin. CC17 isolates are presumably better adapted than other E. faecium isolates to the constraints of the hospital environment and most contain mobile genetic elements, phage genes, genes encoding membrane proteins, regulatory genes, a putative pathogenicity island and megaplasmids. Colonization and persistence are major features of VREm. Inherent characteristics of E. faecium including a remarkable genome plasticity, in part due to acquisition of IS elements, in particular IS16, have facilitated niche adaptation of this distinct E. faecium subpopulation that is multiply resistant to antibiotics. Quinupristin/dalfopristin and linezolid are licensed for the treatment of VREm infections, with linezolid often used as a first-line treatment. However, the emergence of plasmid-mediated resistance to linezolid by production of a Cfr methyltransferase in Enterococcus faecalis is worrying. Daptomycin has not been extensively evaluated for the treatment of VREm infections and resistant mutants have been selected under daptomycin therapy. Although control of VRE is challenging, a laissez-faire policy would result in an increased number of infections and would create an irreversible situation. Although so far unsuccessful, dissemination of glycopeptide-resistant Staphylococcus aureus with van genes acquired from resistant enterococci cannot be ruled out.


Assuntos
Infecção Hospitalar/epidemiologia , Infecção Hospitalar/microbiologia , Surtos de Doenças , Enterococcus/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/epidemiologia , Infecções por Bactérias Gram-Positivas/microbiologia , Resistência a Vancomicina , Portador Sadio/epidemiologia , Portador Sadio/microbiologia , DNA Bacteriano/genética , Enterococcus/classificação , Enterococcus/genética , Enterococcus/isolamento & purificação , Genótipo , Saúde Global , Humanos , Controle de Infecções/métodos
6.
J Antimicrob Chemother ; 68(11): 2464-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23812683

RESUMO

OBJECTIVES: Unique resistance to lincosamides (L phenotype) due to the production of nucleotidyltransferases (Lnu) is uncommon among Gram-positive bacteria. The aim of the study was to characterize the L phenotype in a clinical isolate of the Streptococcus milleri group. METHODS: The strain UCN93 was recovered from neonatal specimens and from the mother's vaginal swab. Identification was confirmed by sequencing of the sodA gene. Antimicrobial susceptibility testing was carried out by the disc diffusion method, while MICs were determined using the agar dilution method. Screening for lnu(A), lnu(B), lnu(C) and lnu(D) genes was performed by PCR. Genetic environment and support were determined by thermal asymmetric interlaced PCR and PCR mapping. The transfer of lincomycin resistance was also attempted by conjugation. RESULTS: UCN93 was unambiguously identified as Streptococcus anginosus. It was susceptible to all tested antibiotics, except lincomycin (MIC, 8 mg/L) and tetracycline (2 mg/L). The lnu(C) gene was found to be responsible for the L phenotype. It was shown that lnu(C) was associated with a gene coding for a transposase within a structure similar to the transposon MTnSag1, described once in Streptococcus agalactiae. Since MTnSag1 was found to be mobilized by Tn916 and S. anginosus UCN93 harboured a Tn916 transposon, several attempts at transfer were performed but they all failed. The lnu(C)-containing genetic element was inserted into a chromosomal intergenic sequence of S. anginosus. CONCLUSIONS: Since lnu(C) has been detected in only one S. agalactiae clinical isolate so far, this is its second description among clinically relevant streptococci.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Farmacorresistência Bacteriana , Lincosamidas/farmacologia , Infecções Estreptocócicas/microbiologia , Streptococcus anginosus/efeitos dos fármacos , Streptococcus anginosus/genética , Conjugação Genética , DNA Bacteriano/genética , Transferência Genética Horizontal , Humanos , Recém-Nascido , Testes de Sensibilidade Microbiana , Mães , Fenótipo , Reação em Cadeia da Polimerase , Streptococcus anginosus/isolamento & purificação
7.
Appl Environ Microbiol ; 79(7): 2428-34, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23377946

RESUMO

To determine if hospital effluent input has an ecological impact on downstream aquatic environment, antibiotic resistance in Enterococcus spp. along a medical center-retirement home-wastewater treatment plant-river continuum in France was determined using a culture-based method. Data on antibiotic consumption among hospitalized and general populations and levels of water contamination by antibiotics were collected. All isolated enterococci were genotypically identified to the species level, tested for in vitro antibiotic susceptibility, and typed by multilocus sequence typing. The erm(B) and mef(A) (macrolide resistance) and tet(M) (tetracycline resistance) genes were detected by PCR. Along the continuum, from 89 to 98% of enterococci, according to the sampled site, were identified as Enterococcus faecium. All E. faecium isolates from hospital and retirement home effluents were multiply resistant to antibiotics, contained erm(B) and mef(A) genes, and belonged to hospital-adapted clonal complex 17 (CC17). Even though this species remained dominant in the downstream continuum, the relative proportion of CC17 isolates progressively decreased in favor of other subpopulations of E. faecium that were more diverse, less resistant to antibiotics, and devoid of the classical macrolide resistance genes and that belonged to various sequence types. Antibiotic concentrations in waters were far below the MICs for susceptible isolates. CC17 E. faecium was probably selected in the gastrointestinal tract of patients under the pressure of administered antibiotics and then excreted together with the resistance genes in waters to progressively decrease along the continuum.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana , Enterococcus/efeitos dos fármacos , Enterococcus/isolamento & purificação , Rios/microbiologia , Águas Residuárias/microbiologia , Centros Médicos Acadêmicos , Proteínas de Bactérias/genética , Enterococcus/classificação , Enterococcus/genética , França , Humanos , Testes de Sensibilidade Microbiana , Tipagem de Sequências Multilocus , Reação em Cadeia da Polimerase
8.
Antimicrob Agents Chemother ; 56(4): 1749-55, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22232283

RESUMO

Rhodococcus equi, the causal agent of rhodococcosis, is a severe pathogen of foals but also of immunodeficient humans, causing bronchopneumonia. The pathogen is often found together with Klebsiella pneumoniae or Streptococcus zooepidemicus in foals. Of great concern is the fact that some R. equi strains are already resistant to commonly used antibiotics. In the present study, we evaluated the in vitro potential of two equine antimicrobial peptides (AMPs), eCATH1 and DEFA1, as new drugs against R. equi and its associated pathogens. The peptides led to growth inhibition and death of R. equi and S. zooepidemicus at low micromolar concentrations. Moreover, eCATH1 was able to inhibit growth of K. pneumoniae. Both peptides caused rapid disruption of the R. equi membrane, leading to cell lysis. Interestingly, eCATH1 had a synergic effect together with rifampin. Furthermore, eCATH1 was not cytotoxic against mammalian cells at bacteriolytic concentrations and maintained its high killing activity even at physiological salt concentrations. Our data suggest that equine AMPs, especially eCATH1, may be promising candidates for alternative drugs to control R. equi in mono- and coinfections.


Assuntos
Infecções por Actinomycetales/tratamento farmacológico , Infecções por Actinomycetales/microbiologia , Antibacterianos/farmacologia , Doenças dos Cavalos/tratamento farmacológico , Doenças dos Cavalos/microbiologia , Rhodococcus equi , alfa-Defensinas/farmacologia , Infecções por Actinomycetales/veterinária , Animais , Antibacterianos/química , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Dicroísmo Circular , Farmacorresistência Bacteriana , Sinergismo Farmacológico , Feminino , Hemólise , Cavalos , Lipossomos/química , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Varredura , Fosfolipídeos/química , Rhodococcus equi/efeitos dos fármacos , Rhodococcus equi/ultraestrutura , Tolerância ao Sal , Ovinos , Células Vero , alfa-Defensinas/química
9.
J Clin Microbiol ; 50(10): 3345-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22814459

RESUMO

Twenty-three strains of Staphylococcus aureus with borderline resistance to oxacillin were studied. These strains were not detected by the cefoxitin test, tests for penicillin-binding protein 2a (PBP2a), mecA, and mecA(LGA251) were negative, and the strains were genetically unrelated. To detect all strains resistant to oxacillin, laboratories should routinely test for both cefoxitin and oxacillin.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Oxacilina/farmacologia , Infecções Estafilocócicas/microbiologia , Proteínas de Bactérias/genética , Cefoxitina/farmacologia , Análise por Conglomerados , Eletroforese em Gel de Campo Pulsado , Humanos , Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Testes de Sensibilidade Microbiana/métodos , Epidemiologia Molecular , Tipagem Molecular , Proteínas de Ligação às Penicilinas/genética , Infecções Estafilocócicas/epidemiologia , Tunísia/epidemiologia
10.
Antimicrob Agents Chemother ; 55(4): 1470-4, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21245447

RESUMO

Streptococcus agalactiae UCN70, isolated from a vaginal swab obtained in New Zealand, is resistant to lincosamides and streptogramins A (LS(A) phenotype) and also to tiamulin (a pleuromutilin). By whole-genome sequencing, we identified a 5,224-bp chromosomal extra-element that comprised a 1,479-bp open reading frame coding for an ABC protein (492 amino acids) 45% identical to Lsa(A), a protein related to intrinsic LS(A) resistance in Enterococcus faecalis. Expression of this novel gene, named lsa(C), in S. agalactiae BM132 after cloning led to an increase in MICs of lincomycin (0.06 to 4 µg/ml), clindamycin (0.03 to 2 µg/ml), dalfopristin (2 to >32 µg/ml), and tiamulin (0.12 to 32 µg/ml), whereas no change in MICs of erythromycin (0.06 µg/ml), azithromycin (0.03 µg/ml), spiramycin (0.25 µg/ml), telithromycin (0.03 µg/ml), and quinupristin (8 µg/ml) was observed. The phenotype was renamed the LS(A)P phenotype on the basis of cross-resistance to lincosamides, streptogramins A, and pleuromutilins. This gene was also identified in similar genetic environments in 17 other S. agalactiae clinical isolates from New Zealand exhibiting the same LS(A)P phenotype, whereas it was absent in susceptible S. agalactiae strains. Interestingly, this extra-element was bracketed by a 7-bp duplication of a target site (ATTAGAA), suggesting that this structure was likely a mobile genetic element. In conclusion, we identified a novel gene, lsa(C), responsible for the acquired LS(A)P resistance phenotype in S. agalactiae. Dissection of the biochemical basis of resistance, as well as demonstration of in vitro mobilization of lsa(C), remains to be performed.


Assuntos
Antibacterianos/farmacologia , Lincosamidas/farmacologia , Streptococcus agalactiae/efeitos dos fármacos , Estreptograminas/farmacologia , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Diterpenos/farmacologia , Farmacorresistência Bacteriana Múltipla/genética , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Compostos Policíclicos , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos , Streptococcus agalactiae/genética , Pleuromutilinas
11.
Antimicrob Agents Chemother ; 55(10): 4606-12, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21807981

RESUMO

Enterococcus faecium UCN71, isolated from a blood culture, was resistant to low levels of vancomycin (MIC, 16 µg/ml) but susceptible to teicoplanin (MIC, 0.5 µg/ml). No amplification was observed with primers specific for the previously described glycopeptide resistance ligase genes, but a PCR product corresponding to a gene called vanN was obtained using degenerate primers and was sequenced. The deduced VanN protein was related (65% identity) to the d-alanine:d-serine VanL ligase. The organization of the vanN gene cluster, determined using degenerate primers and by thermal asymmetric interlaced (TAIL)-PCR, was similar to that of the vanC operons. A single promoter upstream from the resistance operon was identified by rapid amplification of cDNA ends (RACE)-PCR. The presence of peptidoglycan precursors ending in d-serine and d,d-peptidase activities in the absence of vancomycin indicated constitutive expression of the resistance operon. VanN-type resistance was transferable by conjugation to E. faecium. This is the first report of transferable d-Ala-d-Ser-type resistance in E. faecium.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/genética , Peptídeo Sintases/genética , Resistência a Vancomicina/genética , Vancomicina/farmacologia , Sequência de Aminoácidos , Sequência de Bases , Enterococcus faecium/metabolismo , Genes Bacterianos , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Família Multigênica , Alinhamento de Sequência , Análise de Sequência de DNA , Teicoplanina/farmacologia
12.
J Antimicrob Chemother ; 66(4): 713-21, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21393182

RESUMO

OBJECTIVES: Unprecedented outbreaks of vancomycin-resistant enterococci (VRE) have occurred in French hospitals since 2004. The aim of this study was to provide a picture of the spread and control of VRE in France and to characterize the isolates. METHODS: Notification of VRE cases to Institut de Veille Sanitaire has been mandatory since 2001. Isolates of VRE were sent to the National Reference Centre for species and vancomycin-resistance gene identification. Isolates were tested for antimicrobial susceptibility and typed by PFGE and multilocus sequence typing. RESULTS: Five hundred and four VRE notifications from 195 hospitals were recorded, corresponding to 2475 cases of infection (n=243) or colonization (n=2232) and 74 episodes of clustered cases. Outbreaks were controlled by implementation of infection control measures, although the number of new hospitals reporting isolation of VRE was increasing. The majority of 902 VRE isolated from 2006 to 2008 were Enterococcus faecium (94.8%) with the vanA or vanB gene. No isolate was resistant to linezolid, tigecycline or fusidic acid. PFGE analysis showed 161 different patterns. Generally a few predominant clones and several minor clones spread in a single hospital. In a subset of 46 representatives of PFGE clones, 13 different sequence types were characterized, all belonging to clonal complex CC17, while the esp and hyl genes were inconsistently detected. CONCLUSIONS: The national mandatory notification of unusual nosocomial events allowed rapid identification of VRE outbreaks and early implementation of control measures that have proved effective. However, VRE continue to emerge in a growing number of hospitals.


Assuntos
Infecção Hospitalar/epidemiologia , Enterococcus/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/epidemiologia , Infecções por Bactérias Gram-Positivas/microbiologia , Resistência a Vancomicina , Vancomicina/farmacologia , Proteínas de Bactérias/genética , Técnicas de Tipagem Bacteriana , Carbono-Oxigênio Ligases/genética , Infecção Hospitalar/microbiologia , Surtos de Doenças , Eletroforese em Gel de Campo Pulsado , Enterococcus/isolamento & purificação , França/epidemiologia , Genótipo , Hospitais , Humanos , Proteínas de Membrana/genética , Testes de Sensibilidade Microbiana , Tipagem Molecular , Tipagem de Sequências Multilocus
13.
J Bacteriol ; 192(8): 2266-76, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20139183

RESUMO

Streptococcus gallolyticus (formerly known as Streptococcus bovis biotype I) is an increasing cause of endocarditis among streptococci and frequently associated with colon cancer. S. gallolyticus is part of the rumen flora but also a cause of disease in ruminants as well as in birds. Here we report the complete nucleotide sequence of strain UCN34, responsible for endocarditis in a patient also suffering from colon cancer. Analysis of the 2,239 proteins encoded by its 2,350-kb-long genome revealed unique features among streptococci, probably related to its adaptation to the rumen environment and its capacity to cause endocarditis. S. gallolyticus has the capacity to use a broad range of carbohydrates of plant origin, in particular to degrade polysaccharides derived from the plant cell wall. Its genome encodes a large repertoire of transporters and catalytic activities, like tannase, phenolic compounds decarboxylase, and bile salt hydrolase, that should contribute to the detoxification of the gut environment. Furthermore, S. gallolyticus synthesizes all 20 amino acids and more vitamins than any other sequenced Streptococcus species. Many of the genes encoding these specific functions were likely acquired by lateral gene transfer from other bacterial species present in the rumen. The surface properties of strain UCN34 may also contribute to its virulence. A polysaccharide capsule might be implicated in resistance to innate immunity defenses, and glucan mucopolysaccharides, three types of pili, and collagen binding proteins may play a role in adhesion to tissues in the course of endocarditis.


Assuntos
Endocardite/microbiologia , Genoma Bacteriano/fisiologia , Streptococcus/genética , Streptococcus/patogenicidade , Animais , Bovinos , Genoma Bacteriano/genética , Modelos Genéticos , Dados de Sequência Molecular , Filogenia , Polissacarídeos/metabolismo , Streptococcus/classificação , Streptococcus/metabolismo , Vitaminas/metabolismo
14.
J Clin Microbiol ; 48(2): 581-5, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19955271

RESUMO

Fifty-three strains belonging to the pathogenic species Leptospira interrogans and Leptospira kirschneri were analyzed by multilocus sequence analysis. The species formed two distinct branches. In the L. interrogans branch, the phylogenetic tree clustered the strains into three subgroups. Genogroups and serogroups were superimposed but not strictly.


Assuntos
Técnicas de Tipagem Bacteriana/métodos , Impressões Digitais de DNA/métodos , Leptospira/classificação , Leptospira/genética , Análise de Sequência , Animais , Análise por Conglomerados , DNA Bacteriano/química , DNA Bacteriano/genética , Genótipo , Humanos , Leptospira/isolamento & purificação , Leptospirose/microbiologia , Leptospirose/veterinária , Dados de Sequência Molecular , Filogenia , Sorotipagem
15.
Med Sci (Paris) ; 26(11): 936-42, 2010 Nov.
Artigo em Francês | MEDLINE | ID: mdl-21106175

RESUMO

Enterococci are responsible for various community- and hospital-acquired infections. Glycopeptides (vancomycin and teicoplanin) are active against these microorganisms by inhibiting cell wall synthesis through binding to cell wall precursors. Enterococcus faecium has developed multidrug resistance, including resistance to glycopeptides. Resistance to glycopeptides is due to the acquisition of an operon of genes cooperating to synthesize precursors devoid of affinity for the glycopeptides. Outbreaks were recently reported in hospital settings. These outbreaks were due to E. faecium isolates belonging to an hospital-adapted clonal complex (CC17) characterized by high level resistance to ampicillin and fluoroquinolones and frequently containing virulence factors. Outbreaks may be controlled by appropriate measures and new antibiotics are available in therapy. However, spreading of clonal strains adapted to hospitals require close surveillance.


Assuntos
Farmacorresistência Bacteriana/efeitos dos fármacos , Enterococcus/efeitos dos fármacos , Glicopeptídeos/uso terapêutico , Cromossomos Bacterianos/efeitos dos fármacos , Cromossomos Bacterianos/genética , Infecções Comunitárias Adquiridas/tratamento farmacológico , Infecção Hospitalar/tratamento farmacológico , Dipeptídeos/farmacologia , Enterococcus faecium/efeitos dos fármacos , Enterococcus faecium/genética , Europa (Continente) , França , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/epidemiologia , Humanos , Vancomicina/uso terapêutico
16.
Emerg Infect Dis ; 15(10): 1648-50, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19861064

RESUMO

The plasmid-borne mph(A) gene that confers resistance to azithromycin and has recently emerged in Shigella sonnei is present in multidrug- and non-multidrug-resistant Escherichia coli isolates from 4 continents. Further spread of mph(A) to Shigella and Salmonella spp. may be expected.


Assuntos
Farmacorresistência Bacteriana Múltipla/genética , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Macrolídeos/farmacologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Testes de Sensibilidade Microbiana
17.
Antimicrob Agents Chemother ; 53(9): 4007-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19546366

RESUMO

We compared the propensities of Enterococcus faecalis JH2-2 and of the recombination-deficient JH2-2 recA strain to develop mutational resistance to linezolid. In both organisms, a mutation in a single rrl copy conferred resistance to linezolid. Delay in acquisition of the mutation by other rrl copies in JH2-2 recA showed that gene conversion contributed to the acquisition of resistance.


Assuntos
Acetamidas/farmacologia , Antibacterianos/farmacologia , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/genética , Oxazolidinonas/farmacologia , Farmacorresistência Bacteriana/genética , Linezolida , Testes de Sensibilidade Microbiana , Mutação
18.
Emerg Infect Dis ; 14(8): 1297-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18680661

RESUMO

Shigella sonnei UCN59, isolated during an outbreak of S. sonnei in January 2007, was resistant to azithromycin (MIC 64 mg/L). The isolate contained a plasmid-borne mph(A) gene encoding a macrolide 2 -phosphotransferase that inactivates macrolides. Emergence of the mph(A) gene in S. sonnei may limit usefulness of azithromycin for treatment of shigellosis.


