Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Antimicrob Chemother ; 77(4): 1155-1165, 2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-35016205

RESUMO

BACKGROUND: DAV132 (colon-targeted adsorbent) has prevented antibiotic-induced effects on microbiota in healthy volunteers. OBJECTIVES: To assess DAV132 safety and biological efficacy in patients. PATIENTS AND METHODS: An open-label, randomized [stratification: fluoroquinolone (FQ) indication] multicentre trial comparing DAV132 (7.5 g, 3 times a day, orally) with No-DAV132 in hospitalized patients requiring 5-21 day treatment with FQs and at risk of Clostridioides difficile infection (CDI). FQ and DAV132 were started simultaneously, DAV132 was administered for 48 h more, and patients were followed up for 51 days. The primary endpoint was the rate of adverse events (AEs) independently adjudicated as related to DAV132 and/or FQ. The planned sample size of 260 patients would provide a 95% CI of ±11.4%, assuming a 33% treatment-related AE rate. Plasma and faecal FQ concentrations, intestinal microbiota diversity, intestinal colonization with C. difficile, MDR bacteria and yeasts, and ex vivo resistance to C. difficile faecal colonization were assessed. RESULTS: Two hundred and forty-three patients (median age 71 years; 96% with chronic comorbidity) were included (No-DAV132, n = 120; DAV132, n = 123). DAV132- and/or FQ-related AEs did not differ significantly: 18 (14.8%) versus 13 (10.8%) in DAV132 versus No-DAV132 patients (difference 3.9%; 95% CI: -4.7 to 12.6). Day 4 FQ plasma levels were unaffected. DAV132 was associated with a >98% reduction in faecal FQ levels (Day 4 to end of treatment; P < 0.001), less impaired microbiota diversity (Shannon index; P = 0.003), increased ex vivo resistance to C. difficile colonization (P = 0.0003) and less frequent FQ-induced VRE acquisition (P = 0.01). CONCLUSIONS: In FQ-treated hospitalized patients, DAV132 was well tolerated, and FQ plasma concentrations unaffected. DAV132 preserved intestinal microbiota diversity and C. difficile colonization resistance.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Microbioma Gastrointestinal , Idoso , Antibacterianos/efeitos adversos , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/prevenção & controle , Fluoroquinolonas/efeitos adversos , Humanos
2.
Artigo em Inglês | MEDLINE | ID: mdl-33139292

RESUMO

A healthy, intact gut microbiota is often resistant to colonization by gastrointestinal pathogens. During periods of dysbiosis, however, organisms such as Clostridioides difficile can thrive. We describe an optimized in vitro colonization resistance assay for C. difficile in stool (CRACS) and demonstrate the utility of this assay by assessing changes in colonization resistance following antibiotic exposure. Fecal samples were obtained from healthy volunteers (n = 6) and from healthy subjects receiving 5 days of moxifloxacin (n = 11) or no antibiotics (n = 10). Samples were separated and either not manipulated (raw) or sterilized (autoclaved or filtered) prior to inoculation with C. difficile ribotype 027 spores and anaerobic incubation for 72 h. Different methods of storing fecal samples were also investigated in order to optimize the CRACS. In healthy, raw fecal samples, incubation with spores did not lead to increased C. difficile total viable counts (TVCs) or cytotoxin detection. In contrast, increased C. difficile TVCs and cytotoxin detection occurred in sterilized healthy fecal samples or those from antibiotic-treated individuals. The CRACS was functional with fecal samples stored at either 4°C or -80°C but not with those stored with glycerol (12% or 30% [vol/vol]). Our data show that the CRACS successfully models in vitro the loss of colonization resistance and subsequent C. difficile proliferation and toxin production. The CRACS could be used as a proxy for C. difficile infection in clinical studies or to determine if an individual is at risk of developing C. difficile infection or other potential infections occurring due to a loss of colonization resistance.


Assuntos
Clostridioides difficile , Infecções por Clostridium , Antibacterianos/farmacologia , Clostridioides , Infecções por Clostridium/tratamento farmacológico , Voluntários Saudáveis , Humanos
3.
J Antimicrob Chemother ; 75(3): 709-717, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31821452

RESUMO

OBJECTIVES: Routine amoxicillin for children with uncomplicated severe acute malnutrition raises concerns of increasing antibiotic resistance. We performed an ancillary study nested within a double-blind, placebo-controlled trial in Niger testing the role of routine 7 day amoxicillin therapy in nutritional recovery of children 6 to 59 months of age with uncomplicated severe acute malnutrition. METHODS: We screened 472 children for rectal carriage of ESBL-producing Enterobacteriaceae (ESBL-E) as well as their household siblings under 5 years old, at baseline and Week 1 (W1) and Week 4 (W4) after start of therapy, and characterized strains by WGS. ClinicalTrials.gov: NCT01613547. RESULTS: Carriage in index children at baseline was similar in the amoxicillin and the placebo groups (33.8% versus 27.9%, P = 0.17). However, acquisition of ESBL-E in index children at W1 was higher in the amoxicillin group than in the placebo group (53.7% versus 32.2%, adjusted risk ratio = 2.29, P = 0.001). Among 209 index and sibling households possibly exposed to ESBL-E transmission, 16 (7.7%) had paired strains differing by ≤10 SNPs, suggesting a high probability of transmission. This was more frequent in households from the amoxicillin group than from the placebo group [11.5% (12/104) versus 3.8% (4/105), P = 0.04]. CONCLUSIONS: Among children exposed to amoxicillin, ESBL-E colonization was more frequent and the risk of transmission to siblings higher. Routine amoxicillin should be carefully balanced with the risks associated with ESBL-E colonization.


Assuntos
Infecções por Enterobacteriaceae , Enterobacteriaceae , Amoxicilina , Antibacterianos/uso terapêutico , Pré-Escolar , Infecções por Enterobacteriaceae/tratamento farmacológico , Humanos , Lactente , Níger , beta-Lactamases
4.
Artigo em Inglês | MEDLINE | ID: mdl-31307985

RESUMO

Although the global deleterious impact of antibiotics on the intestinal microbiota is well known, temporal changes in microbial diversity during and after an antibiotic treatment are still poorly characterized. We used plasma and fecal samples collected frequently during treatment and up to one month after from 22 healthy volunteers assigned to a 5-day treatment by moxifloxacin (n = 14) or no intervention (n = 8). Moxifloxacin concentrations were measured in both plasma and feces, and bacterial diversity was determined in feces by 16S rRNA gene profiling and quantified using the Shannon index and number of operational taxonomic units (OTUs). Nonlinear mixed effect models were used to relate drug pharmacokinetics and bacterial diversity over time. Moxifloxacin reduced bacterial diversity in a concentration-dependent manner, with a median maximal loss of 27.5% of the Shannon index (minimum [min], 17.5; maximum [max], 27.7) and 47.4% of the number of OTUs (min, 30.4; max, 48.3). As a consequence of both the long fecal half-life of moxifloxacin and the susceptibility of the gut microbiota to moxifloxacin, bacterial diversity indices did not return to their pretreatment levels until days 16 and 21, respectively. Finally, the model characterized the effect of moxifloxacin on bacterial diversity biomarkers and provides a novel framework for analyzing antibiotic effects on the intestinal microbiome.


Assuntos
Antibacterianos/uso terapêutico , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Microbiota/efeitos dos fármacos , Microbiota/genética , Adulto , Bactérias/efeitos dos fármacos , Bactérias/genética , Fezes/microbiologia , Feminino , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Moxifloxacina/uso terapêutico , RNA Ribossômico 16S/genética , Análise de Sequência de DNA/métodos , Adulto Jovem
5.
Artigo em Inglês | MEDLINE | ID: mdl-31636067

RESUMO

Fluoroquinolone treatments induce dysbiosis of the intestinal microbiota, resulting in loss of resistance to colonization by exogenous bacteria such as Clostridioides difficile that may cause severe diarrhea in humans and lethal infection in hamsters. We show here that DAV131A, a charcoal-based adsorbent, decreases the intestinal levels of the fluoroquinolone antibiotics levofloxacin and ciprofloxacin in hamsters, protects their intestinal microbiota, and prevents lethal infection by C. difficile.


Assuntos
Carvão Vegetal/administração & dosagem , Clostridioides difficile , Infecções por Clostridium/prevenção & controle , Administração Oral , Adsorção , Animais , Antibacterianos/efeitos adversos , Antibacterianos/farmacocinética , Ciprofloxacina/efeitos adversos , Ciprofloxacina/farmacocinética , Clostridioides difficile/patogenicidade , Modelos Animais de Doenças , Disbiose/induzido quimicamente , Disbiose/metabolismo , Disbiose/prevenção & controle , Fluoroquinolonas/efeitos adversos , Fluoroquinolonas/farmacocinética , Microbioma Gastrointestinal/efeitos dos fármacos , Levofloxacino/efeitos adversos , Levofloxacino/farmacocinética , Masculino , Mesocricetus
6.
Artigo em Inglês | MEDLINE | ID: mdl-30936104

RESUMO

Ceftriaxone has a higher biliary elimination than cefotaxime (40% versus 10%), which may result in a more pronounced impact on the intestinal microbiota. We performed a monocenter, randomized open-label clinical trial in 22 healthy volunteers treated by intravenous ceftriaxone (1 g/24 h) or cefotaxime (1 g/8 h) for 3 days. We collected fecal samples for phenotypic analyses, 16S rRNA gene profiling, and measurement of the antibiotic concentration and compared the groups for the evolution of microbial counts and indices of bacterial diversity over time. Plasma samples were drawn at day 3 for pharmacokinetic analysis. The emergence of 3rd-generation-cephalosporin-resistant Gram-negative enteric bacilli (Enterobacterales), Enterococcus spp., or noncommensal microorganisms was not significantly different between the groups. Both antibiotics reduced the counts of total Gram-negative enteric bacilli and decreased the bacterial diversity, but the differences between the groups were not significant. All but one volunteer from each group exhibited undetectable levels of antibiotic in feces. Plasma pharmacokinetic endpoints were not correlated to alteration of the bacterial diversity of the gut. Both antibiotics markedly impacted the intestinal microbiota, but no significant differences were detected when standard clinical doses were administered for 3 days. This might be related to the similar daily amounts of antibiotics excreted through the bile using a clinical regimen. (This study has been registered at ClinicalTrials.gov under identifier NCT02659033.).


Assuntos
Antibacterianos/uso terapêutico , Cefotaxima/farmacologia , Ceftriaxona/uso terapêutico , Microbioma Gastrointestinal/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Adolescente , Adulto , Cefalosporinas/uso terapêutico , Fezes , Feminino , Infecções por Bactérias Gram-Negativas/tratamento farmacológico , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , RNA Ribossômico 16S/efeitos dos fármacos , Adulto Jovem
7.
J Infect Dis ; 217(4): 628-636, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29186529

RESUMO

Background: Antibiotics are life-saving drugs but severely affect the gut microbiome with short-term consequences including diarrhea and selection of antibiotic-resistant bacteria. Long-term links to allergy and obesity are also suggested. We devised a product, DAV132, and previously showed its ability to deliver a powerful adsorbent, activated charcoal, in the late ileum of human volunteers. Methods: We performed a randomized controlled trial in 28 human volunteers treated with a 5-day clinical regimen of the fluoroquinolone antibiotic moxifloxacin in 2 parallel groups, with or without DAV132 coadministration. Two control goups of 8 volunteers each receiving DAV132 alone, or a nonactive substitute, were added. Results: The coadministration of DAV132 decreased free moxifloxacin fecal concentrations by 99%, while plasmatic levels were unaffected. Shotgun quantitative metagenomics showed that the richness and composition of the intestinal microbiota were largely preserved in subjects co-treated with DAV132 in addition to moxifloxacin. No adverse effect was observed. In addition, DAV132 efficiently adsorbed a wide range of clinically relevant antibiotics ex vivo. Conclusions: DAV132 was highly effective to protect the gut microbiome of moxifloxacin-treated healthy volunteers and may constitute a clinical breakthrough by preventing adverse health consequences of a wide range of antibiotic treatments. Clinical Trials Registration: NCT02176005.


Assuntos
Antibacterianos/administração & dosagem , Carvão Vegetal/administração & dosagem , Microbioma Gastrointestinal/efeitos dos fármacos , Microbiota/efeitos dos fármacos , Moxifloxacina/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibacterianos/análise , Fezes/química , Fezes/microbiologia , Feminino , Voluntários Saudáveis , Humanos , Masculino , Metagenômica , Pessoa de Meia-Idade , Moxifloxacina/análise , Resultado do Tratamento , Adulto Jovem
8.
Artigo em Inglês | MEDLINE | ID: mdl-30061286

RESUMO

Antibiotic disruption of the intestinal microbiota favors colonization by Clostridium difficile Using a charcoal-based adsorbent to decrease intestinal antibiotic concentrations, we studied the relationship between antibiotic concentrations in feces and the intensity of dysbiosis and quantified the link between this intensity and mortality. We administered either moxifloxacin (n = 70) or clindamycin (n = 60) to hamsters by subcutaneous injection from day 1 (D1) to D5 and challenged them with a C. difficile toxigenic strain at D3 Hamsters received various doses of a charcoal-based adsorbent, DAV131A, to modulate intestinal antibiotic concentrations. Gut dysbiosis was evaluated at D0 and D3 using diversity indices determined from 16S rRNA gene profiling. Survival was monitored until D16 We analyzed the relationship between fecal antibiotic concentrations and dysbiosis at the time of C. difficile challenge and studied their capacity to predict subsequent death of the animals. Increasing doses of DAV131A reduced fecal concentrations of both antibiotics, lowered dysbiosis, and increased survival from 0% to 100%. Mortality was related to the level of dysbiosis (P < 10-5 for the change of Shannon index in moxifloxacin-treated animals and P < 10-9 in clindamycin-treated animals). The Shannon diversity index and unweighted UniFrac distance best predicted death, with areas under the receiver operating curve (ROC) of 0.89 (95% confidence interval [CI], 0.82, 0.95) and 0.95 (0.90, 0.98), respectively. Altogether, moxifloxacin and clindamycin disrupted the diversity of the intestinal microbiota with a dependency on the DAV131A dose; mortality after C. difficile challenge was related to the intensity of dysbiosis in similar manners with the two antibiotics.


Assuntos
Antibacterianos/efeitos adversos , Infecções por Clostridium/tratamento farmacológico , Infecções por Clostridium/mortalidade , Disbiose/induzido quimicamente , Animais , Antibacterianos/uso terapêutico , Clindamicina/uso terapêutico , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/patogenicidade , Cricetinae , Disbiose/mortalidade , Microbioma Gastrointestinal/efeitos dos fármacos , Masculino , Mesocricetus , Moxifloxacina/uso terapêutico
9.
Mol Biol Evol ; 33(4): 898-914, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26613786

RESUMO

In the context of the great concern about the impact of human activities on the environment, we studied 403 commensal Escherichia coli/Escherichia clade strains isolated from several animal and human populations that have variable contacts to one another. Multilocus sequence typing (MLST) showed a decrease of diversity 1) in strains isolated from animals that had an increasing contact with humans and 2) in all strains that had increased antimicrobial resistance. A specific B1 phylogroup clonal complex (CC87, Institut Pasteur schema nomenclature) of animal origin was identified and characterized as being responsible for the increased antimicrobial resistance prevalence observed in strains from the environments with a high human-mediated antimicrobial pressure. CC87 strains have a high capacity of acquiring and disseminating resistance genes with specific metabolic and genetic determinants as demonstrated by high-throughput sequencing and phenotyping. They are good mouse gut colonizers but are not virulent. Our data confirm the predominant role of human activities in the emergence of antimicrobial resistance in the environmental bacterial strains and unveil a particular E. coli clonal complex of animal origin capable of spreading antimicrobial resistance to other members of microbial communities.


Assuntos
Farmacorresistência Bacteriana Múltipla/genética , Escherichia coli/genética , Variação Genética , Animais , Anti-Infecciosos/efeitos adversos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Proteínas de Escherichia coli/genética , Genótipo , Humanos , Camundongos , Tipagem de Sequências Multilocus , Filogenia
10.
Artigo em Inglês | MEDLINE | ID: mdl-28739791

RESUMO

Lowering the gut exposure to antibiotics during treatments can prevent microbiota disruption. We evaluated the effects of an activated charcoal-based adsorbent, DAV131A, on the fecal free moxifloxacin concentration and mortality in a hamster model of moxifloxacin-induced Clostridium difficile infection. A total of 215 hamsters receiving moxifloxacin subcutaneously (day 1 [D1] to D5) were orally infected at D3 with C. difficile spores. They received various doses (0 to 1,800 mg/kg of body weight/day) and schedules (twice a day [BID] or three times a day [TID]) of DAV131A (D1 to D8). Moxifloxacin concentrations and C. difficile counts were determined at D3, and mortality was determined at D12 We compared mortality rates, moxifloxacin concentrations, and C. difficile counts according to DAV131A regimen and modeled the links between DAV131A regimen, moxifloxacin concentration, and mortality. All hamsters that received no DAV131A died, but none of those that received 1,800 mg/kg/day died. Significant dose-dependent relationships between DAV131A dose and (i) mortality, (ii) moxifloxacin concentration, and (iii) C. difficile count were evidenced. Mathematical modeling suggested that (i) lowering the moxifloxacin concentration at D3, which was 58 µg/g (95% confidence interval [CI] = 50 to 66 µg/g) without DAV131A, to 17 µg/g (14 to 21 µg/g) would reduce mortality by 90%; and (ii) this would be achieved with a daily DAV131A dose of 703 mg/kg (596 to 809 mg/kg). In this model of C. difficile infection, DAV131A reduced mortality in a dose-dependent manner by decreasing the fecal free moxifloxacin concentration.


Assuntos
Clostridioides difficile/patogenicidade , Infecções por Clostridium/induzido quimicamente , Disbiose/induzido quimicamente , Enterocolite Pseudomembranosa/induzido quimicamente , Fluoroquinolonas/efeitos adversos , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Animais , Carvão Vegetal/farmacologia , Infecções por Clostridium/tratamento farmacológico , Cricetinae , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Enterocolite Pseudomembranosa/tratamento farmacológico , Enterocolite Pseudomembranosa/mortalidade , Fluoroquinolonas/farmacologia , Trato Gastrointestinal/microbiologia , Moxifloxacina
11.
Appl Environ Microbiol ; 83(13)2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28411228

RESUMO

To get insights into the temporal pattern of commensal Escherichia coli populations, we sampled the feces of four healthy cows from the same herd in the Hwange District of Zimbabwe daily over 25 days. The cows had not received antibiotic treatment during the previous 3 months. We performed viable E. coli counts and characterized the 326 isolates originating from the 98 stool samples at a clonal level, screened them for stx and eae genes, and tested them for their antibiotic susceptibilities. We observed that E. coli counts and dominant clones were different among cows, and very few clones were shared. No clone was shared by three or four cows. Clone richness and evenness were not different between cows. Within each host, the variability in the E. coli count was evidenced between days, and no clone was found to be dominant during the entire sampling period, suggesting the existence of clonal interference. Dominant clones tended to persist longer than subdominant ones and were mainly from phylogenetic groups A and B1. Five E. coli clones were found to contain both the stx1 and stx2 genes, representing 6.3% of the studied isolates. All cows harbored at least one Shiga toxin-producing E. coli (STEC) strain. Resistance to tetracycline, penicillins, trimethoprim, and sulfonamides was rare and observed in three clones that were shed at low levels in two cows. This study highlights the fact that the commensal E. coli population, including the STEC population, is host specific, is highly dynamic over a short time frame, and rarely carries antibiotic resistance determinants in the absence of antibiotic treatment.IMPORTANCE The literature about the dynamics of commensal Escherichia coli populations is very scarce. Over 25 days, we followed the total E. coli counts daily and characterized the sampled clones in the feces of four cows from the same herd living in the Hwange District of Zimbabwe. This study deals with the day-to-day dynamics of both quantitative and qualitative aspects of E. coli commensal populations, with a focus on both Shiga toxin-producing E. coli and antibiotic-resistant E. coli strains. We show that the structure of these commensal populations was highly specific to the host, even though the cows ate and roamed together, and was highly dynamic between days. Such data are of importance to understand the ecological forces that drive the dynamics of the emergence of E. coli clones of particular interest within the gastrointestinal tract and their transmission between hosts.


Assuntos
Bovinos/microbiologia , Escherichia coli/fisiologia , Animais , Bovinos/fisiologia , Escherichia coli/classificação , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Fezes/microbiologia , Filogenia , Toxina Shiga/genética , Toxina Shiga/metabolismo , Simbiose , Zimbábue
12.
J Water Health ; 15(2): 175-184, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28362299

RESUMO

Clinically relevant antimicrobial resistant bacteria, genetic resistance elements, and antibiotic residues (so-called AMR) from human and animal waste are abundantly present in environmental samples. This presence could lead to human exposure to AMR. In 2015, the World Health Organization (WHO) developed a Global Action Plan for Antimicrobial Resistance with one of its strategic objectives being to strengthen knowledge through surveillance and research. With respect to a strategic research agenda on water, sanitation and hygiene and AMR, WHO organized a workshop to solicit input by scientists and other stakeholders. The workshop resulted in three main conclusions. The first conclusion was that guidance is needed on how to reduce the spread of AMR to humans via the environment and to introduce effective intervention measures. Second, human exposure to AMR via water and its health impact should be investigated and quantified, in order to compare with other human exposure routes, such as direct transmission or via food consumption. Finally, a uniform and global surveillance strategy that complements existing strategies and includes analytical methods that can be used in low-income countries too, is needed to monitor the magnitude and dissemination of AMR.


Assuntos
Anti-Infecciosos/farmacologia , Resistência Microbiana a Medicamentos , Saneamento , Microbiologia da Água , Humanos , Saneamento/normas , Microbiologia da Água/normas , Organização Mundial da Saúde
13.
Clin Infect Dis ; 63(8): 1087-1093, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27439526

RESUMO

Antimicrobial use in food animals selects for antimicrobial resistance in bacteria, which can spread to people. Reducing use of antimicrobials-particularly those deemed to be critically important for human medicine-in food production animals continues to be an important step for preserving the benefits of these antimicrobials for people. The World Health Organization ranking of antimicrobials according to their relative importance in human medicine was recently updated. Antimicrobials considered the highest priority among the critically important antimicrobials were quinolones, third- and fourth-generation cephalosporins, macrolides and ketolides, and glycopeptides. The updated ranking allows stakeholders in the agriculture sector and regulatory agencies to focus risk management efforts on drugs used in food animals that are the most important to human medicine. In particular, the current large-scale use of fluoroquinolones, macrolides, and third-generation cephalosporins and any potential use of glycopeptides and carbapenems need to be addressed urgently.


Assuntos
Anti-Infecciosos , Resistência Microbiana a Medicamentos , Controle de Medicamentos e Entorpecentes , Inocuidade dos Alimentos , Animais , Anti-Infecciosos/farmacologia , Anti-Infecciosos/uso terapêutico , Humanos , Gestão de Riscos , Organização Mundial da Saúde
14.
Antimicrob Agents Chemother ; 60(1): 507-14, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26552974

RESUMO

Epidemiological and individual risk factors for colonization by enterobacteria producing extended-spectrum beta-lactamases (E-ESBL) have been studied extensively, but whether such colonization is associated with significant changes in the composition of the rest of the microbiota is still unknown. To address this issue, we assessed in an isolated Amerindian Guianese community whether intestinal carriage of E-ESBL was associated with specificities in gut microbiota using metagenomic and metatranscriptomic approaches. While the richness of taxa of the active microbiota of carriers was similar to that of noncarriers, the taxa were less homogeneous. In addition, species of four genera, Desulfovibrio, Oscillospira, Parabacteroides, and Coprococcus, were significantly more abundant in the active microbiota of noncarriers than in the active microbiota of carriers, whereas such was the case only for species of Desulfovibrio and Oscillospira in the total microbiota. Differential genera in noncarrier microbiota could either be associated with resistance to colonization or be the consequence of the colonization by E-ESBL.


Assuntos
Infecções por Enterobacteriaceae/epidemiologia , Enterobacteriaceae/genética , Microbioma Gastrointestinal/genética , Genes Bacterianos , Indígenas Norte-Americanos , Transcriptoma , beta-Lactamases/genética , Adulto , Idoso , Portador Sadio , Desulfovibrio/genética , Desulfovibrio/isolamento & purificação , Enterobacteriaceae/classificação , Enterobacteriaceae/enzimologia , Enterobacteriaceae/isolamento & purificação , Infecções por Enterobacteriaceae/microbiologia , Fezes/microbiologia , Feminino , Guiana Francesa/epidemiologia , Expressão Gênica , Humanos , Masculino , Metagenoma , Pessoa de Meia-Idade , Filogenia , RNA Ribossômico 16S/genética , Análise de Sequência de DNA , beta-Lactamases/metabolismo
16.
Clin Infect Dis ; 61(4): 593-600, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25904368

RESUMO

BACKGROUND: Multidrug-resistant Enterobacteriaceae (MRE) are widespread in the community, especially in tropical regions. Travelers are at risk of acquiring MRE in these regions, but the precise extent of the problem is not known. METHODS: From February 2012 to April 2013, travelers attending 6 international vaccination centers in the Paris area prior to traveling to tropical regions were asked to provide a fecal sample before and after their trip. Those found to have acquired MRE were asked to send fecal samples 1, 2, 3, 6, and 12 months after their return, or until MRE was no longer detected. The fecal relative abundance of MRE among all Enterobacteriaceae was determined in each carrier. RESULTS: Among 824 participating travelers, 574 provided fecal samples before and after travel and were not MRE carriers before departure. Of these, 292 (50.9%) acquired an average of 1.8 MRE. Three travelers (0.5%) acquired carbapenemase-producing Enterobacteriaceae. The acquisition rate was higher in Asia (142/196 [72.4%]) than in sub-Saharan Africa (93/195 [47.7%]) or Latin America (57/183 [31.1%]). MRE acquisition was associated with the type of travel, diarrhea, and exposure to ß-lactams during the travel. Three months after return, 4.7% of the travelers carried MRE. Carriage lasted longer in travelers returning from Asia and in travelers with a high relative abundance of MRE at return. CONCLUSIONS: MRE acquisition is very frequent among travelers to tropical regions. Travel to these regions should be considered a risk factor of MRE carriage during the first 3 months after return, but not beyond. CLINICAL TRIALS REGISTRATION: NCT01526187.


Assuntos
Portador Sadio/epidemiologia , Portador Sadio/microbiologia , Farmacorresistência Bacteriana Múltipla , Infecções por Enterobacteriaceae/epidemiologia , Infecções por Enterobacteriaceae/microbiologia , Enterobacteriaceae/isolamento & purificação , Viagem , Adolescente , Adulto , Idoso , Enterobacteriaceae/efeitos dos fármacos , Fezes/microbiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Paris/epidemiologia , Fatores de Tempo , Clima Tropical , Adulto Jovem
17.
Appl Environ Microbiol ; 82(5): 1459-1467, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26712551

RESUMO

At a human/livestock/wildlife interface, Escherichia coli populations were used to assess the risk of bacterial and antibiotic resistance dissemination between hosts. We used phenotypic and genotypic characterization techniques to describe the structure and the level of antibiotic resistance of E. coli commensal populations and the resistant Enterobacteriaceae carriage of sympatric African buffalo (Syncerus caffer caffer) and cattle populations characterized by their contact patterns in the southern part of Hwange ecosystem in Zimbabwe. Our results (i) confirmed our assumption that buffalo and cattle share similar phylogroup profiles, dominated by B1 (44.5%) and E (29.0%) phylogroups, with some variability in A phylogroup presence (from 1.9 to 12%); (ii) identified a significant gradient of antibiotic resistance from isolated buffalo to buffalo in contact with cattle and cattle populations expressed as the Murray score among Enterobacteriaceae (0.146, 0.258, and 0.340, respectively) and as the presence of tetracycline-, trimethoprim-, and amoxicillin-resistant subdominant E. coli strains (0, 5.7, and 38%, respectively); (iii) evidenced the dissemination of tetracycline, trimethoprim, and amoxicillin resistance genes (tet, dfrA, and blaTEM-1) in 26 isolated subdominant E. coli strains between nearby buffalo and cattle populations, that led us (iv) to hypothesize the role of the human/animal interface in the dissemination of genetic material from human to cattle and toward wildlife. The study of antibiotic resistance dissemination in multihost systems and at anthropized/natural interface is necessary to better understand and mitigate its multiple threats. These results also contribute to attempts aiming at using E. coli as a tool for the identification of pathogen transmission pathway in multihost systems.


Assuntos
Farmacorresistência Bacteriana , Infecções por Escherichia coli/veterinária , Escherichia coli/classificação , Escherichia coli/efeitos dos fármacos , Animais , Antibacterianos/farmacologia , Bovinos , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Infecções por Escherichia coli/epidemiologia , Infecções por Escherichia coli/microbiologia , Variação Genética , Genótipo , Humanos , Filogenia , Zimbábue/epidemiologia
18.
Bull World Health Organ ; 93(4): 217-8, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26229185

RESUMO

This year's World Health Day is on food safety. The mass use of antibiotics in animal husbandry is contaminating the food supply with resistant bacteria, causing difficult-to-treat infections while reducing the power of antibiotics to cure human infections. Antoine Andremont talks to Fiona Fleck.


Assuntos
Criação de Animais Domésticos/métodos , Microbiologia de Alimentos , Inocuidade dos Alimentos , Animais , Antibacterianos/administração & dosagem , Bactérias/efeitos dos fármacos , Farmacorresistência Bacteriana , Humanos
19.
PLoS Comput Biol ; 10(9): e1003840, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25210849

RESUMO

Fecal excretion of antibiotics and resistant bacteria in the environment are major public health threats associated with extensive farming and modern medical care. Innovative strategies that can reduce the intestinal antibiotic concentrations during treatments are in development. However, the effect of lower exposure on the amount of resistant enterobacteria excreted has not been quantified, making it difficult to anticipate the impact of these strategies. Here, we introduce a bacterial kinetic model to capture the complex relationships between drug exposure, loss of susceptible enterobacteria and growth of resistant strains in the feces of piglets receiving placebo, 1.5 or 15 mg/kg/day ciprofloxacin, a fluoroquinolone, for 5 days. The model could well describe the kinetics of drug susceptible and resistant enterobacteria observed during treatment, and up to 22 days after treatment cessation. Next, the model was used to predict the expected amount of resistant enterobacteria excreted over an average piglet's lifetime (150 days) when varying drug exposure and treatment duration. For the clinically relevant dose of 15 mg/kg/day for 5 days, the total amount of resistant enterobacteria excreted was predicted to be reduced by 75% and 98% when reducing treatment duration to 3 and 1 day treatment, respectively. Alternatively, for a fixed 5-days treatment, the level of resistance excreted could be reduced by 18%, 33%, 57.5% and 97% if 3, 5, 10 and 30 times lower levels of colonic drug concentrations were achieved, respectively. This characterization on in vivo data of the dynamics of resistance to antibiotics in the colonic flora could provide new insights into the mechanism of dissemination of resistance and can be used to design strategies aiming to reduce it.


Assuntos
Antibacterianos/farmacologia , Antibacterianos/farmacocinética , Farmacorresistência Bacteriana , Enterobacteriaceae/efeitos dos fármacos , Fezes/química , Fezes/microbiologia , Animais , Ciprofloxacina/farmacocinética , Ciprofloxacina/farmacologia , Colo/microbiologia , Fluoroquinolonas/farmacocinética , Fluoroquinolonas/farmacologia , Cinética , Modelos Biológicos , Suínos
20.
Clin Microbiol Rev ; 26(4): 744-58, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24092853

RESUMO

In the last 10 years, extended-spectrum ß-lactamase-producing enterobacteria (ESBL-E) have become one of the main challenges for antibiotic treatment of enterobacterial infections, largely because of the current CTX-M enzyme pandemic. However, most studies have focused on hospitalized patients, though today it appears that the community is strongly affected as well. We therefore decided to devote our investigation to trends in ESBL-E fecal carriage rates and comprehensively reviewed data from studies conducted on healthy populations in various parts of the world. We show that (i) community ESBL-E fecal carriage, which was unknown before the turn of the millennium, has since increased significantly everywhere, with developing countries being the most affected; (ii) intercontinental travel may have emphasized and globalized the issue; and (iii) CTX-M enzymes, especially CTX-M-15, are the dominant type of ESBL. Altogether, these results suggest that CTX-M carriage is evolving toward a global pandemic but is still insufficiently described. Only a better knowledge of its dynamics and biology will lead to further development of appropriate control measures.


Assuntos
Portador Sadio/epidemiologia , Infecções por Enterobacteriaceae/epidemiologia , Infecções por Enterobacteriaceae/microbiologia , Enterobacteriaceae/enzimologia , Enterobacteriaceae/isolamento & purificação , Fezes/microbiologia , beta-Lactamases/metabolismo , Portador Sadio/microbiologia , Infecções Comunitárias Adquiridas/epidemiologia , Infecções Comunitárias Adquiridas/microbiologia , Surtos de Doenças , Reservatórios de Doenças/microbiologia , Infecções por Enterobacteriaceae/transmissão , Humanos , Internacionalidade , Organização Mundial da Saúde , beta-Lactamases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA