Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.493
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 185(7): 1157-1171.e22, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35259335

RESUMEN

Enterococci are a part of human microbiota and a leading cause of multidrug resistant infections. Here, we identify a family of Enterococcus pore-forming toxins (Epxs) in E. faecalis, E. faecium, and E. hirae strains isolated across the globe. Structural studies reveal that Epxs form a branch of ß-barrel pore-forming toxins with a ß-barrel protrusion (designated the top domain) sitting atop the cap domain. Through a genome-wide CRISPR-Cas9 screen, we identify human leukocyte antigen class I (HLA-I) complex as a receptor for two members (Epx2 and Epx3), which preferentially recognize human HLA-I and homologous MHC-I of equine, bovine, and porcine, but not murine, origin. Interferon exposure, which stimulates MHC-I expression, sensitizes human cells and intestinal organoids to Epx2 and Epx3 toxicity. Co-culture with Epx2-harboring E. faecium damages human peripheral blood mononuclear cells and intestinal organoids, and this toxicity is neutralized by an Epx2 antibody, demonstrating the toxin-mediated virulence of Epx-carrying Enterococcus.


Asunto(s)
Toxinas Bacterianas/metabolismo , Enterococcus , Leucocitos Mononucleares , Factores de Virulencia/metabolismo , Animales , Bovinos , Enterococcus/metabolismo , Enterococcus/patogenicidad , Caballos , Ratones , Pruebas de Sensibilidad Microbiana , Porcinos
2.
Cell ; 169(5): 849-861.e13, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28502769

RESUMEN

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.


Asunto(s)
Evolución Biológica , Enterococcus/genética , Animales , Enfermedades Transmisibles Emergentes/microbiología , Infección Hospitalaria/microbiología , Farmacorresistencia Bacteriana , Enterococcus/clasificación , Enterococcus/citología , Enterococcus/efectos de los fármacos , Especiación Genética , Interacciones Huésped-Patógeno , Larva/microbiología , Mariposas Nocturnas/crecimiento & desarrollo , Mariposas Nocturnas/microbiología , Filogenia , ARN Ribosómico 16S/genética
3.
Nature ; 626(7998): 419-426, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38052229

RESUMEN

Determining the structure and phenotypic context of molecules detected in untargeted metabolomics experiments remains challenging. Here we present reverse metabolomics as a discovery strategy, whereby tandem mass spectrometry spectra acquired from newly synthesized compounds are searched for in public metabolomics datasets to uncover phenotypic associations. To demonstrate the concept, we broadly synthesized and explored multiple classes of metabolites in humans, including N-acyl amides, fatty acid esters of hydroxy fatty acids, bile acid esters and conjugated bile acids. Using repository-scale analysis1,2, we discovered that some conjugated bile acids are associated with inflammatory bowel disease (IBD). Validation using four distinct human IBD cohorts showed that cholic acids conjugated to Glu, Ile/Leu, Phe, Thr, Trp or Tyr are increased in Crohn's disease. Several of these compounds and related structures affected pathways associated with IBD, such as interferon-γ production in CD4+ T cells3 and agonism of the pregnane X receptor4. Culture of bacteria belonging to the Bifidobacterium, Clostridium and Enterococcus genera produced these bile amidates. Because searching repositories with tandem mass spectrometry spectra has only recently become possible, this reverse metabolomics approach can now be used as a general strategy to discover other molecules from human and animal ecosystems.


Asunto(s)
Amidas , Ácidos y Sales Biliares , Ésteres , Ácidos Grasos , Metabolómica , Animales , Humanos , Bifidobacterium/metabolismo , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Clostridium/metabolismo , Estudios de Cohortes , Enfermedad de Crohn/metabolismo , Enterococcus/metabolismo , Ésteres/química , Ésteres/metabolismo , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Metabolómica/métodos , Fenotipo , Receptor X de Pregnano/metabolismo , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem , Amidas/química , Amidas/metabolismo
4.
Nature ; 611(7937): 780-786, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36385534

RESUMEN

Enteric pathogens are exposed to a dynamic polymicrobial environment in the gastrointestinal tract1. This microbial community has been shown to be important during infection, but there are few examples illustrating how microbial interactions can influence the virulence of invading pathogens2. Here we show that expansion of a group of antibiotic-resistant, opportunistic pathogens in the gut-the enterococci-enhances the fitness and pathogenesis of Clostridioides difficile. Through a parallel process of nutrient restriction and cross-feeding, enterococci shape the metabolic environment in the gut and reprogramme C. difficile metabolism. Enterococci provide fermentable amino acids, including leucine and ornithine, which increase C. difficile fitness in the antibiotic-perturbed gut. Parallel depletion of arginine by enterococci through arginine catabolism provides a metabolic cue for C. difficile that facilitates increased virulence. We find evidence of microbial interaction between these two pathogenic organisms in multiple mouse models of infection and patients infected with C. difficile. These findings provide mechanistic insights into the role of pathogenic microbiota in the susceptibility to and the severity of C. difficile infection.


Asunto(s)
Clostridioides difficile , Enterococcus , Interacciones Microbianas , Animales , Humanos , Ratones , Antibacterianos/farmacología , Arginina/deficiencia , Arginina/metabolismo , Clostridioides difficile/metabolismo , Clostridioides difficile/patogenicidad , Clostridioides difficile/fisiología , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Enterococcus/efectos de los fármacos , Enterococcus/metabolismo , Enterococcus/patogenicidad , Enterococcus/fisiología , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/metabolismo , Intestinos/microbiología , Leucina/metabolismo , Ornitina/metabolismo , Virulencia , Susceptibilidad a Enfermedades
5.
Nature ; 607(7919): 563-570, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35831502

RESUMEN

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.


Asunto(s)
Bacterias , Traslocación Bacteriana , Evolución Biológica , Microbioma Gastrointestinal , Hígado , Bacterias/genética , Bacterias/inmunología , Bacterias/patogenicidad , Traslocación Bacteriana/genética , Pared Celular/genética , Enterococcus/genética , Enterococcus/inmunología , Microbioma Gastrointestinal/genética , Genómica , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamación/microbiología , Inflamación/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Limosilactobacillus reuteri/genética , Limosilactobacillus reuteri/inmunología , Hígado/microbiología , Hígado/patología , Ganglios Linfáticos/microbiología , Mutación , Procesos Estocásticos , Simbiosis/genética , Simbiosis/inmunología
6.
Proc Natl Acad Sci U S A ; 121(10): e2310852121, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38416678

RESUMEN

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.


Asunto(s)
Enterococcus faecium , Infecciones por Bacterias Grampositivas , Animales , Humanos , Enterococcus/genética , Antibacterianos/farmacología , Enterococcus faecium/genética , Enterococcus faecalis/genética , Filogenia , Pruebas de Sensibilidad Microbiana , Farmacorresistencia Bacteriana
7.
Clin Microbiol Rev ; 37(2): e0012123, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38466110

RESUMEN

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.


Asunto(s)
Infecciones por Bacterias Grampositivas , Factores de Virulencia , Humanos , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Factores de Virulencia/genética , Animales , Farmacorresistencia Bacteriana , Antibacterianos/uso terapéutico , Antibacterianos/farmacología , Enterococcus/patogenicidad , Enterococcus/efectos de los fármacos , Enterococcus/genética , Virulencia
8.
PLoS Pathog ; 19(6): e1011424, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37267422

RESUMEN

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.


Asunto(s)
Enterococcus faecium , Enterococcus , Humanos , Enterococcus/genética , Elementos Transponibles de ADN/genética , Antibacterianos/farmacología , Farmacorresistencia Microbiana , Enterococcus faecalis/genética
9.
BMC Genomics ; 25(1): 28, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172677

RESUMEN

BACKGROUND: Enterococcus faecium and E. lactis are phylogenetically closely related lactic acid bacteria that are ubiquitous in nature and are known to be beneficial or pathogenic. Despite their considerable industrial and clinical importance, comprehensive studies on their evolutionary relationships and genomic, metabolic, and pathogenic traits are still lacking. Therefore, we conducted comparative pangenome analyses using all available dereplicated genomes of these species. RESULTS: E. faecium was divided into two subclades: subclade I, comprising strains derived from humans, animals, and food, and the more recent phylogenetic subclade II, consisting exclusively of human-derived strains. In contrast, E. lactis strains, isolated from diverse sources including foods, humans, animals, and the environment, did not display distinct clustering based on their isolation sources. Despite having similar metabolic features, noticeable genomic differences were observed between E. faecium subclades I and II, as well as E. lactis. Notably, E. faecium subclade II strains exhibited significantly larger genome sizes and higher gene counts compared to both E. faecium subclade I and E. lactis strains. Furthermore, they carried a higher abundance of antibiotic resistance, virulence, bacteriocin, and mobile element genes. Phylogenetic analysis of antibiotic resistance and virulence genes suggests that E. faecium subclade II strains likely acquired these genes through horizontal gene transfer, facilitating their effective adaptation in response to antibiotic use in humans. CONCLUSIONS: Our study offers valuable insights into the adaptive evolution of E. faecium strains, enabling their survival as pathogens in the human environment through horizontal gene acquisitions.


Asunto(s)
Enterococcus faecium , Animales , Humanos , Filogenia , Enterococcus , Genómica , Antibacterianos
10.
BMC Genomics ; 25(1): 718, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39054474

RESUMEN

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.


Asunto(s)
Bifidobacterium , Enterococcus , Microbioma Gastrointestinal , Humanos , Zimbabwe , Lactante , Microbioma Gastrointestinal/genética , Enterococcus/genética , Enterococcus/efectos de los fármacos , Enterococcus/aislamiento & purificación , Bifidobacterium/genética , Bifidobacterium/aislamiento & purificación , Bifidobacterium/efectos de los fármacos , Genómica , Genoma Bacteriano , Heces/microbiología , Secuenciación Completa del Genoma , Estudios de Cohortes , Filogenia
11.
Mol Microbiol ; 120(6): 805-810, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38012814

RESUMEN

Regulation of the first committed step of peptidoglycan precursor synthesis by MurA-enzyme homologs has recently taken center stage in many different bacteria. In different low-GC Gram-positive bacteria, regulation of this step has been shown to be regulated by phosphorylation of homologs of the IreB/ReoM regulatory protein by PASTA-domain Ser/Thr-protein kinases. In this issue, Mascari, Little, and Kristich determine this regulatory pathway and its links to resistance to cephalosporin ß-lactam antibiotics in the major human pathogen, Enterococcus faecalis (Efa). Unbiased genetic selections identified MurAA (MurA-family homolog) as the downstream target of IreB regulation in the absence of the IreK Ser/Thr-protein kinase. Physiological and biochemical approaches, including determination of MICs to ceftriaxone, Western blotting of MurAA cellular amounts, isotope incorporation into peptidoglycan sacculi, and thermal-shift binding assays of purified proteins, demonstrated that unphosphorylated IreB, together with proteins MurAB (MurZ-family homolog), and ReoY(Efa) negatively regulate MurAA stability and cellular amount by the ClpCP protease. Importantly, this paper supports the idea that ceftriaxone stimulates phosphorylation of IreB, which leads to increased cellular MurAA amount and precursor pathway flux required for E. faecalis cephalosporin resistance. Overall, findings in this paper significantly contribute to understanding variations of this central regulatory pathway in other low-GC Gram-positive bacteria.


Asunto(s)
Ceftriaxona , Enterococcus , Humanos , Fosforilación , Enterococcus/metabolismo , Peptidoglicano/metabolismo , Enterococcus faecalis/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
12.
Chembiochem ; 25(12): e202400212, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38648232

RESUMEN

The ß-hemolytic factor streptolysin S (SLS) is an important linear azol(in)e-containing peptide (LAP) that contributes significantly to the virulence of Streptococcus pyogenes. Despite its discovery 85 years ago, SLS has evaded structural characterizing owing to its notoriously problematic physicochemical properties. Here, we report the discovery and characterization of a structurally analogous hemolytic peptide from Enterococcus caccae, termed enterolysin S (ELS). Through heterologous expression, site-directed mutagenesis, chemoselective modification, and high-resolution mass spectrometry, we found that ELS contains an intriguing contiguous octathiazole moiety. The discovery of ELS expands our knowledge of hemolytic LAPs by adding a new member to this virulence-promoting family of modified peptides.


Asunto(s)
Enterococcus , Enterococcus/metabolismo , Tiazoles/química , Tiazoles/metabolismo , Péptidos/química , Péptidos/metabolismo , Hemólisis/efectos de los fármacos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Secuencia de Aminoácidos
13.
J Antimicrob Chemother ; 79(5): 997-1005, 2024 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-38501366

RESUMEN

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.


Asunto(s)
Reacción en Cadena en Tiempo Real de la Polimerasa , Sensibilidad y Especificidad , Resistencia a la Vancomicina , Enterococos Resistentes a la Vancomicina , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Resistencia a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/aislamiento & purificación , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Enterococcus/genética , Enterococcus/efectos de los fármacos , Enterococcus/aislamiento & purificación , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/diagnóstico , Proteínas Bacterianas/genética , Factores de Tiempo , Genes Bacterianos/genética
14.
J Antimicrob Chemother ; 79(4): 846-850, 2024 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-38366373

RESUMEN

OBJECTIVES: To investigate the global distribution of an optrA-harbouring linezolid-resistant Enterococcus faecalis ST476 clonal lineage. METHODS: Comprehensive searches of the NCBI database were performed to identify published peer-reviewed articles and genomes of E. faecalis ST476. Each genome was analysed for resistome, virulome, OptrA variant and optrA genetic contexts. A phylogenetic comparison of ST476 genomes with publicly available genomes of other STs was also performed. RESULTS: Sixty-six E. faecalis ST476 isolates from 15 countries (China, Japan, South Korea, Austria, Denmark, Spain, Czech Republic, Colombia, Tunisia, Italy, Malaysia, Belgium, Germany, United Arab Emirates and Switzerland) mainly of human and animal origin were identified. Thirty available ST476 genomes compared with genomes of 591 STs indicated a progressive radiation of E. faecalis STs starting from ST21. The closest ancestral node for ST476 was ST1238. Thirty E. faecalis ST476 genomes exhibited 3-916 SNP differences. Several antimicrobial resistance and virulence genes were conserved among the ST476 genomes. The optrA genetic context exhibited a high degree of or complete identity to the chromosomal transposon Tn6674. Only three isolates displayed an optrA-carrying plasmid with complete or partial Tn6674. The WT OptrA protein was most widespread in the ST476 lineage. CONCLUSIONS: Linezolid-resistant optrA-carrying E. faecalis of the clonal lineage ST476 is globally distributed in human, animal and environmental settings. The presence of such an emerging clone can be of great concern for public health. Thus, a One Health approach is needed to counteract the spread and the evolution of this enterococcal clonal lineage.


Asunto(s)
Enterococcus faecium , Infecciones por Bacterias Grampositivas , Animales , Humanos , Linezolid/farmacología , Antibacterianos/farmacología , Enterococcus faecalis , Filogenia , Farmacorresistencia Bacteriana/genética , Enterococcus , Infecciones por Bacterias Grampositivas/epidemiología , Infecciones por Bacterias Grampositivas/veterinaria , Enterococcus faecium/genética , Pruebas de Sensibilidad Microbiana
15.
PLoS Pathog ; 18(9): e1010829, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36103556

RESUMEN

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.


Asunto(s)
Infecciones por Bacterias Grampositivas , Proteínas de la Membrana , Proteínas Bacterianas/metabolismo , Biopelículas , Enterococcus/genética , Enterococcus/metabolismo , Enterococcus faecalis , Humanos , Proteínas de la Membrana/metabolismo , Péptido Hidrolasas/metabolismo
16.
BMC Microbiol ; 24(1): 98, 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38528458

RESUMEN

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.


Asunto(s)
Cardiopatías Congénitas , Insuficiencia Cardíaca , Lactante , Humanos , Interleucina-10 , Factor de Necrosis Tumoral alfa , Interleucina-6 , Interleucina-4 , Insuficiencia Cardíaca/complicaciones , Cardiopatías Congénitas/complicaciones , Enterococcus/genética , Inflamación
17.
BMC Microbiol ; 24(1): 110, 2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38570789

RESUMEN

BACKGROUND: All gastrointestinal pathogens, including Enterococcus faecalis and Enterococcus faecium, undergo adaptation processes during colonization and infection. In this study, we investigated by data-independent acquisition mass spectrometry (DIA-MS) two crucial adaptations of these two Enterococcus species at the proteome level. Firstly, we examined the adjustments to cope with bile acid concentrations at 0.05% that the pathogens encounter during a potential gallbladder infection. Therefore, we chose the primary bile acids cholic acid (CA) and chenodeoxycholic acid (CDCA) as well as the secondary bile acid deoxycholic acid (DCA), as these are the most prominent bile acids. Secondly, we investigated the adaptations from an aerobic to a microaerophilic environment, as encountered after oral-fecal infection, in the absence and presence of deoxycholic acid (DCA). RESULTS: Our findings showed similarities, but also species-specific variations in the response to the different bile acids. Both Enterococcus species showed an IC50 in the range of 0.01- 0.023% for DCA and CDCA in growth experiments and both species were resistant towards 0.05% CA. DCA and CDCA had a strong effect on down-expression of proteins involved in translation, transcription and replication in E. faecalis (424 down-expressed proteins with DCA, 376 down-expressed proteins with CDCA) and in E. faecium (362 down-expressed proteins with DCA, 391 down-expressed proteins with CDCA). Proteins commonly significantly altered in their expression in all bile acid treated samples were identified for both species and represent a "general bile acid response". Among these, various subunits of a V-type ATPase, different ABC-transporters, multi-drug transporters and proteins related to cell wall biogenesis were up-expressed in both species and thus seem to play an essential role in bile acid resistance. Most of the differentially expressed proteins were also identified when E. faecalis was incubated with low levels of DCA at microaerophilic conditions instead of aerobic conditions, indicating that adaptations to bile acids and to a microaerophilic atmosphere can occur simultaneously. CONCLUSIONS: Overall, these findings provide a detailed insight into the proteomic stress response of two Enterococcus species and help to understand the resistance potential and the stress-coping mechanisms of these important gastrointestinal bacteria.


Asunto(s)
Ácidos y Sales Biliares , Enterococcus faecium , Ácidos y Sales Biliares/farmacología , Enterococcus faecalis/genética , Enterococcus faecalis/metabolismo , Enterococcus faecium/genética , Enterococcus faecium/metabolismo , Ácido Desoxicólico/farmacología , Proteómica , Ácido Cólico , Ácido Quenodesoxicólico/metabolismo , Enterococcus
18.
FASEB J ; 37(10): e23186, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37665578

RESUMEN

Enterococcus faecalis (E. faecalis) is one of the major pathogenic bacteria responsible for surgical site infections. Biofilm infections are major hospital-acquired infections. Previous studies suggested that ions could regulate biofilm formation in microbes. Volatile anesthetics, frequently administered in surgical setting, target ion channels. Here, we investigated the role of ion channels/transporters and volatile anesthetics in the biofilm formation by E. faecalis MMH594 strain and its ion transporter mutants. We found that a chloride transporter mutant significantly reduced biofilm formation compared to the parental strain. Downregulation of teichoic acid biosynthesis in the chloride transporter mutant impaired biofilm matrix formation and cellular adhesion, leading to mitigated biofilm formation. Among anesthetics, isoflurane exposure enhanced biofilm formation in vitro and in vivo. The upregulation of de novo purine biosynthesis pathway by isoflurane exposure potentially enhanced biofilm formation, an essential process for DNA, RNA, and ATP synthesis. We also demonstrated that isoflurane exposure to E. faecalis increased cyclic-di-AMP and extracellular DNA production, consistent with the increased purine biosynthesis. We further showed that isoflurane enhanced the enzymatic activity of phosphoribosyl pyrophosphate synthetase (PRPP-S). With the hypothesis that isoflurane directly bound to PRPP-S, we predicted isoflurane binding site on it using rigid docking. Our study provides a better understanding of the underlying mechanisms of E. faecalis biofilm formation and highlights the potential impact of an ion transporter and volatile anesthetic on this process. These findings may lead to the development of novel strategies for preventing E. faecalis biofilm formation and improving patient outcomes in clinical settings.


Asunto(s)
Anestésicos , Infecciones Bacterianas , Isoflurano , Humanos , Isoflurano/farmacología , Cloruros , Biopelículas , Proteínas de Transporte de Membrana , AMP Cíclico , Enterococcus
19.
Arch Microbiol ; 206(2): 84, 2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38296886

RESUMEN

Enterococcus mundtii AM_AQ_BC8 isolated from biofouled filtration membrane was characterised as a potential probiotic bacterium showing strong L-lactic acid-producing capability. Experimental studies revealed that E. mundtii AM_AQ_BC8 possess antibiofilm and antimicrobial ability too, as tested against strong biofilm-forming bacteria like Pseudomonas spp. The present study has evaluated the genetic potential of E. mundtii AM_AQ_BC8 through genome sequencing. Whole genome analysis revealed the presence of key genes like ldh_1 and ldh_2 responsible for lactic acid production along with genes encoding probiotic features such as acid and bile salt resistance (dnaK, dnaJ, argS), fatty acid synthesis (fabD, fabE) and lactose utilisation (lacG, lacD). The phylogenomic analysis based on OrthoANI (99.85%) and dDDH (96.8%) values revealed that the strain AM_AQ_BC8 shared the highest homology with E. mundtii. The genome sequence of strain AM_AQ_BC8 has been deposited to NCBI and released with GenBank accession no. SAMN32531201. The study primarily demonstrated the probiotic potential of E. mundtii AM_AQ_BC8 isolate, for L-lactate synthesis in high concentration (8.98 g/L/day), which also showed anti-biofilm and antimicrobial activities.


Asunto(s)
Antiinfecciosos , Enterococcus , Probióticos , Genómica , Biopelículas
20.
Arch Microbiol ; 206(10): 397, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39249569

RESUMEN

Amongst all Enterococcus spp., E. faecalis and E. faecium are most known notorious pathogen and their biofilm formation has been associated with endocarditis, oral, urinary tract, and wound infections. Biofilm formation involves a pattern of initial adhesion, microcolony formation, and mature biofilms. The initial adhesion and microcolony formation involve numerous surface adhesins e.g. pili Ebp and polysaccharide Epa. The mature biofilms are maintained by eDNA, It's worth noting that phage-mediated dispersal plays a prominent role. Further, the involvement of peptide pheromones in regulating biofilm maintenance sets it apart from other pathogens and facilitating the horizontal transfer of resistance genes. The role of fsr based regulation by regulating gelE expression is also discussed. Thus, we provide a concise overview of the significant determinants at each stage of Enterococcus spp. biofilm formation. These elements could serve as promising targets for antibiofilm strategies.


Asunto(s)
Biopelículas , Enterococcus , Infecciones por Bacterias Grampositivas , Enterococcus/genética , Enterococcus/metabolismo , Regulación Bacteriana de la Expresión Génica , Infecciones por Bacterias Grampositivas/epidemiología , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/fisiopatología , Adhesión Bacteriana/genética , Adhesinas Bacterianas/genética , Adhesinas Bacterianas/metabolismo , Polisacáridos Bacterianos/metabolismo , Transferencia de Gen Horizontal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA