Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.678
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 169(5): 849-861.e13, 2017 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-28502769

RESUMO

We examined the evolutionary history of leading multidrug resistant hospital pathogens, the enterococci, to their origin hundreds of millions of years ago. Our goal was to understand why, among the vast diversity of gut flora, enterococci are so well adapted to the modern hospital environment. Molecular clock estimation, together with analysis of their environmental distribution, phenotypic diversity, and concordance with host fossil records, place the origins of the enterococci around the time of animal terrestrialization, 425-500 mya. Speciation appears to parallel the diversification of hosts, including the rapid emergence of new enterococcal species following the End Permian Extinction. Major drivers of speciation include changing carbohydrate availability in the host gut. Life on land would have selected for the precise traits that now allow pathogenic enterococci to survive desiccation, starvation, and disinfection in the modern hospital, foreordaining their emergence as leading hospital pathogens.


Assuntos
Evolução Biológica , Enterococcus/genética , Animais , Doenças Transmissíveis Emergentes/microbiologia , Infecção Hospitalar/microbiologia , Farmacorresistência Bacteriana , Enterococcus/classificação , Enterococcus/citologia , Enterococcus/efeitos dos fármacos , Especiação Genética , Interações Hospedeiro-Patógeno , Larva/microbiologia , Mariposas/crescimento & desenvolvimento , Mariposas/microbiologia , Filogenia , RNA Ribossômico 16S/genética
2.
Nature ; 607(7919): 563-570, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35831502

RESUMO

Gut commensal bacteria with the ability to translocate across the intestinal barrier can drive the development of diverse immune-mediated diseases1-4. However, the key factors that dictate bacterial translocation remain unclear. Recent studies have revealed that gut microbiota strains can adapt and evolve throughout the lifetime of the host5-9, raising the possibility that changes in individual commensal bacteria themselves over time may affect their propensity to elicit inflammatory disease. Here we show that within-host evolution of the model gut pathobiont Enterococcus gallinarum facilitates bacterial translocation and initiation of inflammation. Using a combination of in vivo experimental evolution and comparative genomics, we found that E. gallinarum diverges into independent lineages adapted to colonize either luminal or mucosal niches in the gut. Compared with ancestral and luminal E. gallinarum, mucosally adapted strains evade detection and clearance by the immune system, exhibit increased translocation to and survival within the mesenteric lymph nodes and liver, and induce increased intestinal and hepatic inflammation. Mechanistically, these changes in bacterial behaviour are associated with non-synonymous mutations or insertion-deletions in defined regulatory genes in E. gallinarum, altered microbial gene expression programs and remodelled cell wall structures. Lactobacillus reuteri also exhibited broadly similar patterns of divergent evolution and enhanced immune evasion in a monocolonization-based model of within-host evolution. Overall, these studies define within-host evolution as a critical regulator of commensal pathogenicity that provides a unique source of stochasticity in the development and progression of microbiota-driven disease.


Assuntos
Bactérias , Translocação Bacteriana , Evolução Biológica , Microbioma Gastrointestinal , Fígado , Bactérias/genética , Bactérias/imunologia , Bactérias/patogenicidade , Translocação Bacteriana/genética , Parede Celular/genética , Enterococcus/genética , Enterococcus/imunologia , Microbioma Gastrointestinal/genética , Genômica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamação/microbiologia , Inflamação/patologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/imunologia , Fígado/microbiologia , Fígado/patologia , Linfonodos/microbiologia , Mutação , Processos Estocásticos , Simbiose/genética , Simbiose/imunologia
3.
Proc Natl Acad Sci U S A ; 121(10): e2310852121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38416678

RESUMO

Enterococci are gut microbes of most land animals. Likely appearing first in the guts of arthropods as they moved onto land, they diversified over hundreds of millions of years adapting to evolving hosts and host diets. Over 60 enterococcal species are now known. Two species, Enterococcus faecalis and Enterococcus faecium, are common constituents of the human microbiome. They are also now leading causes of multidrug-resistant hospital-associated infection. The basis for host association of enterococcal species is unknown. To begin identifying traits that drive host association, we collected 886 enterococcal strains from widely diverse hosts, ecologies, and geographies. This identified 18 previously undescribed species expanding genus diversity by >25%. These species harbor diverse genes including toxins and systems for detoxification and resource acquisition. Enterococcus faecalis and E. faecium were isolated from diverse hosts highlighting their generalist properties. Most other species showed a more restricted distribution indicative of specialized host association. The expanded species diversity permitted the Enterococcus genus phylogeny to be viewed with unprecedented resolution, allowing features to be identified that distinguish its four deeply rooted clades, and the entry of genes associated with range expansion such as B-vitamin biosynthesis and flagellar motility to be mapped to the phylogeny. This work provides an unprecedentedly broad and deep view of the genus Enterococcus, including insights into its evolution, potential new threats to human health, and where substantial additional enterococcal diversity is likely to be found.


Assuntos
Enterococcus faecium , Infecções por Bactérias Gram-Positivas , Animais , Humanos , Enterococcus/genética , Antibacterianos/farmacologia , Enterococcus faecium/genética , Enterococcus faecalis/genética , Filogenia , Testes de Sensibilidade Microbiana , Farmacorresistência Bacteriana
4.
Clin Microbiol Rev ; 37(2): e0012123, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38466110

RESUMO

SUMMARYEnterococci are a diverse group of Gram-positive bacteria that are typically found as commensals in humans, animals, and the environment. Occasionally, they may cause clinically relevant diseases such as endocarditis, septicemia, urinary tract infections, and wound infections. The majority of clinical infections in humans are caused by two species: Enterococcus faecium and Enterococcus faecalis. However, there is an increasing number of clinical infections caused by non-faecium non-faecalis (NFF) enterococci. Although NFF enterococcal species are often overlooked, studies have shown that they may harbor antimicrobial resistance (AMR) genes and virulence factors that are found in E. faecium and E. faecalis. In this review, we present an overview of the NFF enterococci with a particular focus on human clinical manifestations, epidemiology, virulence genes, and AMR genes.


Assuntos
Infecções por Bactérias Gram-Positivas , Fatores de Virulência , Humanos , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Fatores de Virulência/genética , Animais , Farmacorresistência Bacteriana , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Enterococcus/patogenicidade , Enterococcus/efeitos dos fármacos , Enterococcus/genética , Virulência
5.
PLoS Pathog ; 19(6): e1011424, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37267422

RESUMO

Insertion sequences (IS) are simple transposons implicated in the genome evolution of diverse pathogenic bacterial species. Enterococci have emerged as important human intestinal pathogens with newly adapted virulence potential and antibiotic resistance. These genetic features arose in tandem with large-scale genome evolution mediated by mobile elements. Pathoadaptation in enterococci is thought to be mediated in part by the IS element IS256 through gene inactivation and recombination events. However, the regulation of IS256 and the mechanisms controlling its activation are not well understood. Here, we adapt an IS256-specfic deep sequencing method to describe how chronic lytic phage infection drives widespread diversification of IS256 in E. faecalis and how antibiotic exposure is associated with IS256 diversification in E. faecium during a clinical human infection. We show through comparative genomics that IS256 is primarily found in hospital-adapted enterococcal isolates. Analyses of IS256 transposase gene levels reveal that IS256 mobility is regulated at the transcriptional level by multiple mechanisms in E. faecalis, indicating tight control of IS256 activation in the absence of selective pressure. Our findings reveal that stressors such as phages and antibiotic exposure drives rapid genome-scale transposition in the enterococci. IS256 diversification can therefore explain how selective pressures mediate evolution of the enterococcal genome, ultimately leading to the emergence of dominant nosocomial lineages that threaten human health.


Assuntos
Enterococcus faecium , Enterococcus , Humanos , Enterococcus/genética , Elementos de DNA Transponíveis/genética , Antibacterianos/farmacologia , Resistência Microbiana a Medicamentos , Enterococcus faecalis/genética
6.
BMC Genomics ; 25(1): 718, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39054474

RESUMO

INTRODUCTION: Our understanding of particular gut microbiota members such as Bifidobacterium and Enterococcus in low-middle-income countries remains very limited, particularly early life strain-level beneficial traits. This study addresses this gap by exploring a collection of bacterial strains isolated from the gut of Zimbabwean infants; comparing their genomic characteristics with strains isolated from infants across North America, Europe, and other regions of Africa. MATERIALS AND METHOD: From 110 infant stool samples collected in Harare, Zimbabwe, 20 randomly selected samples were used to isolate dominant early-life gut microbiota members Bifidobacterium and Enterococcus. Isolated strains were subjected to whole genome sequencing and bioinformatics analysis including functional annotation of carbohydrates, human milk oligosaccharide (HMO) and protein degradation genes and clusters, and the presence of antibiotic resistance genes (ARGs). RESULTS: The study observed some location-based clustering within the main five identified taxonomic groups. Furthermore, there were varying and overall species-specific numbers of genes belonging to different GH families encoded within the analysed dataset. Additionally, distinct strain- and species-specific variances were identified in the potential of Bifidobacterium for metabolizing HMOs. Analysis of putative protease activity indicated a consistent presence of gamma-glutamyl hydrolases in Bifidobacterium, while Enterococcus genomes exhibited a high abundance of aspartyl peptidases. Both genera harboured resistance genes against multiple classes of antimicrobial drugs, with Enterococcus genomes containing a higher number of ARGs compared to Bifidobacterium, on average. CONCLUSION: This study identified promising probiotic strains within Zimbabwean isolates, offering the potential for early-life diet and microbial therapies. However, the presence of antibiotic resistance genes in infant-associated microbes raises concerns for infection risk and next-stage probiotic development. Further investigation in larger cohorts, particularly in regions with limited existing data on antibiotic and probiotic use, is crucial to validate these initial insights. IMPACT STATEMENT: This research represents the first investigation of its kind in the Zimbabwean context, focusing on potential probiotic strains within the early-life gut microbiota. By identifying local probiotic strains, this research can contribute to the development of probiotic interventions that are tailored to the Zimbabwean population, which can help address local health challenges and promote better health outcomes for infants. Another essential aspect of the study is the investigation of antimicrobial resistance genes present in Zimbabwean bacterial strains. Antimicrobial resistance is a significant global health concern, and understanding the prevalence and distribution of resistance genes in different regions can help inform public health policies and interventions.


Assuntos
Bifidobacterium , Enterococcus , Microbioma Gastrointestinal , Humanos , Zimbábue , Lactente , Microbioma Gastrointestinal/genética , Enterococcus/genética , Enterococcus/efeitos dos fármacos , Enterococcus/isolamento & purificação , Bifidobacterium/genética , Bifidobacterium/isolamento & purificação , Bifidobacterium/efeitos dos fármacos , Genômica , Genoma Bacteriano , Fezes/microbiologia , Sequenciamento Completo do Genoma , Estudos de Coortes , Filogenia
7.
J Antimicrob Chemother ; 79(5): 997-1005, 2024 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-38501366

RESUMO

BACKGROUND: VRE are increasingly described worldwide. Screening of hospitalized patients at risk for VRE carriage is mandatory to control their dissemination. Here, we have developed the Bfast [VRE Panel] PCR kit, a rapid and reliable quantitative PCR assay for detection of vanA, vanB, vanD and vanM genes, from solid and liquid cultures adaptable to classical and ultrafast real-time PCR platforms. METHODS: Validation was carried out on 133 well characterized bacterial strains, including 108 enterococci of which 64 were VRE. Analytical performances were determined on the CFX96 Touch (Bio-Rad) and Chronos Dx (BforCure), an ultrafast qPCR machine. Widely used culture plates and broths for enterococci selection/growth were tested. RESULTS: All targeted van alleles (A, B, D and M) were correctly detected without cross-reactivity with other van genes (C, E, G, L and N) and no interference with the different routinely used culture media. A specificity and sensitivity of 100% and 99.7%, respectively, were determined, with limits of detection ranging from 21 to 238 cfu/reaction depending on the targets. The Bfast [VRE Panel] PCR kit worked equally well on the CFX and Chronos Dx platforms, with differences in multiplexing capacities (five and four optical channels, respectively) and in turnaround time (45 and 16 minutes, respectively). CONCLUSIONS: The Bfast [VRE Panel] PCR kit is robust, easy to use, rapid and easily implementable in clinical microbiology laboratories for ultra-rapid confirmation of the four main acquired van genes. Its features, especially on Chronos Dx, seem to be unmatched compared to other tools for screening of VRE.


Assuntos
Reação em Cadeia da Polimerase em Tempo Real , Sensibilidade e Especificidade , Resistência a Vancomicina , Enterococos Resistentes à Vancomicina , Humanos , Reação em Cadeia da Polimerase em Tempo Real/métodos , Resistência a Vancomicina/genética , Enterococos Resistentes à Vancomicina/genética , Enterococos Resistentes à Vancomicina/isolamento & purificação , Enterococos Resistentes à Vancomicina/efeitos dos fármacos , Enterococcus/genética , Enterococcus/efeitos dos fármacos , Enterococcus/isolamento & purificação , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/diagnóstico , Proteínas de Bactérias/genética , Fatores de Tempo , Genes Bacterianos/genética
8.
PLoS Pathog ; 18(9): e1010829, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36103556

RESUMO

Multidrug-resistant (MDR) Enterococcus faecalis are major causes of hospital-acquired infections. Numerous clinical strains of E. faecalis harbor a large pathogenicity island that encodes enterococcal surface protein (Esp), which is suggested to promote biofilm production and virulence, but this remains controversial. To resolve this issue, we characterized the Esp N-terminal region, the portion implicated in biofilm production. Small angle X-ray scattering indicated that the N-terminal region had a globular head, which consisted of two DEv-Ig domains as visualized by X-ray crystallography, followed by an extended tail. The N-terminal region was not required for biofilm production but instead significantly strengthened biofilms against mechanical or degradative disruption, greatly increasing retention of Enterococcus within biofilms. Biofilm strengthening required low pH, which resulted in Esp unfolding, aggregating, and forming amyloid-like structures. The pH threshold for biofilm strengthening depended on protein stability. A truncated fragment of the first DEv-Ig domain, plausibly generated by a host protease, was the least stable and sufficient to strengthen biofilms at pH ≤ 5.0, while the entire N-terminal region and intact Esp on the enterococcal surface was more stable and required a pH ≤ 4.3. These results suggested a virulence role of Esp in strengthening enterococcal biofilms in acidic abiotic or host environments.


Assuntos
Infecções por Bactérias Gram-Positivas , Proteínas de Membrana , Proteínas de Bactérias/metabolismo , Biofilmes , Enterococcus/genética , Enterococcus/metabolismo , Enterococcus faecalis , Humanos , Proteínas de Membrana/metabolismo , Peptídeo Hidrolases/metabolismo
9.
BMC Microbiol ; 24(1): 98, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38528458

RESUMO

OBJECTIVE: The association between heart failure (HF) and intestinal inflammation caused by a disturbed intestinal microbiota in infants with congenital heart disease (CHD) was investigated. METHODS: Twenty infants with HF and CHD who were admitted to our hospital between October 2021 and March 2022 were included in this study. Twenty age- and sex-matched infants without HF at our hospital were selected as the control group. Faecal samples were obtained from each participant and analysed by enzyme-linked immunoassay and 16 S rDNA sequencing to assess intestinal inflammatory factors and the microbiota. RESULTS: The levels of intestinal inflammatory factors, including IL-1ß, IL-4, IL-6, IL-17 A and TNF-α, were greatly increased, while the levels of IL-10 were significantly decreased in the HF group compared to the control group (p < 0.05). The intestinal microbial diversity of patients in the HF group was markedly lower than that in the control group (p < 0.05). The abundance of Enterococcus was significantly increased in the HF group compared to the control group (p < 0.05), but the abundance of Bifidobacterium was significantly decreased in the HF group compared to the control group (p < 0.05). The diversity of the intestinal microbiota was negatively correlated with the levels of IL-1ß, IL-4, IL-6 and TNF-α in the intestinal tract but was positively correlated with that of IL-10. The abundance of Enterococcus was positively associated with the levels of IL-1ß, IL-4, IL-6 and TNF-α in the intestinal tract but was negatively correlated with that of IL-10. NT-proBNP was positively associated with the levels of IL-1ß, IL-4, IL-6 and TNF-α in the HF group but was negatively correlated with that of IL-10. The heart function score was positively associated with the levels of IL-1ß, IL-4, IL-6 and TNF-α in the HF group but was negatively correlated with that of IL-10. CONCLUSIONS: Infants with CHD-related HF had a disordered intestinal microbiota, decreased diversity of intestinal microbes, increased levels of pathogenic bacteria and decreased levels of beneficial bacteria. The increased abundance of Enterococcus and the significant decrease in the diversity of the intestinal microbiota may exacerbate the intestinal inflammatory response, which may be associated with the progression of HF.


Assuntos
Cardiopatias Congênitas , Insuficiência Cardíaca , Lactente , Humanos , Interleucina-10 , Fator de Necrose Tumoral alfa , Interleucina-6 , Interleucina-4 , Insuficiência Cardíaca/complicações , Cardiopatias Congênitas/complicações , Enterococcus/genética , Inflamação
10.
Eur J Clin Microbiol Infect Dis ; 43(8): 1645-1650, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38811483

RESUMO

E. innesii is a recently described Enterococcus species which may be difficult to differentiate from the more common E. casseliflavus. We present the first clinical report of invasive E. innesii infection, featuring two cases of biliary sepsis. Whole genome sequencing confirmed the taxonomic assignment and the presence of vanC-4. Analysis of public genomes identified 13 deposited E. innesii and 13 deposited E. casselifalvus/E.gallinarum genomes which could be reassigned as E. innesii. Improved laboratory diagnosis of E. innesii is expected to generate additional data concerning its clinical relevance and support the future diagnosis and treatment of this uncommon pathogen.


Assuntos
Enterococcus , Infecções por Bactérias Gram-Positivas , Humanos , Enterococcus/genética , Enterococcus/classificação , Enterococcus/isolamento & purificação , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/diagnóstico , Masculino , Sequenciamento Completo do Genoma , Genoma Bacteriano , Idoso , Feminino , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Pessoa de Meia-Idade , Sepse/microbiologia
11.
Microbiol Immunol ; 68(8): 254-266, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38873884

RESUMO

To prevent nosocomial infection, it is important to screen for potential vancomycin-resistant Enterococcus (VRE) among patients. In this study, we analyzed enterococcal isolates from inpatients in one hospital without any apparent outbreak of VRE. Enterococcal isolates were collected from inpatients at Hiroshima University Hospital from April 1 to June 30, 2021 using selective medium for Enterococci. Multilocus sequence typing, antimicrobial susceptibility testing, and whole-genome sequencing were performed. A total of 164 isolates, including Enterococcus faecium (41 isolates), Enterococcus faecalis (80 isolates), Enterococcus raffinosus (11 isolates), Enterococcus casseliflavus (nine isolates), Enterococcus avium (12 isolates), Enterococcus lactis (eight isolates), Enterococcus gallinarum (two isolates), and Enterococcus malodoratus (one isolate), were analyzed. We found one vanA-positive E. faecium, which was already informed when the patient was transferred to the hospital, nine vanC-positive E. casseliflavus, and two vanC-positive E. gallinarum. E. faecium isolates showed resistance to ampicillin (95.1%), imipenem (95.1%), and levofloxacin (87.8%), and E. faecalis isolates showed resistance to minocycline (49.4%). Ampicillin- and levofloxacin-resistant E. faecium had multiple mutations in penicillin-binding protein 5 (PBP5) (39/39 isolates) and ParC/GyrA (21/36 isolates), respectively. E. raffinosus showed resistance to ampicillin (81.8%), imipenem (45.5%), and levofloxacin (45.5%), and E. lactis showed resistance to ampicillin (37.5%) and imipenem (50.0%). The linezolid resistance genes optrA and cfr(B) were found only in one isolate of E. faecalis and E. raffinosus, respectively. This study, showing the status of enterococci infection in hospitalized patients, is one of the important information when considering nosocomial infection control of VRE.


Assuntos
Antibacterianos , Infecções por Bactérias Gram-Positivas , Testes de Sensibilidade Microbiana , Sequenciamento Completo do Genoma , Humanos , Japão/epidemiologia , Antibacterianos/farmacologia , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/epidemiologia , Pacientes Internados , Infecção Hospitalar/microbiologia , Infecção Hospitalar/epidemiologia , Genoma Bacteriano/genética , Enterococos Resistentes à Vancomicina/genética , Enterococos Resistentes à Vancomicina/efeitos dos fármacos , Enterococos Resistentes à Vancomicina/isolamento & purificação , Tipagem de Sequências Multilocus , Surtos de Doenças , Enterococcus/genética , Enterococcus/efeitos dos fármacos , Enterococcus/isolamento & purificação , Enterococcus/classificação , Vancomicina/farmacologia
12.
Avian Pathol ; 53(4): 312-320, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38525653

RESUMO

Worldwide outbreaks make infections with pathogenic strains of Enterococcus cecorum (EC) one of the most important diseases in the broiler industry. Although research has increased knowledge about the pathogen, the transmission is not fully understood. Samples from different locations were collected from two broiler farms in Germany over a total of six production cycles. Samples were collected at days 1, 5, 10, 15, 21, 27, 34, 41 post-hatch and after cleaning and disinfection (C&D). A total of 1017 samples were collected from 25 different locations on the farms. Samples were analysed in the laboratory for EC by quantitative real-time PCR. Overall, 7.5% of the samples were positive. The probabilities for positive and negative samples did not differ between the farms. The number of findings differed significantly between the cycles. Compared to other samples, the chances of detecting EC in faecal samples were significantly higher. Most positive samples were found in the last week of the production periods, indicating an accumulation of EC in the barn environment. After C&D, positive PCR results were obtained in four out of 14 locations. A re-introduction from contaminated environment seemed possible. However, one pooled faecal sample was positive 1 day post-hatch. The locations that showed positive results after C&D and the positive faecal sample 1 day post-hatch indicated the persistence of EC in broiler houses of clinically healthy flocks that could lead to potential horizontal transmission routes. The present study detected potential EC sources and may help to improve hygienic measures to avoid transmissions.RESEARCH HIGHLIGHTSMethodology is suitable to detect EC during production and after C&D.Locations were detected that may serve as a reservoir for EC.Cycles with fewer positive samples were observed.Cleaning and disinfection had a major impact on the detection of EC.


Assuntos
Galinhas , Enterococcus , Fezes , Infecções por Bactérias Gram-Positivas , Abrigo para Animais , Doenças das Aves Domésticas , Animais , Galinhas/microbiologia , Fezes/microbiologia , Enterococcus/isolamento & purificação , Enterococcus/genética , Doenças das Aves Domésticas/microbiologia , Doenças das Aves Domésticas/epidemiologia , Infecções por Bactérias Gram-Positivas/veterinária , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/epidemiologia , Infecções por Bactérias Gram-Positivas/transmissão , Alemanha/epidemiologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Desinfecção , Microbiologia Ambiental
13.
J Appl Microbiol ; 135(8)2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-39081072

RESUMO

AIMS: Antibiotic resistance is a global health crisis. Roughly two-thirds of all antibiotics used are in production animals, which have the potential to impact the development of antibiotic resistance in bacterial pathogens of humans. There is little visibility on the extent of antibiotic resistance in the Australian food chain. This study sought to establish the incidence of antibiotic resistance among enterococci from poultry in Victoria. METHODS AND RESULTS: In 2016, poultry from a Victorian processing facility were swabbed immediately post-slaughter and cultured for Enterococcus species. All isolates recovered were speciated and tested for antibiotic susceptibility to 12 antibiotics following the Clinical Laboratory Standards Institute guidelines. A total of 6 farms and 207 birds were sampled and from these 285 isolates of Enterococcus were recovered. Eight different enterococcal species were identified as follows: E. faecalis (n = 122; 43%), E. faecium (n = 92; 32%), E. durans (n = 35; 12%), E. thailandicus (n = 23; 8%), E. hirae (n = 10; 3%), and a single each of E. avium, E. gallinarum, and E. mundtii. Reduced susceptibility to older classes of antibiotics was common, in particular: erythromycin (73%), rifampin (49%), nitrofurantoin (40%), and ciprofloxacin (39%). Two vancomycin-intermediate isolates were recovered, but no resistance was detected to either linezolid or gentamicin. CONCLUSIONS: The relatively high numbers of a recently described species, E. thailandicus, suggest this species might be well adapted to colonize poultry. The incidence of antibiotic resistance is lower in isolates from poultry than in human medicine in Australia. These results suggest that poultry may serve as a reservoir for older antibiotic resistance genes but is not driving the emergence of antimicrobial resistance in human bacterial pathogens. This is supported by the absence of resistance to linezolid and gentamicin.


Assuntos
Antibacterianos , Enterococcus , Testes de Sensibilidade Microbiana , Aves Domésticas , Animais , Enterococcus/isolamento & purificação , Enterococcus/efeitos dos fármacos , Enterococcus/genética , Antibacterianos/farmacologia , Aves Domésticas/microbiologia , Vitória , Incidência , Infecções por Bactérias Gram-Positivas/veterinária , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/epidemiologia , Farmacorresistência Bacteriana , Galinhas/microbiologia , Doenças das Aves Domésticas/microbiologia , Doenças das Aves Domésticas/epidemiologia
14.
Ann Clin Microbiol Antimicrob ; 23(1): 41, 2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38704577

RESUMO

BACKGROUND: Infections caused by linezolid-resistant enterococci (LRE) are clinically difficult to treat and threaten patient health. However, there is a lack of studies on long time-span LRE strains in China. For this reason, our study comprehensively revealed the resistance mechanisms of LRE strains collected in a Chinese tertiary care hospital from 2011 to 2022. METHODS: Enterococcal strains were screened and verified after retrospective analysis of microbial data. Subsequently, 65 LRE strains (61 Enterococcus faecalis and 4 Enterococcus faecium, MIC ≥ 8 µg/ml), 1 linezolid-intermediate Enterococcus faecium (MIC = 4 µg/ml) and 1 linezolid-susceptible Enterococcus faecium (MIC = 1.5 µg/ml) were submitted for whole-genome sequencing (WGS) analysis and bioinformatics analysis. RESULTS: The optrA gene was found to be the most common linezolid resistance mechanism in our study. We identified the wild-type OptrA and various OptrA variants in 98.5% of LRE strains (61 Enterococcus faecalis and 3 Enterococcus faecium). We also found one linezolid-resistant Enterococcus faecium strain carried both optrA and cfr(D) gene, while one linezolid-resistant Enterococcus faecium only harbored the poxtA gene. Most optrA genes (55/64) were located on plasmids, with impB-fexA-optrA, impB-fexA-optrA-erm(A), fexA-optrA-erm(A), and fexA-optrA segments. A minority of optrA genes (9/64) were found on chromosomes with the Tn6674-like platform. Besides, other possible linezolid resistance-associated mechanisms (mutations in the rplC and rplD genes) were also found in 26 enterococcal strains. CONCLUSIONS: Our study suggested that multiple mechanisms of linezolid resistance exist among clinical LRE strains in China.


Assuntos
Antibacterianos , Farmacorresistência Bacteriana , Enterococcus faecalis , Enterococcus faecium , Infecções por Bactérias Gram-Positivas , Linezolida , Testes de Sensibilidade Microbiana , Sequenciamento Completo do Genoma , Linezolida/farmacologia , China/epidemiologia , Humanos , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/epidemiologia , Enterococcus faecium/genética , Enterococcus faecium/efeitos dos fármacos , Farmacorresistência Bacteriana/genética , Enterococcus faecalis/efeitos dos fármacos , Enterococcus faecalis/genética , Antibacterianos/farmacologia , Estudos Retrospectivos , Enterococcus/efeitos dos fármacos , Enterococcus/genética , Proteínas de Bactérias/genética , Genoma Bacteriano , Epidemiologia Molecular , Centros de Atenção Terciária , Genômica
15.
Antonie Van Leeuwenhoek ; 117(1): 40, 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38393447

RESUMO

Enterococci are ubiquitous microorganisms in almost all environments, from the soil we step on to the food we eat. They are frequently found in naturally fermented foods, contributing to ripening through protein, lipid, and sugar metabolism. On the other hand, these organisms are also leading the current antibiotic resistance crisis. In this study, we performed whole-genome sequencing and comparative genomics of an Enterococcus faecium strain isolated from an artisanal Mexican Cotija cheese, namely QD-2. We found clear genomic differences between commensal and pathogenic strains, particularly in their carbohydrate metabolic pathways, resistance to vancomycin and other antibiotics, bacteriocin production, and bacteriophage and CRISPR content. Furthermore, a bacteriocin transcription analysis performed by RT-qPCR revealed that, at the end of the log phase, besides enterocins A and X, two putative bacteriocins not reported previously are also transcribed as a bicistronic operon in E. faecium QD-2, and are expressed 1.5 times higher than enterocin A when cultured in MRS broth.


Assuntos
Bacteriocinas , Queijo , Enterococcus faecium , Bacteriocinas/metabolismo , Enterococcus faecium/genética , Enterococcus faecium/metabolismo , Enterococcus/genética , Genômica
16.
Antonie Van Leeuwenhoek ; 117(1): 85, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38811466

RESUMO

Kars Kashar cheese is an artisanal pasta-filata type cheese and geographically marked in Eastern Anatolia of Turkey. The aims of this research were to determine for the first time thermophilic lactic acid bacteria (LAB) of Kars Kashar cheese and characterize the technological properties of obtained isolates. In our research, a number of 15 samples of whey were collected from the different villages in Kars. These samples were incubated at 45 °C and used as the source material for isolating thermophilic LAB. A total of 250 colonies were isolated from thermophilic whey, and 217 of them were determined to be presumptive LAB based on their Gram staining and catalase test. A total of 170 isolates were characterized by their phenotypic properties and identified using the MALDI-TOF mass spectrometry method. Phenotypic identification of isolates displayed that Enterococcus and Lactobacillus were the predominant microbiota. According to MALDI-TOF MS identification, 89 isolates were identified as Enterococcus (52.35%), 57 isolates as Lactobacillus (33.53%), 23 isolates as Streptococcus (13.53%), and one isolate as Lactococcus (0.59%). All thermophilic LAB isolates were successfully identified to the species level and it has been observed that MALDI-TOF MS can be successfully used for the identification of selected LAB. The acidification and proteolytic activities of the isolated thermophilic LAB were examined, and the isolates designated for use as starter cultures were also genotypically defined.


Assuntos
Queijo , Lactobacillales , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Queijo/microbiologia , Lactobacillales/isolamento & purificação , Lactobacillales/classificação , Lactobacillales/genética , Lactobacillales/metabolismo , Soro do Leite/microbiologia , Soro do Leite/química , Microbiologia de Alimentos , Turquia , Lactobacillus/isolamento & purificação , Lactobacillus/genética , Lactobacillus/classificação , Lactobacillus/metabolismo , Enterococcus/isolamento & purificação , Enterococcus/classificação , Enterococcus/genética , Enterococcus/metabolismo
17.
Can J Microbiol ; 70(8): 303-335, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38696839

RESUMO

The rapid increase of antimicrobial-resistant bacteria in humans and livestock is concerning. Antimicrobials are essential for the treatment of disease in modern day medicine, and their misuse in humans and food animals has contributed to an increase in the prevalence of antimicrobial-resistant bacteria. Globally, antimicrobial resistance is recognized as a One Health problem affecting humans, animals, and environment. Enterococcal species are Gram-positive bacteria that are widely distributed in nature. Their occurrence, prevalence, and persistence across the One Health continuum make them an ideal candidate to study antimicrobial resistance from a One Health perspective. The objective of this review was to summarize the role of enterococci as an indicator of antimicrobial resistance across One Health sectors. We also briefly address the prevalence of enterococci in human, animal, and environmental settings. In addition, a 16S RNA gene-based phylogenetic tree was constructed to visualize the evolutionary relationship among enterococcal species and whether they segregate based on host environment. We also review the genomic basis of antimicrobial resistance in enterococcal species across the One Health continuum.


Assuntos
Antibacterianos , Farmacorresistência Bacteriana , Enterococcus , Saúde Única , Filogenia , Enterococcus/efeitos dos fármacos , Enterococcus/genética , Humanos , Antibacterianos/farmacologia , Animais , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , RNA Ribossômico 16S/genética
18.
Curr Microbiol ; 81(8): 225, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38877167

RESUMO

Linezolid resistance in Enterococcus spp. is increasingly considered critically important and a public health threat which mandates the need to understand their genomic contents and dissemination patterns. Here, we used whole-genome sequencing to characterize the resistome, virulome and mobile genetic elements of nine linezolid-resistant (LZDR) enterococci (seven optrA-E. faecalis, one poxtA-E. faecium and one optrA-E. casseliflavus) previously obtained from the nares of healthy dogs, pigs, pig farmers and tracheal samples of nestling storks in Spain. Also, the relatedness of the isolates with publicly available genomes was accessed by core-genome single nucleotide polymorphism (SNP) analysis. The optrA gene of the E. faecalis and E. casseliflavus isolates was located downstream of the fexA gene. The optrA gene in the E. casseliflavus isolate was carried in a plasmid (pURX4962), while those in the seven E. faecalis isolates were chromosomally located. The OptrA proteins were mostly variants of wild type (DP-2: Y176D/T481P; RDK: I104R/Y176D/E256K; DD-3: Y176D/G393D; and EDD: K3E/Y176D/G393D), except two that were wild type (one E. faecalis and one E. casseliflavus). The poxtA gene in the E. faecium isolate was found alone within its contig. The cfrD was upstream of ermB gene in the E. casseliflavus isolate and flanked by ISNCY and IS1216. All the LZDR enterococci carried plasmid rep genes (2-3) containing tetracycline, chloramphenicol and aminoglycoside resistance genes. All isolates except E. casseliflavus carried at least one intact prophage, of which E. faecalis-ST330 (X4957) from a pig carried the highest (n = 5). Tn6260 was associated with lnuG in E. faecalis-ST330 while Tn554 was with fexA in E. feaecalis-ST59 isolates. All except E. casseliflavus (n = 0) carried at least two metal resistance genes (MRGs), of which poxtA-carrying E. faecium-ST1739 isolate contained the most (arsA, copA, fief, ziaA, znuA, zosA, zupT, and zur). SNP-based analyses identified closely related optrA-E. faecalis isolates from a pig and a pig farmer on the same farm (SNP = 4). Moreover, optrA- carrying E. faecalis-ST32, -ST59, and -ST474 isolates from pigs were related to those previously described from humans (sick and healthy) and cattle in Spain, Belgium, and Switzerland (SNP range 43-86). These findings strongly suggest the transmission of LZDR-E. faecalis between a pig and a pig farmer and potential inter-country dissemination. These highlight the need to strengthen molecular surveillance of LZDR enterococci in all ecological niches and body parts to direct appropriate control strategies.


Assuntos
Antibacterianos , Farmacorresistência Bacteriana , Enterococcus , Genoma Bacteriano , Linezolida , Filogenia , Animais , Linezolida/farmacologia , Suínos/microbiologia , Farmacorresistência Bacteriana/genética , Cães , Antibacterianos/farmacologia , Enterococcus/genética , Enterococcus/efeitos dos fármacos , Enterococcus/isolamento & purificação , Enterococcus/classificação , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/transmissão , Infecções por Bactérias Gram-Positivas/veterinária , Humanos , Sequenciamento Completo do Genoma , Espanha , Polimorfismo de Nucleotídeo Único , Testes de Sensibilidade Microbiana , Proteínas de Bactérias/genética , Genômica , Plasmídeos/genética
19.
Lett Appl Microbiol ; 77(7)2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38925640

RESUMO

Faecal contamination of surface waters has the potential to spread not only pathogenic organisms but also antimicrobial resistant organisms. During the bathing season of 2021, weekly water samples, from six selected coastal bathing locations (n = 93) and their freshwater tributaries (n = 93), in Northern Ireland (UK), were examined for concentrations of faecal indicator bacteria Escherichia coli and intestinal enterococci. Microbial source tracking involved detection of genetic markers from the genus Bacteroides using PCR assays for the general AllBac marker, the human HF8 marker and the ruminant BacR marker for the detection of human, and ruminant sources of faecal contamination. The presence of beta-lactamase genes blaOXA-48, blaKPC, and blaNDM-1 was determined using PCR assays for the investigation of antimicrobial resistance genes that are responsible for lack of efficacy in major broad-spectrum antibiotics. The beta-lactamase gene blaOXA-48 was found in freshwater tributary samples at all six locations. blaOXA-48 was detected in 83% of samples that tested positive for the human marker and 69% of samples that tested positive for the ruminant marker over all six locations. This study suggests a risk of human exposure to antimicrobial resistant bacteria where bathing waters receive at least episodically substantial transfers from such tributaries.


Assuntos
Proteínas de Bactérias , Escherichia coli , Fezes , Água Doce , beta-Lactamases , beta-Lactamases/genética , Irlanda do Norte , Água Doce/microbiologia , Proteínas de Bactérias/genética , Escherichia coli/genética , Escherichia coli/isolamento & purificação , Escherichia coli/efeitos dos fármacos , Humanos , Fezes/microbiologia , Microbiologia da Água , Enterococcus/genética , Enterococcus/isolamento & purificação , Enterococcus/enzimologia , Enterococcus/efeitos dos fármacos , Antibacterianos/farmacologia , Animais
20.
Food Microbiol ; 120: 104492, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38431334

RESUMO

Enterococci are widely distributed in dairy sector. They are commensals of the gastrointestinal tract of animals, thus, via fecal contamination, could reach raw milk and dairy products. The aims of this study were: 1) to investigate the enterococcal diversity in cow feces and milk samples and 2) to evaluate the antibiotic resistance (AR) of dairy-related enterococci and their ability to transfer resistance genes. E. faecalis (59.9%), E. faecium (18.6%) and E. lactis (12.4%) were prevalent in milk, while E. faecium (84.2%) and E. hirae (15.0%) were dominant in bovine feces. RAPD-PCR highlighted a high number of Enterococcus biotypes (45 from milk and 37 from feces) and none of the milk strains exhibited genetic profiles similar to those of feces biotypes. A high percentage of enterococci isolated from milk (71%) were identified as multidrug resistant and resistance against streptomycin and tetracycline were widespread among milk strains while enterococci from feces were commonly resistant to linezolid and quinupristin/dalfopristin. Only E. faecalis strains were able to transfer horizontally the tetM gene to Lb. delbrueckii subsp. lactis. Our results indicated that Enterococcus biotypes from milk and bovine feces belong to different community and the ability of these microorganisms to transfer AR genes is strain-dependent.


Assuntos
Enterococcus faecium , Enterococcus , Feminino , Bovinos , Animais , Enterococcus/genética , Leite , Técnica de Amplificação ao Acaso de DNA Polimórfico , Testes de Sensibilidade Microbiana , Antibacterianos/farmacologia , Resistência Microbiana a Medicamentos , Fezes , Biodiversidade , Farmacorresistência Bacteriana/genética , Enterococcus faecalis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA