Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38559250

ABSTRACT

Quorum sensing (QS) is a cell-cell signaling system that enables bacteria to coordinate population density-dependent changes in behavior. This chemical communication pathway is mediated by diffusible N-acyl L-homoserine lactone signals and cytoplasmic signal-responsive LuxR-type receptors in Gram-negative bacteria. As many common pathogenic bacteria use QS to regulate virulence, there is significant interest in disrupting QS as a potential therapeutic strategy. Prior studies have implicated the natural products salicylic acid, cinnamaldehyde and other related benzaldehyde derivatives as inhibitors of QS in the opportunistic pathogen Pseudomonas aeruginosa, yet we lack an understanding of the mechanisms by which these compounds function. Herein, we evaluate the activity of a set of benzaldehyde derivatives using heterologous reporters of the P. aeruginosa LasR and RhlR QS signal receptors. We find that most tested benzaldehyde derivatives can antagonize LasR or RhlR reporter activation at micromolar concentrations, although certain molecules also caused mild growth defects and nonspecific reporter antagonism. Notably, several compounds showed promising RhlR or LasR specific inhibitory activities over a range of concentrations below that causing toxicity. Ortho-Vanillin, a previously untested compound, was the most promising within this set. Competition experiments against the native ligands for LasR and RhlR revealed that ortho-vanillin can interact competitively with RhlR but not with LasR. Overall, these studies expand our understanding of benzaldehyde activities in the LasR and RhlR receptors and reveal potentially promising effects of ortho-vanillin as a small molecule QS modulator against RhlR.

2.
J Bacteriol ; 203(21): e0037121, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34424034

ABSTRACT

The ability of Enterococcus faecalis to use a variety of carbon sources enables colonization at various anatomic sites within a mammalian host. N-Acetylglucosamine (GlcNAc) is one of the most abundant natural sugars and provides bacteria with a source of carbon and nitrogen when metabolized. N-Acetylglucosamine is also a component of bacterial peptidoglycan, further highlighting the significance of N-acetylglucosamine utilization. In this study, we show that CcpA-regulated enzymes are required for growth on the poly-ß1,4-linked GlcNAc substrate, chitopentaose (ß1,4-linked GlcNAc5). We also show that EF0114 (EndoE) is required for growth on chitobiose (ß1,4-linked GlcNAc2) and that the GH20 domain of EndoE is required for the conversion of GlcNAc2 to N-acetylglucosamine. GlcNAc is transported into the cell via two separate phosphotransferase system (PTS) complexes, either the PTS IICBA encoded by ef1516 (nagE) or the Mpt glucose/mannose permease complex (MptBACD). The Mpt PTS is also the primary glucosamine transporter. In order for N-acetylglucosamine to be utilized as a carbon source, phosphorylated N-acetylglucosamine (GlcNAc-6-P) must be deacetylated, and here, we show that this activity is mediated by EF1317 (an N-acetylglucosamine-6-phosphate deacetylase; NagA homolog), as a deletion of ef1317 is unable to grow on GlcNAc as the carbon source. Deamination of glucosamine to fructose-6-phosphate is required for entry into glycolysis, and we show that growth on glucosamine is dependent on EF0466 (a glucosamine-6-phosphate deaminase; NagB homolog). Collectively, our data highlight the chitinolytic machinery required for breaking down exogenous chitinous substrates, as well as the uptake and cytosolic enzymes needed for metabolizing N-acetylglucosamine. IMPORTANCE Enterococcus faecalis causes life-threatening health care-associated infections in part due to its intrinsic and acquired antibiotic resistance, its ability to form biofilms, and its nutrient versatility. Alternative nutrient acquisition systems are key factors that contribute to enterococcal colonization at biologically unique host anatomic sites. Although E. faecalis can metabolize an array of carbon sources, little is known of how this bacterium acquires these secondary nutrient sources in mammalian hosts. Our research identifies the glycosidase machinery required for degrading exogenous chitinous substrates into N-acetylglucosamine monomers for transport and metabolism of one of the most abundant naturally occurring sugars, N-acetylglucosamine. Disrupting the function of this N-acetylglucosamine acquisition pathway may lead to new treatments against multidrug-resistant enterococcal infections.


Subject(s)
Acetylglucosamine/analogs & derivatives , Acetylglucosamine/metabolism , Enterococcus faecalis/metabolism , Gene Expression Regulation, Bacterial/physiology , Glucosamine/metabolism , Glycoside Hydrolases/metabolism , Aldose-Ketose Isomerases/genetics , Aldose-Ketose Isomerases/metabolism , Amidohydrolases/metabolism , Bacterial Proteins , Biological Transport , Enterococcus faecalis/drug effects , Gene Deletion , Gene Expression Regulation, Bacterial/genetics , Gene Expression Regulation, Enzymologic/genetics , Glycoside Hydrolases/genetics
3.
Infect Immun ; 89(11): e0034321, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34424752

ABSTRACT

The ability of Enterococcus faecalis to colonize host anatomical sites is dependent on its adaptive response to host conditions. Three glycosyl hydrolase gene clusters, each belonging to glycosyl hydrolase family 18 (GH18) (ef0114, ef0361, and ef2863), in E. faecalis were previously found to be upregulated under glucose-limiting conditions. The GH18 catalytic domain is present in proteins that are classified as either chitinases or ß-1,4 endo-ß-N-acetylglucosaminidases (ENGases) based on their ß-1,4 endo-N-acetyl-ß-d-glucosaminidase activity, and ENGase activity is commonly associated with cleaving N-linked glycoprotein, an abundant glycan structure on host epithelial surfaces. Here, we show that all three hydrolases are negatively regulated by the transcriptional regulator carbon catabolite protein A (CcpA). Additionally, we demonstrate that a constitutively active CcpA variant represses the expression of CcpA-regulated genes irrespective of glucose availability. Previous studies showed that the GH18 catalytic domains of EndoE (EF0114) and EfEndo18A (EF2863) were capable of deglycosylating RNase B, a model high-mannose-type glycoprotein. However, it remained uncertain which glycosidase is primarily responsible for the deglycosylation of high-mannose-type glycoproteins. In this study, we show by mutation analysis as well as a dose-dependent analysis of recombinant protein expression that EfEndo18A is primarily responsible for deglycosylating high-mannose glycoproteins and that the glycans removed by EfEndo18A support growth under nutrient-limiting conditions in vitro. In contrast, IgG is representative of a complex-type glycoprotein, and we demonstrate that the GH18 domain of EndoE is primarily responsible for the removal of this glycan decoration. Finally, our data highlight the combined contribution of glycosidases to the virulence of E. faecalis in vivo.


Subject(s)
Enterococcus faecalis/metabolism , Glycoside Hydrolases/physiology , Bacterial Proteins/physiology , Biofilms , Catalytic Domain , Enterococcus faecalis/genetics , Enterococcus faecalis/pathogenicity , Gene Expression Regulation, Bacterial , Mannose/metabolism , Nutrients/metabolism , Polysaccharides/metabolism , Ribonucleases/metabolism , Transcription Initiation Site
4.
J Bacteriol ; 203(20): e0025821, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34370556

ABSTRACT

Enterococcus faecalis is a gut commensal but transitions to a pathogenic state as a consequence of intestinal dysbiosis and/or the presence of indwelling medical devices, causing a wide range of infections. One of the unique features of E. faecalis is its ability to display high level resistance to lysozyme, an important host defense of the innate immune response. Lysozyme resistance in E. faecalis is known to be mediated by the extracytoplasmic function (ECF) sigma factor SigV. PgdA and RsiV expression is directly regulated by SigV, but pgdA and rsiV mutants display nominal changes in lysozyme resistance, suggesting that additional gene products in the SigV regulon contribute to lysozyme resistance. Using transcriptome sequencing (RNA-seq) analysis, we compared the transcriptional profile of the parental strain to that of an isogenic sigV mutant and show that apart from sigV, only rsiV and pgdA expression was induced upon lysozyme exposure. The combined deletion mutant of both rsiV and pgdA rendered E. faecalis sensitive to lysozyme at a level comparable to that of the sigV mutant, highlighting the limited SigV regulon. Several additional genes were also induced upon lysozyme exposure, but in a SigV-independent fashion. Overexpression of pgdA from a SigV-independent promoter restored lysozyme resistance in a sigV deletion mutant and also induced cell chaining. Overexpression of rsiV from a SigV-independent promoter only partially restored lysozyme resistance in a sigV mutant. Overall, we provide evidence for a simple adaptation to lysozyme stress, in which SigV controls the expression of rsiV and pgdA, and that both gene products contribute to lysozyme resistance. IMPORTANCE Enterococcus faecalis causes health care-associated infections and displays resistance to a variety of antibiotics and molecules of the innate immune system. SigV has been shown to play an important role in enterococcal lysozyme resistance. Even though several proteins have been implicated in enterococcal lysozyme resistance, a complete SigV-dependent regulon has not been functionally characterized as being responsible for the dramatic increase in lysozyme susceptibility displayed by a sigV mutant. Using RNA-seq, we have identified the SigV regulon to be comprised of two gene loci, sigV-rsiV and pgdA. Deletion of both rsiV and pgdA renders E. faecalis susceptible to lysozyme on par with a sigV mutant. We also demonstrate that overproduction of rsiV and pgdA contributes to lysozyme resistance in susceptible strains.


Subject(s)
Enterococcus faecalis/metabolism , Gene Expression Regulation, Bacterial/physiology , Muramidase , Sigma Factor/metabolism , Cell Wall/metabolism , Enterococcus faecalis/genetics , Regulon/genetics , Regulon/physiology , Sigma Factor/genetics
5.
mBio ; 12(3)2021 05 18.
Article in English | MEDLINE | ID: mdl-34006651

ABSTRACT

The alternative sigma factor σ54 has been shown to regulate the expression of a wide array of virulence-associated genes, as well as central metabolism, in bacterial pathogens. In Gram-positive organisms, the σ54 is commonly associated with carbon metabolism. In this study, we show that the Enterococcus faecalis alternative sigma factor σ54 (RpoN) and its cognate enhancer binding protein MptR are essential for mannose utilization and are primary contributors to glucose uptake through the Mpt phosphotransferase system. To gain further insight into how RpoN contributes to global transcriptional changes, we performed microarray transcriptional analysis of strain V583 and an isogenic rpoN mutant grown in a chemically defined medium with glucose as the sole carbon source. Transcripts of 340 genes were differentially affected in the rpoN mutant; the predicted functions of these genes mainly related to nutrient acquisition. These differentially expressed genes included those with predicted catabolite-responsive element (cre) sites, consistent with loss of repression by the major carbon catabolite repressor CcpA. To determine if the inability to efficiently metabolize glucose/mannose affected infection outcome, we utilized two distinct infection models. We found that the rpoN mutant is significantly attenuated in both rabbit endocarditis and murine catheter-associated urinary tract infection (CAUTI). Here, we examined a ccpA mutant in the CAUTI model and showed that the absence of carbon catabolite control also significantly attenuates bacterial tissue burden in this model. Our data highlight the contribution of central carbon metabolism to growth of E. faecalis at various sites of infection.IMPORTANCE Hospital-acquired infections account for 2 billion dollars annually in increased health care expenses and cause more than 100,000 deaths in the United States alone. Enterococci are the second leading cause of hospital-acquired infections. They form biofilms at surgical sites and are often associated with infections of the urinary tract following catheterization. Nutrient uptake and growth are key factors that influence their ability to cause disease. Our research identified a large set of genes that illuminate nutrient uptake pathways in enterococci. Perturbation of the metabolic circuit reduces virulence in a rabbit endocarditis model, as well as in catheter-associated urinary tract infection in mice. Targeting metabolic pathways that are important in infection may lead to new treatments against multidrug-resistant enterococcal infections.


Subject(s)
Bacterial Proteins/genetics , Carbon/metabolism , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Gene Expression , Sigma Factor/genetics , Animals , Bacterial Proteins/metabolism , Biofilms , Gene Expression Regulation, Bacterial , Rabbits , Sigma Factor/classification , Virulence
6.
J Bacteriol ; 202(17)2020 08 10.
Article in English | MEDLINE | ID: mdl-32540933

ABSTRACT

Enterococcus faecalis is an opportunistic pathogen capable of causing infections, including endocarditis and urinary tract infections (UTI). One of the well-characterized quorum-sensing pathways in E. faecalis involves coordination of the conjugal transfer of pheromone-responsive plasmids by PrgX, a member of the RRNPP protein family. Members of this protein family in various Firmicutes have also been shown to contribute to numerous cellular processes, including sporulation, competence, conjugation, nutrient sensing, biofilm formation, and virulence. As PrgX is a plasmid-encoded RRNPP family member, we surveyed the genome of the multidrug-resistant strain V583 for additional RRNPP homologs using computational searches and refined those identified hits for predicted structural similarities to known RRNPP family members. This led us to investigate the contribution of the chromosomally encoded RRNPP homologs to biofilm processes and pathogenesis in a catheter-associated urinary tract infection (CAUTI) model. In this study, we identified five such homologs and report that 3 of the 5 homologs, EF0073, EF1599, and EF1316, affect biofilm formation as well as outcomes in the CAUTI model.IMPORTANCEEnterococcus faecalis causes health care-associated infections and displays resistance to a variety of broad-spectrum antibiotics by acquisition of resistance traits as well as the ability to form biofilms. Even though a growing number of factors related to biofilm formation have been identified, mechanisms that contribute to biofilm formation are still largely unknown. Members of the RRNPP protein family regulate a diverse set of biological reactions in low-G+C Gram-positive bacteria (Firmicutes). Here, we identify three predicted structural homologs of the RRNPP family, EF0073, EF1599, and EF1316, which affect biofilm formation and CAUTI pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Enterococcus faecalis/metabolism , Urinary Tract Infections/microbiology , Bacterial Proteins/genetics , Drug Resistance, Multiple, Bacterial , Enterococcus faecalis/drug effects , Enterococcus faecalis/genetics , Gene Expression Regulation, Bacterial/physiology , Humans
7.
mSphere ; 1(3)2016.
Article in English | MEDLINE | ID: mdl-27306929

ABSTRACT

Enterococcus faecalis is an opportunistic pathogen and is one of the leading causes of nosocomial infections. E. faecalis harbors a number of antibiotic resistance genes, and most of these are present on mobile genetic elements (MGEs) that can be disseminated within the species, as well as to other members of the human microflora. In an article by Price and colleagues [V. J. Price et al., mSphere 1(3):e00064-16, 2016, http://dx.doi.org/10.1128/mSphere.00064-16], the authors demonstrated how E. faecalis uses a restriction-modification system along with a clustered regularly interspaced short palindromic repeat (CRISPR)-Cas to function as a bacterial innate and adaptive immune system to regulate the influx of MGEs. The absence of these systems in high-risk hospital-adapted lineages of E. faecalis, including the prototypical V583 strain, appears to allow the ready acquisition of new traits that aid in the adaptation to new environmental stresses, including the evolution of resistance to many of our best antibiotics.

8.
Sci Transl Med ; 7(286): 286ra68, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25947163

ABSTRACT

Even under the most expert care, a properly constructed intestinal anastomosis can fail to heal, resulting in leakage of its contents, peritonitis, and sepsis. The cause of anastomotic leak remains unknown, and its incidence has not changed in decades. We demonstrate that the commensal bacterium Enterococcus faecalis contributes to the pathogenesis of anastomotic leak through its capacity to degrade collagen and to activate tissue matrix metalloproteinase 9 (MMP9) in host intestinal tissues. We demonstrate in rats that leaking anastomotic tissues were colonized by E. faecalis strains that showed an increased collagen-degrading activity and also an increased ability to activate host MMP9, both of which contributed to anastomotic leakage. We demonstrate that the E. faecalis genes gelE and sprE were required for E. faecalis-mediated MMP9 activation. Either elimination of E. faecalis strains through direct topical antibiotics applied to rat intestinal tissues or pharmacological suppression of intestinal MMP9 activation prevented anastomotic leak in rats. In contrast, the standard recommended intravenous antibiotics used in patients undergoing colorectal surgery did not eliminate E. faecalis at anastomotic tissues nor did they prevent leak in our rat model. Finally, we show in humans undergoing colon surgery and treated with the standard recommended intravenous antibiotics that their anastomotic tissues still contained E. faecalis and other bacterial strains with collagen-degrading/MMP9-activating activity. We suggest that intestinal microbes with the capacity to produce collagenases and to activate host metalloproteinase MMP9 may break down collagen in the intestinal tissue contributing to anastomotic leak.


Subject(s)
Anastomotic Leak/pathology , Collagen/chemistry , Enterococcus faecalis/pathogenicity , Intestinal Mucosa/metabolism , Intestines/microbiology , Matrix Metalloproteinase 9/metabolism , Anastomotic Leak/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Caenorhabditis elegans , Cell Line , Humans , Intestines/pathology , Ischemia/pathology , Macrophages/metabolism , Male , Mice , RNA, Ribosomal, 16S/genetics , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Treatment Outcome
9.
Appl Environ Microbiol ; 81(6): 1949-58, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25576607

ABSTRACT

The modification of microbial membranes to achieve biotechnological strain improvement with exogenous small molecules, such as oligopolyphenylenevinylene-conjugated oligoelectrolyte (OPV-COE) membrane insertion molecules (MIMs), is an emerging biotechnological field. Little is known about the interactions of OPV-COEs with their target, the bacterial envelope. We studied the toxicity of three previously reported OPV-COEs with a selection of Gram-negative and Gram-positive organisms and demonstrated that Gram-positive bacteria are more sensitive to OPV-COEs than Gram-negative bacteria. Transmission electron microscopy demonstrated that these MIMs disrupt microbial membranes and that this occurred to a much greater degree in Gram-positive organisms. We used a number of mutants to probe the nature of MIM interactions with the microbial envelope but were unable to align the membrane perturbation effects of these compounds to previously reported membrane disruption mechanisms of, for example, cationic antimicrobial peptides. Instead, the data support the notion that OPV-COEs disrupt microbial membranes through a suspected interaction with diphosphatidylglycerol (DPG), a major component of Gram-positive membranes. The integrity of model membranes containing elevated amounts of DPG was disrupted to a greater extent by MIMs than those prepared from Escherichia coli total lipid extracts alone.


Subject(s)
Cell Membrane/drug effects , Gram-Positive Bacteria/drug effects , Polyvinyls/metabolism , Polyvinyls/toxicity , Gram-Negative Bacteria/cytology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/cytology , Microscopy, Electron, Transmission
10.
mBio ; 5(5): e01726-14, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25249282

ABSTRACT

UNLABELLED: Enterococci are leading causes of hospital-acquired infection in the United States and continue to develop resistances to new antibiotics. Many Enterococcus faecalis isolates harbor pheromone-responsive plasmids that mediate horizontal transfer of even large blocks of chromosomal genes, resulting in hospital-adapted strains over a quarter of whose genomes consist of mobile elements. Pheromones to which the donor cells respond derive from lipoprotein signal peptides. Using a novel bacterial killing assay dependent on the presence of sex pheromones, we screened a transposon mutant library for functions that relate to the production and/or activity of the effector pheromone. Here we describe a previously uncharacterized, but well-conserved, ABC transporter that contributes to pheromone production. Using three distinct pheromone-dependent mating systems, we show that mutants defective in expressing this transporter display a 5- to 6-order-of-magnitude reduction in conjugation efficiency. In addition, we demonstrate that the ABC transporter mutant displays an altered biofilm architecture, with a significant reduction in biofilm biomass compared to that of its isogenic parent, suggesting that pheromone activity also influences biofilm development. The conservation of this peptide transporter across the Firmicutes suggests that it may also play an important role in cell-cell communication in other species within this important phylum. IMPORTANCE: Enterococcus faecalis ranks as one of the leading causes of hospital-associated infections. Strains possessing resistance to multiple antibiotics are becoming all too common in clinical settings. Pheromone-responsive plasmids play an important role in harboring and disseminating these antibiotic resistance genes. Here we have identified a novel ABC transporter that is responsible for the secretion of peptide pheromones, which enables communication between cells to mediate plasmid transfer. We have also shown that this transporter is important for biofilm formation, providing a strong rationale for its use as a viable therapeutic target which could be targeted to curb infection, as well as the spread of existing drug resistance.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Enterococcus faecalis/genetics , Peptides/metabolism , Pheromones/metabolism , ATP-Binding Cassette Transporters/genetics , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , Biofilms/growth & development , Chromosome Mapping , DNA Transposable Elements/genetics , Drug Resistance, Bacterial , Escherichia coli/genetics , Gene Deletion , Plasmids
11.
PLoS One ; 8(5): e64740, 2013.
Article in English | MEDLINE | ID: mdl-23734216

ABSTRACT

Enterococcus faecalis V583 is a vancomycin-resistant clinical isolate which belongs to the hospital-adapted clade, CC2. This strain harbours several factors that have been associated with virulence, including the fsr quorum-sensing regulatory system that is known to control the expression of GelE and SprE proteases. To discriminate between genes directly regulated by Fsr, and those indirectly regulated as the result of protease expression or activity, we compared gene expression in isogenic mutants of V583 variously defective in either Fsr quorum sensing or protease expression. Quorum sensing was artificially induced by addition of the quorum signal, GBAP, exogenously in a controlled manner. The Fsr regulon was found to be restricted to five genes, gelE, sprE, ef1097, ef1351 and ef1352. Twelve additional genes were found to be dependent on the presence of GBAP-induced proteases. Induction of GelE and SprE by GBAP via Fsr resulted in accumulation of mRNA encoding lrgAB, and this induction was found to be lytRS dependent. Drosophila infection was used to discern varying levels of toxicity stemming from mutations in the fsr quorum regulatory system and the genes that it regulates, highlighting the contribution of LrgAB and bacteriocin EF1097 to infection toxicity. A contribution of SprE to infection toxicity was also detected. This work brought to light new players in E. faecalis success as a pathogen and paves the way for future studies on host tolerance mechanisms to infections caused by this important nosocomial pathogen.


Subject(s)
Drosophila melanogaster/microbiology , Enterococcus faecalis/physiology , Quorum Sensing/physiology , Virulence Factors/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Enterococcus faecalis/genetics , Enterococcus faecalis/pathogenicity , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Male , Mutation , Oligonucleotide Array Sequence Analysis , Quorum Sensing/genetics , Reverse Transcriptase Polymerase Chain Reaction , Virulence/genetics , Virulence Factors/genetics
12.
J Bacteriol ; 195(14): 3125-34, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23645601

ABSTRACT

Enterococcus faecalis is a commensal bacterium found in the gastrointestinal tract of most mammals, including humans, and is one of the leading causes of nosocomial infections. One of the hallmarks of E. faecalis pathogenesis is its unusual ability to tolerate high concentrations of lysozyme, which is an important innate immune component of the host. Previous studies have shown that the presence of lysozyme leads to the activation of SigV, an extracytoplasmic function (ECF) sigma factor in E. faecalis, and that the deletion of sigV increases the susceptibility of the bacterium toward lysozyme. Here, we describe the contribution of Eep, a membrane-bound zinc metalloprotease, to the activation of SigV under lysozyme stress by its effects on the stability of the anti-sigma factor RsiV. We demonstrate that the Δeep mutant phenocopies the ΔsigV mutant in lysozyme, heat, ethanol, and acid stress susceptibility. We also show, using an immunoblot analysis, that in an eep deletion mutant, the anti-sigma factor RsiV is only partially degraded after lysozyme exposure, suggesting that RsiV is processed by unknown protease(s) prior to the action of Eep. An additional observation is that the deletion of rsiV, which results in constitutive SigV expression, leads to chaining of cells, suggesting that SigV might be involved in regulating cell wall-modifying enzymes important in cell wall turnover. We also demonstrate that, in the absence of eep or sigV, enterococci bind significantly more lysozyme, providing a plausible explanation for the increased sensitivity of these mutants toward lysozyme.


Subject(s)
Bacterial Proteins/metabolism , Enterococcus faecalis/metabolism , Immune Evasion , Membrane Proteins/metabolism , Muramidase/antagonists & inhibitors , Sigma Factor/metabolism , Bacterial Proteins/genetics , Enterococcus faecalis/genetics , Gene Deletion , Membrane Proteins/genetics , Stress, Physiological
13.
J Bacteriol ; 194(2): 368-75, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22081387

ABSTRACT

Transcription initiation is a critical step in bacterial gene regulation and is often controlled by transcription regulators. The alternate sigma factor (σ(54)) is one such regulator that facilitates activator-dependent transcription initiation and thus modulates the expression of a variety of genes involved in metabolism and pathogenesis in bacteria. This study describes the role of σ(54) in the nosocomial pathogen Enterococcus faecalis. Biofilm formation is one of the important pathogenic mechanisms of E. faecalis, as it elevates the organism's potential to cause surgical site and urinary tract infections. Lysis of bacterial cells within the population contributes to biofilm formation by providing extracellular DNA (eDNA) as a key component of the biofilm matrix. Deletion of rpoN rendered E. faecalis resistant to autolysis, which in turn impaired eDNA release. Despite the significant reduction in eDNA levels compared to the parental strain, the rpoN mutant formed more robust biofilms as observed using laser scanning confocal microscopy and Comstat analysis, indicating and emphasizing the presence of other matrix components. Initial adherence to a polystyrene surface was also enhanced in the mutant. Proteinase K treatment at early stages of biofilm development significantly reduced the accumulation of biofilm by the rpoN mutant. In conclusion, our data indicate that other factors in addition to eDNA might contribute to the overall composition of the enterococcal biofilm and that the regulatory role of σ(54) governs the nature and composition of the biofilm matrix.


Subject(s)
Bacteriolysis/physiology , Biofilms/growth & development , Enterococcus faecalis/enzymology , Enterococcus faecalis/physiology , RNA Polymerase Sigma 54/metabolism , Bacterial Adhesion/genetics , Bacterial Adhesion/physiology , DNA, Bacterial , Enterococcus faecalis/genetics , Gene Deletion , Gene Expression Regulation, Bacterial/physiology , Genetic Complementation Test , RNA Polymerase Sigma 54/genetics , Time Factors
14.
Microbiology (Reading) ; 158(Pt 2): 519-528, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22117005

ABSTRACT

The concomitant presence of a complete fsr quorum-sensing system and gelE-sprE operons in Enterococcus faecalis is known to be essential for the detection of gelatinase activity. However, there are reports of the absence of gelatinase activity despite the presence of complete fsr and gelE loci. In order to understand this incongruence between genotype and phenotype we sequenced fsr and gelE loci of the E. faecalis LN68 strain, which was previously found to carry both operons but to lack gelatinase activity. Of the 59 nucleotide differences detected compared with the gelatinase-positive V583 strain, we found a nonsense mutation (a premature STOP codon) predicted to truncate the ATPase sensor domain of the FsrC protein, responsible for sensing and transducing the signal from the quorum-sensing molecule. Strain LN68 was highly affected in the expression of the gelE and sprE genes, further supporting the lack of Fsr-dependent gelE induction. When we constructed a V583 mutant with the same premature stop mutation in the fsrC gene the resulting strain was no longer able to degrade gelatin. We conclude that the reduced ability to transduce the quorum-sensing signal of the prematurely truncated FsrC protein is sufficient to explain the negative gelatinase phenotype. As the incongruent genotype and phenotype is detected in natural isolates, we believe that the silencing of the quorum-sensing system Fsr may be beneficial for some E. faecalis strains.


Subject(s)
Bacterial Proteins/metabolism , Enterococcus faecalis/enzymology , Gelatinases/metabolism , Lactones/metabolism , Peptides, Cyclic/metabolism , Quorum Sensing , Signal Transduction , Amino Acid Sequence , Bacterial Proteins/genetics , Base Sequence , Enterococcus faecalis/genetics , Enterococcus faecalis/physiology , Gelatinases/genetics , Gene Expression Regulation, Bacterial , Genotype , Molecular Sequence Data , Phenotype
15.
J Bacteriol ; 193(7): 1643-52, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21257770

ABSTRACT

Selenium has been shown to be present as a labile cofactor in a small class of molybdenum hydroxylase enzymes in several species of clostridia that specialize in the fermentation of purines and pyrimidines. This labile cofactor is poorly understood, yet recent bioinformatic studies have suggested that Enterococcus faecalis could serve as a model system to better understand the way in which this enzyme cofactor is built and the role of these metalloenzymes in the physiology of the organism. An mRNA that encodes a predicted selenium-dependent molybdenum hydroxylase (SDMH) has also been shown to be specifically increased during the transition from planktonic growth to biofilm growth. Based on these studies, we examined whether this organism produces an SDMH and probed whether selenoproteins may play a role in biofilm physiology. We observed a substantial increase in biofilm density upon the addition of uric acid to cells grown in a defined culture medium, but only when molybdate (Mo) and selenite (Se) were also added. We also observed a significant increase in biofilm density in cells cultured in tryptic soy broth with 1% glucose (TSBG) when selenite was added. In-frame deletion of selD, which encodes selenophosphate synthetase, also blocked biofilm formation that occurred upon addition of selenium. Moreover, mutation in the gene encoding the molybdoenzyme (xdh) prevented the induction of biofilm proliferation upon supplementation with selenium. Tungstate or auranofin addition also blocked this enhanced biofilm density, likely through inhibition of molybdenum or selenium cofactor synthesis. A large protein complex labeled with (75)Se is present in higher concentrations in biofilms than in planktonic cells, and the same complex is formed in TSBG. Xanthine dehydrogenase activity correlates with the presence of this labile selenoprotein complex and is absent in a selD or an xdh mutant. Enhanced biofilm density correlates strongly with higher levels of extracellular peroxide, which is produced upon the addition of selenite to TSBG. Peroxide levels are not increased in either the selD or the xdh mutant upon addition of selenite. Extracellular superoxide production, a phenomenon well established to be linked to clinical isolates, is abolished in both mutant strains. Taken together, these data provide evidence that an SDMH is involved in biofilm formation in Enterococcus faecalis, contributing to oxidant production either directly or alternatively through its involvement in redox-dependent processes linked to oxidant production.


Subject(s)
Biofilms/growth & development , Enterococcus faecalis/enzymology , Enterococcus faecalis/physiology , Oxidants/biosynthesis , Selenium/metabolism , Xanthine Dehydrogenase/metabolism , Auranofin/pharmacology , Culture Media , Enterococcus faecalis/drug effects , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Glucose , Molybdenum/metabolism , Tungsten Compounds/pharmacology , Up-Regulation , Uric Acid/pharmacology , Xanthine Dehydrogenase/genetics
16.
Infect Immun ; 78(10): 4166-75, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20696830

ABSTRACT

Catheter-associated urinary tract infections (CAUTIs) constitute the majority of nosocomial UTIs and pose significant clinical challenges. Enterococcal species are among the predominant causative agents of CAUTIs. However, very little is known about the pathophysiology of Enterococcus-mediated UTIs. We optimized a murine model of foreign body-associated UTI in order to mimic conditions of indwelling catheters in patients. In this model, the presence of a foreign body elicits major histological changes and induces the expression of several proinflammatory cytokines in the bladder. In addition, in contrast to naïve mice, infection of catheter-implanted mice with Enterococcus faecalis induced the specific expression of interleukin 1ß (IL-1ß) and macrophage inflammatory protein 1α (MIP-1α) in the bladder. These responses resulted in a favorable niche for the development of persistent E. faecalis infections in the murine bladders and kidneys. Furthermore, biofilm formation on the catheter implant in vivo correlated with persistent infections. However, the enterococcal autolytic factors GelE and Atn (also known as AtlA), which are important in biofilm formation in vitro, are dispensable in vivo. In contrast, the housekeeping sortase A (SrtA) is critical for biofilm formation and virulence in CAUTIs. Overall, this murine model represents a significant advance in the understanding of CAUTIs and underscores the importance of urinary catheterization during E. faecalis uropathogenesis. This model is also a valuable tool for the identification of virulence determinants that can serve as potential antimicrobial targets for the treatment of enterococcal infections.


Subject(s)
Biofilms/growth & development , Catheter-Related Infections/microbiology , Enterococcus faecalis/pathogenicity , Foreign Bodies/complications , Gram-Positive Bacterial Infections/microbiology , Urinary Tract Infections/etiology , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biomarkers , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Enterococcus faecalis/physiology , Female , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic , Gram-Positive Bacterial Infections/etiology , Inflammation/metabolism , Kidney/microbiology , Mice , Mice, Inbred C57BL , Silicones , Urinary Bladder/immunology , Urinary Bladder/microbiology , Urinary Bladder/pathology , Urinary Tract Infections/microbiology , Virulence
17.
Infect Immun ; 78(11): 4936-43, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713628

ABSTRACT

The Gram-positive pathogen Enterococcus faecalis is a leading agent of nosocomial infections, including urinary tract infections, surgical site infections, and bacteremia. Among the infections caused by E. faecalis, endocarditis remains a serious clinical manifestation and unique in that it is commonly acquired in a community setting. Infective endocarditis is a complex disease, with many host and microbial components contributing to the formation of bacterial biofilm-like vegetations on the aortic valve and adjacent areas within the heart. In the current study, we compared the pathogenic potential of the vancomycin-resistant E. faecalis V583 and three isogenic protease mutants (ΔgelE, ΔsprE, and ΔgelE ΔsprE mutants) in a rabbit model of enterococcal endocarditis. The bacterial burdens displayed by GelE(-) mutants (ΔgelE and ΔgelE ΔsprE mutants) in the heart were significantly lower than those of V583 or the SprE(-) mutant. Vegetations on the aortic valve infected with GelE(-) mutants (ΔgelE and ΔgelE ΔsprE mutants) also showed a significant increase in deposition of fibrinous matrix layer and increased chemotaxis of inflammatory cells. In support of a role for proteolytic modulation of the immune response to E. faecalis, we also demonstrate that GelE can cleave the anaphylatoxin complement C5a and that this proteolysis leads to decreased neutrophil migration in vitro. In vivo, a decreased heterophil (neutrophil-like cell) migration was observed at tissue sites infected with GelE-producing strains but not at those infected with SprE-producing strains. Taken together, these observations suggest that of the two enterococcal proteases, gelatinase is the principal mediator of pathogenesis in endocarditis.


Subject(s)
Endocarditis, Bacterial/pathology , Enterococcus faecalis/pathogenicity , Gelatinases/metabolism , Gram-Positive Bacterial Infections/pathology , Animals , Aortic Valve/microbiology , Aortic Valve/pathology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Complement C5a/metabolism , Endocarditis, Bacterial/microbiology , Enterococcus faecalis/enzymology , Enterococcus faecalis/genetics , Gelatinases/genetics , Gram-Positive Bacterial Infections/microbiology , HL-60 Cells , Humans , Mutation , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Rabbits
18.
Proc Natl Acad Sci U S A ; 107(27): 12269-74, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20566881

ABSTRACT

The Enterococcus faecalis pathogenicity island (PAI) encodes known virulence traits and >100 additional genes with unknown roles in enterococcal biology. Phage-related integration and excision genes, and direct repeats flanking the island, suggest it moves as an integrative conjugative element (ICE). However, transfer was observed not to require these genes. Transfer only occurred from donors possessing a pheromone responsive-type of conjugative plasmid, and was invariably accompanied by transfer of flanking donor chromosome sequences. Deletion of plasmid transfer functions, including the cis-acting origin of transfer (oriT), abolished movement. In addition to demonstrating PAI movement by a mechanism involving plasmid integration, we observed transfer of a selectable marker placed virtually anywhere on the chromosome. Transfer of this selectable marker was observed to be accompanied by chromosome-chromosome transfer of vancomycin resistance, MLST markers, and capsule genes as well. Plasmid mobilization therefore appears to be a major mechanism for horizontal gene transfer in the evolution of antibiotic resistant E. faecalis strains capable of causing human infection.


Subject(s)
Chromosomes, Bacterial/genetics , Conjugation, Genetic , Enterococcus faecalis/genetics , Genomic Islands/genetics , Bacterial Capsules/metabolism , Bacterial Infections/microbiology , Bacteriophages/genetics , DNA, Bacterial/genetics , Drug Resistance, Bacterial/genetics , Enterococcus faecalis/metabolism , Enterococcus faecalis/pathogenicity , Gene Transfer, Horizontal , Humans , Mutation , Plasmids/genetics , Transduction, Genetic , Vancomycin Resistance/genetics , Virulence/genetics
19.
Int J Artif Organs ; 32(9): 537-44, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19851979

ABSTRACT

Bacterial autolysis has recently been identified as a key mechanism that regulates different phases of biofilm development including microcolony formation and dispersal. However such autolytic measures are limited to a subfraction of cells within the entire biofilm population. Here we speculate on the role biofilm heterogeneity plays in limiting autolysis within biofilms and further describe the molecular regulation of suicidal and fratricidal mechanisms in biofilm development of Staphylococcus aureus and Enterococcus faecalis.


Subject(s)
Bacteriolysis , Biofilms , Enterococcus faecalis/growth & development , Staphylococcus aureus/growth & development , Bacterial Proteins/metabolism , DNA, Bacterial/metabolism , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Humans , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
20.
Infect Immun ; 77(12): 5551-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19805541

ABSTRACT

It has become increasingly difficult to treat infections caused by Enterococcus faecalis due to its high levels of intrinsic and acquired antibiotic resistance. However, few studies have explored the mechanisms that E. faecalis employs to circumvent the host innate immune response and establish infection. Capsular polysaccharides are important virulence factors that are associated with innate immune evasion. We demonstrate, using cultured macrophages (RAW 264.7), that capsule-producing E. faecalis strains of either serotype C or D are more resistant to complement-mediated opsonophagocytosis than unencapsulated strains. We show that differences in opsonophagocytosis are not due to variations in C3 deposition but are due to the ability of capsule to mask bound C3 from detection on the surface of E. faecalis. Similarly, E. faecalis capsule masks lipoteichoic acid from detection, which correlates with decreased tumor necrosis factor alpha production by cultured macrophages in the presence of encapsulated strains compared to that in the presence of unencapsulated strains. Our studies confirm the important role of the capsule as a virulence factor of E. faecalis and provide several mechanisms by which the presence of the capsule influences evasion of the innate immune response and suggest that the capsule could be a potential target for developing alternative therapies to treat E. faecalis infections.


Subject(s)
Bacterial Capsules/immunology , Bacterial Capsules/physiology , Enterococcus faecalis/immunology , Enterococcus faecalis/pathogenicity , Virulence Factors/immunology , Virulence Factors/physiology , Animals , Cell Line , Complement C3/immunology , Complement C3/metabolism , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Opsonin Proteins/immunology , Phagocytosis/immunology , Protein Binding , Teichoic Acids/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...