Assuntos
Antibacterianos/farmacologia , Farmacorresistência Bacteriana Múltipla , Macrolídeos/farmacologia , Shigella sonnei/efeitos dos fármacos , Pré-Escolar , Surtos de Doenças , Disenteria Bacilar/microbiologia , Eletroforese em Gel de Campo Pulsado , Feminino , Humanos , Filogenia , Reação em Cadeia da Polimerase
19.
Antimicrob Agents Chemother ; 52(8): 2767-70, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18519724

RESUMO

Streptococcus uberis UCN60 was resistant to spiramycin (MIC = 8 microg/ml) but susceptible to erythromycin (MIC = 0.06 microg/ml), azithromycin (MIC = 0.12 microg/ml), josamycin (MIC = 0.25 microg/ml), and tylosin (MIC = 0.5 microg/ml). A 2.5-kb HindIII fragment was cloned from S. uberis UCN60 DNA on plasmid pUC18 and introduced into Escherichia coli AG100A, where it conferred resistance to spiramycin by inactivation. The sequence analysis of the fragment showed the presence of an rdmC-like gene that putatively encoded a protein belonging to the alpha/beta hydrolase family and of the first 196 nucleotides of the mph(B) gene putatively encoding a phosphotransferase known to inactivate 14-, 15-, and 16-membered macrolides in E. coli. The entire mph(B) gene was then identified in S. uberis UCN60. The two genes were expressed alone or in combination in E. coli, Staphylococcus aureus, and Enterococcus faecalis. Analysis of MICs revealed that rdmC-like alone did not confer resistance to erythromycin, tylosin, and josamycin in those three hosts. It conferred resistance to spiramycin in E. coli and E. faecalis but not in S. aureus. mph(B) conferred resistance in E. coli to erythromycin, tylosin, josamycin, and spiramycin but only low levels of resistance in E. faecalis and S. aureus to spiramycin (MIC = 8 microg/ml). The combination of mph(B) and rdmC-like genes resulted in a resistance to spiramycin and tylosin in the three hosts that significantly exceeded the mere addition of the resistance levels conferred by each resistance mechanism alone.


Assuntos
Farmacorresistência Bacteriana/genética , Genes Bacterianos/genética , Macrolídeos/farmacologia , Streptococcus/efeitos dos fármacos , Streptococcus/genética , Azitromicina/farmacologia , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/genética , Eritromicina/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Josamicina/farmacologia , Testes de Sensibilidade Microbiana , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/genética , Tilosina/farmacologia
20.
J Clin Microbiol ; 46(2): 546-50, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18077631

RESUMO

In staphylococci, inducible macrolide-lincosamide-streptogramin B (MLS(B)) resistance is conferred by the erm(C) or erm(A) gene. This phenotype is characterized by the erythromycin-clindamycin "D-zone" test. Although clindamycin appears active in vitro, exposure of MLS(B)-inducible Staphylococcus aureus to this antibiotic may result in the selection of clindamycin-resistant mutants, either in vitro or in vivo. We have compared the frequencies of mutation to clindamycin resistance for 28 isolates of S. aureus inducibly resistant to erythromycin and bearing the erm(C) (n = 18) or erm(A) (n = 10) gene. Seven isolates susceptible to erythromycin or bearing the msr(A) gene (efflux) were used as controls. The frequencies of mutation to clindamycin resistance for the erm(A) isolates (mean +/- standard deviation, 3.4 x 10(-8) +/- 2.4 x 10(-8)) were only slightly higher than those for the controls (1.1 x 10(-8) +/- 6.4 x 10(-9)). By contrast, erm(C) isolates displayed a mean frequency of mutation to clindamycin resistance (4.7 x 10(-7) +/- 5.5 x 10(-7)) 14-fold higher than that of the S. aureus isolates with erm(A). The difference was also observed, although to a lower extent, when erm(C) and erm(A) were cloned into S. aureus RN4220. We conclude that erm(C) and erm(A) have different genetic potentials for selection of clindamycin-resistant mutants. By the disk diffusion method, erm(C) and erm(A) isolates could be distinguished on the basis of high- and low-level resistance to oleandomycin, respectively.


Assuntos
Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Clindamicina/farmacologia , Farmacorresistência Bacteriana/genética , Metiltransferases/genética , Mutação , Seleção Genética , Staphylococcus aureus/genética , DNA Bacteriano/genética , Macrolídeos/farmacologia , Análise de Sequência de DNA , Staphylococcus aureus/efeitos dos fármacos , Estreptogramina Grupo B/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA